Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Nano Lett ; 24(8): 2671-2679, 2024 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-38375804

RESUMO

The emerging two-photon polymerization (TPP) technique enables high-resolution printing of complex 3D structures, revolutionizing micro/nano additive manufacturing. Various fast scanning and parallel processing strategies have been proposed to promote its efficiency. However, obtaining large numbers of uniform focal spots for parallel high-speed scanning remains challenging, which hampers the realization of higher throughput. We report a TPP printing platform that combines galvanometric mirrors and liquid crystal on silicon spatial light modulator (LCoS-SLM). By setting the target light field at LCoS-SLM's diffraction center, sufficient energy is acquired to support simultaneous polymerization of over 400 foci. With fast scanning, the maximum printing speed achieves 1.49 × 108 voxels s-1, surpassing the existing scanning-based TPP methods while maintaining high printing resolution and flexibility. To demonstrate the processing capability, functional 3D microstructure arrays are rapidly fabricated and applied in micro-optics and micro-object manipulation. Our method may expand the prospects of TPP in large-scale micro/nanomanufacturing.

2.
Stem Cells ; 37(3): 306-317, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30471152

RESUMO

Directed differentiation of human induced pluripotent stem cells (iPSCs) toward hepatobiliary lineages has been increasingly used as models of human liver development/diseases. As protein kinases are important components of signaling pathways regulating cell fate changes, we sought to define the key molecular mediators regulating human liver development using inhibitors targeting tyrosine kinases during hepatic differentiation of human iPSCs. A library of tyrosine kinase inhibitors was used for initial screening during the multistage differentiation of human iPSCs to hepatic lineage. Among the 80 kinase inhibitors tested, only Src inhibitors suppressed endoderm formation while none had significant effect on later stages of hepatic differentiation. Transient inhibition of c-Src during endodermal induction of human iPSCs reduced endodermal commitment and expression of endodermal markers, including SOX17 and FOXA2, in a dose-dependent manner. Interestingly, the transiently treated cells later developed into profibrogenic cholangiocyte-like cells expressing both cholangiocyte markers, such as CK7 and CK19, and fibrosis markers, including Collagen1 and smooth muscle actin. Further analysis of these cells revealed colocalized expression of collagen and yes-associated protein (YAP; a marker associated with bile duct proliferation/fibrosis) and an increased production of interleukin-6 and tumor necrosis factor-α. Moreover, treatment with verteporfin, a YAP inhibitor, significantly reduced expression of fibrosis markers. In summary, these results suggest that c-Src has a critical role in cell fate determination during endodermal commitment of human iPSCs, and its alteration in early liver development in human may lead to increased production of abnormal YAP expressing profibrogenic proinflammatory cholangiocytes, similar to those seen in livers of patients with biliary fibrosis. Stem Cells 2019;37:306-317.


Assuntos
Proteína Tirosina Quinase CSK/antagonistas & inibidores , Diferenciação Celular/efeitos dos fármacos , Linhagem da Célula/efeitos dos fármacos , Endoderma/enzimologia , Inibidores de Proteínas Quinases/farmacologia , Ductos Biliares/enzimologia , Ductos Biliares/patologia , Proteína Tirosina Quinase CSK/metabolismo , Endoderma/patologia , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Células-Tronco Pluripotentes Induzidas/enzimologia , Células-Tronco Pluripotentes Induzidas/patologia , Fígado/enzimologia , Fígado/patologia
3.
Biol Pharm Bull ; 43(5): 810-816, 2020 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-32101839

RESUMO

Osteoarthritis is a common disease character with progressive destruction of cartilage. MicroRNA (miR)-140-3p was validated as a biomarker for osteoarthritis. However, the mechanism by which miRNA-140-3p regulates osteoarthritis remains unclear. Thus, this study aims to evaluate the potential function of miRNA-140-3p during the pathogenesis of osteoarthritis. MiRNA-140-3p expression in tissue and CHON-001 chondrocyte cells was determined with quantitative real time (qRT)-PCR. In vitro osteoarthritis model was established by treatment of the chondrocyte cells CHON-001 with interleukin (IL)-1ß. Cell proliferation and apoptosis were measured with cell counting kit-8 (CCK8) and Annexin V/propidium iodide (PI) apoptosis assay, respectively. Protein expressions were evaluated using Western blot. The target gene of miR-140-3p was predicted using Targetscan and miRDB. MiR-140-3p was downregulated in knee tissue from patients with osteoarthritis. IL-1ß inhibited the proliferation of CHON-001 cells via inducing apoptosis. In addition, IL-1ß significantly inhibited the expressions of collagen II and aggrecan and increased the level of matrix metalloproteinase (MMP)13. However, the effects of IL-1ß could be ameliorated by the addition of miR-140-3p mimics. Moreover, luciferase reporter assay demonstrated CXCR4 as a target gene of miR-140-3p. IL-1ß-induced upregulation of CXCR4 could be blocked by miR-140-3p mimics. Our study indicated that miR-140-3p could suppress the progression of osteoarthritis by directly targeting CXCR4. Therefore, miR-140-3p might serve as a potential therapeutic target for the treatment of osteoarthritis.


