Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Cell Physiol ; 238(1): 179-194, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36436185

RESUMO

Hemogenic endothelial (HE) cells are specialized endothelial cells to give rise to hematopoietic stem/progenitor cells during hematopoietic development. The underlying mechanisms that regulate endothelial-to-hematopoietic transition (EHT) of human HE cells are not fully understand. Here, we identified platelet endothelial aggregation receptor-1 (PEAR1) as a novel regulator of early hematopoietic development in human pluripotent stem cells (hPSCs). We found that the expression of PEAP1 was elevated during hematopoietic development. A subpopulation of PEAR1+ cells overlapped with CD34+ CD144+ CD184+ CD73- arterial-type HE cells. Transcriptome analysis by RNA sequencing indicated that TAL1/SCL, GATA2, MYB, RUNX1 and other key transcription factors for hematopoietic development were mainly expressed in PEAR1+ cells, whereas the genes encoding for niche-related signals, such as fibronectin, vitronectin, bone morphogenetic proteins and jagged1, were highly expressed in PEAR1- cells. The isolated PEAR1+ cells exhibited significantly greater EHT capacity on endothelial niche, compared with the PEAR1- cells. Colony-forming unit (CFU) assays demonstrated the multilineage hematopoietic potential of PEAR1+ -derived hematopoietic cells. Furthermore, PEAR1 knockout in hPSCs by CRISPR/Cas9 technology revealed that the hematopoietic differentiation was impaired, resulting in decreased EHT capacity, decreased expression of hematopoietic-related transcription factors, and increased expression of niche-related signals. In summary, this study revealed a novel role of PEAR1 in balancing intrinsic and extrinsic signals for early hematopoietic fate decision.


Assuntos
Hemangioblastos , Hematopoese , Células-Tronco Hematopoéticas , Células-Tronco Pluripotentes , Receptores de Superfície Celular , Humanos , Diferenciação Celular , Hemangioblastos/citologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Pluripotentes/citologia , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Fatores de Transcrição/metabolismo
2.
Blood ; 137(7): 959-968, 2021 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-33094331

RESUMO

Genome-wide association studies have identified common variants associated with platelet-related phenotypes, but because these variants are largely intronic or intergenic, their link to platelet biology is unclear. In 290 normal subjects from the GeneSTAR Research Study (110 African Americans [AAs] and 180 European Americans [EAs]), we generated whole-genome sequence data from whole blood and RNA sequence data from extracted nonribosomal RNA from 185 induced pluripotent stem cell-derived megakaryocyte (MK) cell lines (platelet precursor cells) and 290 blood platelet samples from these subjects. Using eigenMT software to select the peak single-nucleotide polymorphism (SNP) for each expressed gene, and meta-analyzing the results of AAs and EAs, we identify (q-value < 0.05) 946 cis-expression quantitative trait loci (eQTLs) in derived MKs and 1830 cis-eQTLs in blood platelets. Among the 57 eQTLs shared between the 2 tissues, the estimated directions of effect are very consistent (98.2% concordance). A high proportion of detected cis-eQTLs (74.9% in MKs and 84.3% in platelets) are unique to MKs and platelets compared with peak-associated SNP-expressed gene pairs of 48 other tissue types that are reported in version V7 of the Genotype-Tissue Expression Project. The locations of our identified eQTLs are significantly enriched for overlap with several annotation tracks highlighting genomic regions with specific functionality in MKs, including MK-specific DNAse hotspots, H3K27-acetylation marks, H3K4-methylation marks, enhancers, and superenhancers. These results offer insights into the regulatory signature of MKs and platelets, with significant overlap in genes expressed, eQTLs detected, and enrichment within known superenhancers relevant to platelet biology.


Assuntos
Plaquetas/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Megacariócitos/metabolismo , RNA/genética , Transcriptoma , Adulto , População Negra/genética , Plaquetas/citologia , Células Cultivadas , Feminino , Ontologia Genética , Estudo de Associação Genômica Ampla , Humanos , Masculino , Megacariócitos/citologia , Especificidade de Órgãos , Polimorfismo de Nucleotídeo Único , Locos de Características Quantitativas , RNA/biossíntese , RNA-Seq , População Branca/genética , Sequenciamento Completo do Genoma
3.
Biochem Biophys Res Commun ; 553: 165-171, 2021 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-33773139