Assuntos
MicroRNAs , Osteoartrite do Joelho/genética , Receptores CXCR4/genética , Apoptose , Cartilagem Articular/metabolismo , Linhagem Celular , Proliferação de Células , Condrócitos/metabolismo , Progressão da Doença , Humanos , Interleucina-1beta/farmacologia , Osteoartrite do Joelho/metabolismo , Espécies Reativas de Oxigênio/metabolismo
4.
J Pediatr Gastroenterol Nutr ; 68(1): 56-63, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30358741

RESUMO

Biliary atresia (BA) is the most common cause of pediatric end-stage liver disease and the etiology is poorly understood. There is no effective therapy for BA partly due to lack of human BA models. Towards developing in vitro human models of BA, disease-specific induced pluripotent stem cells (iPSCs) from 6 BA patients were generated using non-integrating episomal plasmids. In addition, to determine the functional significance of BA-susceptibility genes identified by genome-wide association studies (GWAS) in biliary development, a genome-editing approach was used to create iPSCs with defined mutations in these GWAS BA loci. Using the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/Cas9 system, isogenic iPSCs deficient in BA-associated genes (GPC1 and ADD3) were created from healthy iPSCs. Both the BA patient-iPSCs and the knock out (KO) iPSCs were studied for their in vitro biliary differentiation potential. These BA-specific iPSCs demonstrated significantly decreased formation of ductal structures, decreased expression of biliary markers including CK7, EpCAM, SOX9, CK19, AE2, and CFTR and increased fibrosis markers such as alpha smooth muscle actin, Loxl2, and Collagen1 compared to controls. Both the patient- and the KO-iPSCs also showed increased yes-associated protein (YAP, a marker of bile duct proliferation/fibrosis). Collagen and YAP were reduced by treatment with the anti-fibrogenic drug pentoxifylline. In summary, these BA-specific human iPSCs showed deficiency in biliary differentiation along with increased fibrosis, the 2 key disease features of BA. These iPSCs can provide new human BA models for understanding the molecular basis of abnormal biliary development and opportunities to identify drugs that have therapeutic effects on BA.


Assuntos
Atresia Biliar/genética , Células-Tronco Pluripotentes Induzidas/citologia , Diferenciação Celular/genética , Feminino , Loci Gênicos/genética , Predisposição Genética para Doença/genética , Estudo de Associação Genômica Ampla , Humanos , Lactente , Masculino , Mutação , Pentoxifilina/farmacologia
5.
Blood ; 125(23): 3637-46, 2015 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-25862562

RESUMO

Atypical hemolytic uremic syndrome (aHUS) is a thrombotic microangiopathy (TMA) characterized by excessive activation of the alternative pathway of complement (APC). Atypical HUS is frequently a diagnosis of exclusion. Differentiating aHUS from other TMAs, especially thrombotic thrombocytopenic purpura (TTP), is difficult due to overlapping clinical manifestations. We sought to develop a novel assay to distinguish aHUS from other TMAs based on the hypothesis that paroxysmal nocturnal hemoglobinuria cells are more sensitive to APC-activated serum due to deficiency of glycosylphosphatidylinositol- anchored complement regulatory proteins (GPI-AP). Here, we demonstrate that phosphatidylinositol-specific phospholipase C-treated EA.hy926 cells and PIGA-mutant TF-1 cells are more susceptible to serum from aHUS patients than parental EA.hy926 and TF-1 cells. We next studied 31 samples from 25 patients with TMAs, including 9 with aHUS and 12 with TTP. Increased C5b-9 deposition was evident by confocal microscopy and flow cytometry on GPI-AP-deficient cells incubated with aHUS serum compared with heat-inactivated control, TTP, and normal serum. Differences in cell viability were observed in biochemically GPI-AP-deficient cells and were further increased in PIGA-deficient cells. Serum from patients with aHUS resulted in a significant increase of nonviable PIGA-deficient TF-1 cells compared with serum from healthy controls (P < .001) and other TMAs (P < .001). The cell viability assay showed high reproducibility, sensitivity, and specificity in detecting aHUS. In conclusion, we developed a simple, rapid, and serum-based assay that helps to differentiate aHUS from other TMAs.


Assuntos
Síndrome Hemolítico-Urêmica Atípica/sangue , Síndrome Hemolítico-Urêmica Atípica/diagnóstico , Adulto , Idoso , Síndrome Hemolítico-Urêmica Atípica/genética , Sobrevivência Celular , Feminino , Humanos , Masculino , Proteínas de Membrana/sangue , Proteínas de Membrana/genética , Pessoa de Meia-Idade , Púrpura Trombocitopênica Trombótica/sangue , Púrpura Trombocitopênica Trombótica/diagnóstico , Púrpura Trombocitopênica Trombótica/genética , Soro/metabolismo
6.
Hum Genet ; 135(9): 1041-58, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27256364

RESUMO

The discovery that mature cells can be reprogrammed to become pluripotent and the development of engineered endonucleases for enhancing genome editing are two of the most exciting and impactful technology advances in modern medicine and science. Human pluripotent stem cells have the potential to establish new model systems for studying human developmental biology and disease mechanisms. Gene correction in patient-specific iPSCs can also provide a novel source for autologous cell therapy. Although historically challenging, precise genome editing in human iPSCs is becoming more feasible with the development of new genome-editing tools, including ZFNs, TALENs, and CRISPR. iPSCs derived from patients of a variety of diseases have been edited to correct disease-associated mutations and to generate isogenic cell lines. After directed differentiation, many of the corrected iPSCs showed restored functionality and demonstrated their potential in cell replacement therapy. Genome-wide analyses of gene-corrected iPSCs have collectively demonstrated a high fidelity of the engineered endonucleases. Remaining challenges in clinical translation of these technologies include maintaining genome integrity of the iPSC clones and the differentiated cells. Given the rapid advances in genome-editing technologies, gene correction is no longer the bottleneck in developing iPSC-based gene and cell therapies; generating functional and transplantable cell types from iPSCs remains the biggest challenge needing to be addressed by the research field.


Assuntos
Edição de Genes , Terapia Genética , Células-Tronco Pluripotentes Induzidas/citologia , Modelos Biológicos , Anemia Falciforme/terapia , Fibrose Cística/terapia , Humanos , Distrofia Muscular de Duchenne/terapia , Medicina Regenerativa , Imunodeficiência Combinada Severa/terapia , Deficiência de alfa 1-Antitripsina/terapia , Talassemia beta/terapia
7.
Stem Cells ; 33(5): 1470-9, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25702619

RESUMO

Human induced pluripotent stem cells (iPSCs) and genome editing provide a precise way to generate gene-corrected cells for disease modeling and cell therapies. Human iPSCs generated from sickle cell disease (SCD) patients have a homozygous missense point mutation in the HBB gene encoding adult ß-globin proteins, and are used as a model system to improve strategies of human gene therapy. We demonstrate that the CRISPR/Cas9 system designer nuclease is much more efficient in stimulating gene targeting of the endogenous HBB locus near the SCD point mutation in human iPSCs than zinc finger nucleases and TALENs. Using a specific guide RNA and Cas9, we readily corrected one allele of the SCD HBB gene in human iPSCs by homologous recombination with a donor DNA template containing the wild-type HBB DNA and a selection cassette that was subsequently removed to avoid possible interference of HBB transcription and translation. We chose targeted iPSC clones that have one corrected and one disrupted SCD allele for erythroid differentiation assays, using an improved xeno-free and feeder-free culture condition we recently established. Erythrocytes from either the corrected or its parental (uncorrected) iPSC line were generated with similar efficiencies. Currently ∼6%-10% of these differentiated erythrocytes indeed lacked nuclei, characteristic of further matured erythrocytes called reticulocytes. We also detected the 16-kDa ß-globin protein expressed from the corrected HBB allele in the erythrocytes differentiated from genome-edited iPSCs. Our results represent a significant step toward the clinical applications of genome editing using patient-derived iPSCs to generate disease-free cells for cell and gene therapies. Stem Cells 2015;33:1470-1479.