RESUMO

The COVID-19 pandemic has caused significant morbidity and mortality. There is an urgent need for serological tests to detect antibodies against SARS-CoV-2, which could be used to assess past infection, evaluate responses to vaccines in development, and determine individuals who may be protected from future infection. Current serological tests developed for SARS-CoV-2 rely on traditional technologies such as enzyme-linked immunosorbent assays (ELISA) and lateral flow assays, which have not scaled to meet the demand of hundreds of millions of antibody tests so far. Herein, we present an alternative method of antibody testing that depends on one protein reagent being added to patient serum/plasma or whole blood with direct, visual readout. Two novel fusion proteins, RBD-2E8 and B6-CH1-RBD, were designed to bind red blood cells (RBCs) via a single-chain variable fragment (scFv), thereby displaying the receptor-binding domain (RBD) of SARS-CoV-2 spike protein on the surface of RBCs. Mixing mammalian-derived RBD-2E8 and B6-CH1-RBD with convalescent COVID-19 patient serum and RBCs led to visible hemagglutination, indicating the presence of antibodies against SARS-CoV-2 RBD. B6-CH1-RBD made in bacteria was not as effective in inducing agglutination, indicating better recognition of RBD epitopes from mammalian cells. Given that our hemagglutination test uses methods routinely used in hospital clinical labs across the world for blood typing, we anticipate the test can be rapidly deployed at minimal cost. We anticipate our hemagglutination assay may find extensive use in low-resource settings for detecting SARS-CoV-2 antibodies.


Assuntos
Anticorpos Antivirais/análise , Anticorpos Antivirais/imunologia , Teste Sorológico para COVID-19/métodos , COVID-19/sangue , COVID-19/imunologia , Testes de Hemaglutinação/métodos , Sistemas Automatizados de Assistência Junto ao Leito , SARS-CoV-2/imunologia , Antígenos Virais/imunologia , COVID-19/diagnóstico , COVID-19/virologia , Teste Sorológico para COVID-19/economia , Eritrócitos/imunologia , Testes de Hemaglutinação/economia , Humanos , Sistemas Automatizados de Assistência Junto ao Leito/economia , Proteínas Recombinantes de Fusão/imunologia , Anticorpos de Cadeia Única/química , Anticorpos de Cadeia Única/imunologia , Glicoproteína da Espícula de Coronavírus/química , Glicoproteína da Espícula de Coronavírus/imunologia , Fatores de Tempo
4.
J Clin Microbiol ; 59(12): e0118621, 2021 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-34469185

RESUMO

Serologic point-of-care tests to detect antibodies against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are an important tool in the COVID-19 pandemic. The majority of current point-of-care antibody tests developed for SARS-CoV-2 rely on lateral flow assays, but these do not offer quantitative information. To address this, we developed a novel antibody test leveraging hemagglutination, employing a dry card format currently used for typing ABO blood groups. Two hundred COVID-19 patient and 200 control plasma samples were reconstituted with O-negative red blood cells (RBCs) to form whole blood and added to dried viral-antibody fusion protein, followed by a stirring step and a tilting step, 3-min incubation, and a second tilting step. The sensitivities of the hemagglutination test, Euroimmun IgG enzyme-linked immunosorbent assay (ELISA), and receptor binding domain (RBD)-based CoronaChek lateral flow assay were 87.0%, 86.5%, and 84.5%, respectively, using samples obtained from recovered COVID-19 individuals. Testing prepandemic samples, the hemagglutination test had a specificity of 95.5%, compared to 97.3% and 98.9% for the ELISA and CoronaChek, respectively. A distribution of agglutination strengths was observed in COVID-19 convalescent-phase plasma samples, with the highest agglutination score (4) exhibiting significantly higher neutralizing antibody titers than weak positives (2) (P < 0.0001). Strong agglutinations were observed within 1 min of testing, and this shorter assay time also increased specificity to 98.5%. In conclusion, we developed a novel rapid, point-of-care RBC agglutination test for the detection of SARS-CoV-2 antibodies that can yield semiquantitative information on neutralizing antibody titer in patients. The 5-min test may find use in determination of serostatus prior to vaccination, postvaccination surveillance, and travel screening.


Assuntos
COVID-19 , SARS-CoV-2 , Anticorpos Antivirais , Ensaio de Imunoadsorção Enzimática , Hemaglutinação , Testes de Hemaglutinação , Humanos , Pandemias , Sistemas Automatizados de Assistência Junto ao Leito , Sensibilidade e Especificidade
5.
Gastrointest Endosc ; 94(6): 1119-1130.e4, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34197834

RESUMO