Assuntos
Anemia Falciforme/genética , Diferenciação Celular , Eritrócitos/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia , Mutação Puntual/genética , Edição de RNA/genética , Globinas beta/genética , Adulto , Anemia Falciforme/patologia , Linhagem Celular , Células Eritroides/citologia , Células Alimentadoras/citologia , Marcação de Genes , Loci Gênicos , Genoma Humano , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo
8.
Mol Ther ; 23(3): 570-7, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25418680

RESUMO

Efficient and precise genome editing is crucial for realizing the full research and therapeutic potential of human induced pluripotent stem cells (iPSCs). Engineered nucleases including CRISPR/Cas9 and transcription activator like effector nucleases (TALENs) provide powerful tools for enhancing gene-targeting efficiency. In this study, we investigated the relative efficiencies of CRISPR/Cas9 and TALENs in human iPSC lines for inducing both homologous donor-based precise genome editing and nonhomologous end joining (NHEJ)-mediated gene disruption. Significantly higher frequencies of NHEJ-mediated insertions/deletions were detected at several endogenous loci using CRISPR/Cas9 than using TALENs, especially at nonexpressed targets in iPSCs. In contrast, comparable efficiencies of inducing homologous donor-based genome editing were observed at disease-associated loci in iPSCs. In addition, we investigated the specificity of guide RNAs used in the CRISPR/Cas9 system in targeting disease-associated point mutations in patient-specific iPSCs. Using myeloproliferative neoplasm patient-derived iPSCs that carry an acquired JAK2-V617F point mutation and α1-antitrypsin (AAT) deficiency patient-derived iPSCs that carry an inherited Z-AAT point mutation, we demonstrate that Cas9 can specifically target either the mutant or the wild-type allele with little disruption at the other allele differing by a single nucleotide. Overall, our results demonstrate the advantages of the CRISPR/Cas9 system in allele-specific genome targeting and in NHEJ-mediated gene disruption.


Assuntos
Alelos , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Genoma Humano , Células-Tronco Pluripotentes Induzidas/metabolismo , Locos de Características Quantitativas , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sequência de Bases , Proteína 9 Associada à CRISPR , Linhagem Celular , Reparo do DNA por Junção de Extremidades , Endonucleases/genética , Endonucleases/metabolismo , Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas/patologia , Janus Quinase 2/genética , Janus Quinase 2/metabolismo , Dados de Sequência Molecular , Mutação , Transtornos Mieloproliferativos/genética , Transtornos Mieloproliferativos/metabolismo , Transtornos Mieloproliferativos/patologia , RNA Guia de Cinetoplastídeos/genética , RNA Guia de Cinetoplastídeos/metabolismo , Reparo de DNA por Recombinação , alfa 1-Antitripsina/genética , alfa 1-Antitripsina/metabolismo
9.
Am J Hum Genet ; 90(2): 295-300, 2012 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-22305531

RESUMO

Phosphatidylinositol glycan class A (PIGA) is involved in the first step of glycosylphosphatidylinositol (GPI) biosynthesis. Many proteins, including CD55 and CD59, are anchored to the cell by GPI. Loss of CD55 and CD59 on erythrocytes causes complement-mediated lysis in paroxysmal nocturnal hemoglobinuria (PNH), a disease that manifests after clonal expansion of hematopoietic cells with somatic PIGA mutations. Although somatic PIGA mutations have been identified in many PNH patients, it has been proposed that germline mutations are lethal. We report a family with an X-linked lethal disorder involving cleft palate, neonatal seizures, contractures, central nervous system (CNS) structural malformations, and other anomalies. An X chromosome exome next-generation sequencing screen identified a single nonsense PIGA mutation, c.1234C>T, which predicts p.Arg412(∗). This variant segregated with disease and carrier status in the family, is similar to mutations known to cause PNH as a result of PIGA dysfunction, and was absent in 409 controls. PIGA-null mutations are thought to be embryonic lethal, suggesting that p.Arg412(∗) PIGA has residual function. Transfection of a mutant p.Arg412(∗) PIGA construct into PIGA-null cells showed partial restoration of GPI-anchored proteins. The genetic data show that the c.1234C>T (p.Arg412(∗)) mutation is present in an affected child, is linked to the affected chromosome in this family, is rare in the population, and results in reduced, but not absent, biosynthesis of GPI anchors. We conclude that c.1234C>T in PIGA results in the lethal X-linked phenotype recognized in the reported family.


Assuntos
Genes Ligados ao Cromossomo X , Mutação em Linhagem Germinativa , Hemoglobinúria Paroxística/genética , Proteínas de Membrana/genética , Adulto , Animais , Cromossomos Humanos X/genética , Exoma/genética , Saúde da Família , Feminino , Genótipo , Heterozigoto , Humanos , Masculino , Camundongos , Linhagem , Fenótipo , Gravidez , Transfecção/métodos
10.
Blood ; 121(15): 2882-90, 2013 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-23372166

RESUMO

Advancements in human pluripotent stem cell (hPSC) research have potential to revolutionize therapeutic transplantation. It has been demonstrated that transcription factors may play key roles in regulating maintenance, expansion, and differentiation of hPSCs. In addition to its regulatory functions in hematopoiesis and blood-related disorders, the transcription factor RUNX1 is also required for the formation of definitive blood stem cells. In this study, we demonstrated that expression of endogenous RUNX1a, an isoform of RUNX1, parallels with lineage commitment and hematopoietic emergence from hPSCs, including both human embryonic stem cells and inducible pluripotent stem cells. In a defined hematopoietic differentiation system, ectopic expression of RUNX1a facilitates emergence of hematopoietic progenitor cells (HPCs) and positively regulates expression of mesoderm and hematopoietic differentiation-related factors, including Brachyury, KDR, SCL, GATA2, and PU.1. HPCs derived from RUNX1a hPSCs show enhanced expansion ability, and the ex vivo-expanded cells are capable of differentiating into multiple lineages. Expression of RUNX1a in embryoid bodies (EBs) promotes definitive hematopoiesis that generates erythrocytes with ß-globin production. Moreover, HPCs generated from RUNX1a EBs possess ≥9-week repopulation ability and show multilineage hematopoietic reconstitution in vivo. Together, our results suggest that RUNX1a facilitates the process of producing therapeutic HPCs from hPSCs.


Assuntos
Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Células-Tronco Embrionárias/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Western Blotting , Diferenciação Celular/genética , Linhagem Celular , Linhagem da Célula/genética , Proliferação de Células , Células Cultivadas , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Corpos Embrioides/citologia , Corpos Embrioides/metabolismo , Células-Tronco Embrionárias/citologia , Proteínas Fetais/genética , Proteínas Fetais/metabolismo , Fator de Transcrição GATA2/genética , Fator de Transcrição GATA2/metabolismo , Expressão Gênica , Células-Tronco Hematopoéticas/citologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Microscopia Confocal , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Transativadores/genética , Transativadores/metabolismo
11.
Stem Cells ; 32(1): 269-78, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24105986

RESUMO

Disease-specific induced pluripotent stem cells (iPSCs) provide an unprecedented opportunity to establish novel disease models and accelerate drug development using distinct tissue target cells generated from isogenic iPSC lines with and without disease-causing mutations. To realize the potential of iPSCs in modeling acquired diseases which are usually heterogeneous, we have generated multiple iPSC lines including two lines that are JAK2-wild-type and four lines homozygous for JAK2-V617F somatic mutation from a single polycythemia vera (PV) patient blood. In vitro differentiation of the same patient-derived iPSC lines have demonstrated the differential contributions of their parental hematopoietic clones to the abnormal erythropoiesis including the formation of endogenous erythroid colonies. This iPSC approach thus may provide unique and valuable insights into the genetic events responsible for disease development. To examine the potential of iPSCs in drug testing, we generated isogenic hematopoietic progenitors and erythroblasts from the same iPSC lines derived from PV patients and normal donors. Their response to three clinical JAK inhibitors, INCB018424 (Ruxolitinib), TG101348 (SAR302503), and the more recent CYT387 was evaluated. All three drugs similarly inhibited erythropoiesis from normal and PV iPSC lines containing the wild-type JAK2 genotype, as well as those containing a homozygous or heterozygous JAK2-V617F activating mutation that showed increased erythropoiesis without a JAK inhibitor. However, the JAK inhibitors had less inhibitory effect on the self-renewal of CD34+ hematopoietic progenitors. The iPSC-mediated disease modeling thus underlies the ineffectiveness of the current JAK inhibitors and provides a modeling system to develop better targeted therapies for the JAK2 mutated hematopoiesis.


Assuntos
Eritroblastos/efeitos dos fármacos , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Janus Quinase 2/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Diferenciação Celular/efeitos dos fármacos , Eritroblastos/enzimologia , Eritropoese/efeitos dos fármacos , Hematopoese/efeitos dos fármacos , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/enzimologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/enzimologia , Janus Quinase 2/genética
12.
Mol Ther ; 22(2): 451-463, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24002691

RESUMO

There is a constant shortage of red blood cells (RBCs) from sufficiently matched donors for patients who need chronic transfusion. Ex vivo expansion and maturation of human erythroid precursors (erythroblasts) from the patients or optimally matched donors could represent a potential solution. Proliferating erythroblasts can be expanded from umbilical cord blood mononuclear cells (CB MNCs) ex vivo for 10(6)-10(7)-fold (in ~50 days) before proliferation arrest and reaching sufficient number for broad application. Here, we report that ectopic expression of three genetic factors (Sox2, c-Myc, and an shRNA against TP53 gene) associated with iPSC derivation enables CB-derived erythroblasts to undergo extended expansion (~10(68)-fold in ~12 months) in a serum-free culture condition without change of cell identity or function. These expanding erythroblasts maintain immature erythroblast phenotypes and morphology, a normal diploid karyotype and dependence on a specific combination of growth factors for proliferation throughout expansion period. When being switched to a terminal differentiation condition, these immortalized erythroblasts gradually exit cell cycle, decrease cell size, accumulate hemoglobin, condense nuclei and eventually give rise to enucleated hemoglobin-containing erythrocytes that can bind and release oxygen. Our result may ultimately lead to an alternative approach to generate unlimited numbers of RBCs for personalized transfusion medicine.


Assuntos
Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/metabolismo , Eritropoese/fisiologia , Sangue Fetal/citologia , Diferenciação Celular/genética , Proliferação de Células , Análise por Conglomerados , Citocinas/metabolismo , Citocinas/farmacologia , Diploide , Eritroblastos/citologia , Eritroblastos/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Genes myb , Genes myc , Hemoglobinas/genética , Hemoglobinas/metabolismo , Hormônios/metabolismo , Hormônios/farmacologia , Humanos , Cariótipo , Fatores de Transcrição SOXB1/genética
13.
Angew Chem Int Ed Engl ; 54(6): 1859-63, 2015 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-25504624

RESUMO

Triptolide is a key component of the traditional Chinese medicinal plant Thunder God Vine and has potent anticancer and immunosuppressive activities. It is an irreversible inhibitor of eukaryotic transcription through covalent modification of XPB, a subunit of the general transcription factor TFIIH. Cys342 of XPB was identified as the residue that undergoes covalent modification by the 12,13-epoxide group of triptolide. Mutation of Cys342 of XPB to threonine conferred resistance to triptolide on the mutant protein. Replacement of the endogenous wild-type XPB with the Cys342Thr mutant in a HEK293T cell line rendered it completely resistant to triptolide, thus validating XPB as the physiologically relevant target of triptolide. Together, these results deepen our understanding of the interaction between triptolide and XPB and have implications for the future development of new analogues of triptolide as leads for anticancer and immunosuppressive drugs.


Assuntos
Cisteína/química , Compostos de Epóxi/metabolismo , Fator de Transcrição TFIIH/metabolismo , Fator de Transcrição TFIIH/química
14.
Hum Mutat ; 35(3): 350-5, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24357517

RESUMO

The phosphatidylinositol glycan class A (PIGA) protein is a member of the glycosylphosphatidylinositol anchor pathway. Germline mutations in PIGA located at Xp22.2 are thought to be lethal in males. However, a nonsense mutation in the last coding exon was recently described in two brothers with multiple congenital anomalies-hypotonia-seizures syndrome 2 (MCAHS2) who survived through birth likely because of the hypomorphic nature of the truncated protein, but died in their first weeks of life. Here, we report on a frameshift mutation early in the PIGA cDNA (c.76dupT; p.Y26Lfs*3) that cosegregates with the disease in a large family diagnosed with a severe syndromic form of X-linked intellectual disability. Unexpectedly, CD59 surface expression suggested the production of a shorter PIGA protein with residual functionality. We provide evidence that the second methionine at position 37 may be used for the translation of a 36 amino acids shorter PIGA. Complementation assays confirmed that this shorter PIGA cDNA was able to partially rescue the surface expression of CD59 in a PIGA-null cell line. Taken together, our data strongly suggest that the early frameshift mutation in PIGA produces a truncated hypomorph, which is sufficient to rescue the lethality in males but not the MCAHS2-like phenotype.


Assuntos
Mutação da Fase de Leitura , Genes Ligados ao Cromossomo X , Deficiência Intelectual/genética , Proteínas de Membrana/genética , Cromossomos Humanos X/genética , Exoma , Éxons , Feminino , Mutação em Linhagem Germinativa , Humanos , Deficiência Intelectual/mortalidade , Masculino , Linhagem , Fenótipo , Análise de Sequência de DNA
15.
Hepatology ; 57(6): 2458-68, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23325555

RESUMO

UNLABELLED: Patient-specific induced pluripotent stem cells (iPSCs) represent a potential source for developing novel drug and cell therapies. Although increasing numbers of disease-specific iPSCs have been generated, there has been limited progress in iPSC-based drug screening/discovery for liver diseases, and the low gene-targeting efficiency in human iPSCs warrants further improvement. Using iPSC lines from patients with alpha-1 antitrypsin (AAT) deficiency, for which there is currently no drug or gene therapy available, we established a platform to discover new drug candidates and correct disease-causing mutation with a high efficiency. A high-throughput format screening assay, based on our hepatic differentiation protocol, was implemented to facilitate automated quantification of cellular AAT accumulation using a 96-well immunofluorescence reader. To expedite the eventual application of lead compounds to patients, we conducted drug screening utilizing our established library of clinical compounds (the Johns Hopkins Drug Library) with extensive safety profiles. Through a blind large-scale drug screening, five clinical drugs were identified to reduce AAT accumulation in diverse patient iPSC-derived hepatocyte-like cells. In addition, using the recently developed transcription activator-like effector nuclease technology, we achieved high gene-targeting efficiency in AAT-deficiency patient iPSCs with 25%-33% of the clones demonstrating simultaneous targeting at both diseased alleles. The hepatocyte-like cells derived from the gene-corrected iPSCs were functional without the mutant AAT accumulation. This highly efficient and cost-effective targeting technology will broadly benefit both basic and translational applications. CONCLUSIONS: Our results demonstrated the feasibility of effective large-scale drug screening using an iPSC-based disease model and highly robust gene targeting in human iPSCs, both of which are critical for translating the iPSC technology into novel therapies for untreatable diseases.


Assuntos
Hepatócitos/efeitos dos fármacos , Hepatopatias/terapia , Células-Tronco Pluripotentes/efeitos dos fármacos , Reparo Gênico Alvo-Dirigido/métodos , Deficiência de alfa 1-Antitripsina/terapia , Diferenciação Celular , Células Cultivadas , Hepatócitos/citologia , Humanos , Hepatopatias/genética
16.
Blood ; 118(7): 1801-5, 2011 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-21628406

RESUMO

EBV-immortalized B lymphocyte cell lines have been widely banked for studying a variety of diseases, including rare genetic disorders. These cell lines represent an important resource for disease modeling with the induced pluripotent stem cell (iPSC) technology. Here we report the generation of iPSCs from EBV-immortalized B-cell lines derived from multiple inherited disease patients via a nonviral method. The reprogramming method for the EBV cell lines involves a distinct protocol compared with that of patient fibroblasts. The B-cell line-derived iPSCs expressed pluripotency markers, retained the inherited mutation and the parental V(D)J rearrangement profile, and differentiated into all 3 germ layer cell types. There was no integration of the reprogramming-related transgenes or the EBV-associated genes in these iPSCs. The ability to reprogram the widely banked patient B-cell lines will offer an unprecedented opportunity to generate human disease models and provide novel drug therapies.


Assuntos
Linfócitos B/citologia , Linfócitos B/virologia , Herpesvirus Humano 4/fisiologia , Células-Tronco Pluripotentes Induzidas/citologia , Linfócitos B/metabolismo , Diferenciação Celular , Linhagem Celular , Transformação Celular Viral , Células Cultivadas , Reprogramação Celular , Fibroblastos/citologia , Fibroblastos/metabolismo , Herpesvirus Humano 4/genética , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Transgenes
17.
Blood ; 117(18): 4773-7, 2011 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-21393480

RESUMO

Cryopreservation of hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs) is crucial for cord blood (CB) banking and transplantation. We evaluated recovery of functional HPC cryopreserved as mononuclear or unseparated cells for up to 23.5 years compared with prefreeze values of the same CB units. Highly efficient recovery (80%-100%) was apparent for granulocyte-macrophage and multipotential hematopoietic progenitors, although some collections had reproducible low recovery. Proliferative potential, response to multiple cytokines, and replating of HPC colonies was extensive. CD34(+) cells isolated from CB cryopreserved for up to 21 years had long-term (≥ 6 month) engrafting capability in primary and secondary immunodeficient mice reflecting recovery of long-term repopulating, self-renewing HSCs. We recovered functionally responsive CD4(+) and CD8(+) T lymphocytes, generated induced pluripotent stem (iPS) cells with differentiation representing all 3 germ cell lineages in vitro and in vivo, and detected high proliferative endothelial colony forming cells, results of relevance to CB biology and banking.


Assuntos
Preservação de Sangue , Criopreservação , Sangue Fetal/citologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Animais , Ensaio de Unidades Formadoras de Colônias , Células Endoteliais/citologia , Sangue Fetal/transplante , Transplante de Células-Tronco Hematopoéticas , Humanos , Técnicas In Vitro , Células-Tronco Pluripotentes Induzidas/transplante , Recém-Nascido , Subunidade gama Comum de Receptores de Interleucina/deficiência , Subunidade gama Comum de Receptores de Interleucina/genética , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/imunologia , Fatores de Tempo , Transplante Heterólogo
18.
Stem Cell Res ; 69: 103113, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37156062

RESUMO

T lymphocytes are the most abundant mononuclear blood cells and can serve as a source for generating induced pluripotent stem cells (iPSCs) for disease modeling or drug development. Here, we report the derivation of two iPSC lines from CD4+ helper T cells and CD8+ cytolytic T cells, respectively. The reprogramming was performed using Sendai virus encoding Klf-4, c-Myc, Oct-4 and Sox-2. Both iPSC lines displayed typical embryonic stem cell-like morphology and normal karyotype. Pluripotency was confirmed using immunocytochemistry methods and teratoma formation assay.


Assuntos
Células-Tronco Pluripotentes Induzidas , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Linfócitos T Citotóxicos/metabolismo , Linhagem Celular , Células-Tronco Embrionárias/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Reprogramação Celular , Diferenciação Celular
19.
Chemosphere ; 328: 138445, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-36963584

RESUMO

To mitigate the sudden increase in the production of waste engineering slurry, predominantly composed of Kaolinite, this study investigated the flocculation and dewatering of Kaolin slurry treated with single- and dual-polymer flocculants. The influence of the flocculant type and dosage, under single- and dual-dose conditions, on slurry's sedimentation and the filtration characteristics, were thoroughly discussed. The results reveal that the adsorption bridging of the polymeric flocculant, resulting from hydrogen bonds, exerts a more significant effect than electrical neutralization on forming a large floc. Under single-dose conditions, nonionic polyacrylamides (NPAMs) with the strongest adsorption bridging leads to biggest flocs and the maximum settling rate of 21.55 mm/s. Under the dual-dose conditions of polymeric aluminium chloride (PAC) and PAM, the size of the slurry's floc decreases with an increase in PAC dosage. Nevertheless, the filtration performance of the slurry improves, with the lowest SRF value of the flocculated slurry being 1.58 × 1011 m/kg as 3‰ PAC and 3‰ NPAM is dosed. The improvement is explained by the micro-pore distribution of sludge. According to Mercury intrusion porosimetry (MIP) test, the slurry treated with the optimal dosage of dual-polymer flocculant exhibits the greatest sludge pore size and connected porosity (with a maximum value of 20.99%). Furthermore, the study discusses and compares the flocculation mechanism of single- and dual-polymer flocculants. The obtained results provide guidance for selecting appropriate flocculants for dewatering inorganic slurries, using different dewatering methods, such as gravitational thickening or filter pressing.


Assuntos
Polímeros , Esgotos , Polímeros/química , Esgotos/química , Caulim , Floculação , Filtração , Cloreto de Alumínio
20.
Mol Imaging ; 10(2): 111-22, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21439256

RESUMO

During the last decade, there has been enormous progress in understanding both multipotent stem cells such as hematopoietic stem cells and pluripotent stem cells such as embryonic stem cells and induced pluripotent stem cells. However, it has been challenging to study developmental potentials of these stem cells because they reside in complex cellular environments and aspects of their distribution, migration, engraftment, survival, proliferation, and differentiation often could not be sufficiently elucidated based on limited snapshot images of location or environment or molecular markers. Therefore, reliable imaging methods to monitor or track the fate of the stem cells are highly desirable. Both short-term and more permanent monitoring of stem cells in cultures and in live organisms have benefited from recently developed imaging approaches that are designed to investigate cell behavior and function. Confocal and multiphoton microscopy, time-lapse imaging technology, and series of noninvasive imaging technologies enable us to investigate cell behavior in the context of a live organism. In turn, the knowledge gained has brought our understanding of stem cell biology to a new level. In this review, we discuss the application of current imaging modalities for research of hematopoietic stem cells and pluripotent stem cells and the challenges ahead.


Assuntos
Imagem Molecular/métodos , Pesquisa com Células-Tronco , Animais , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Coloração e Rotulagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA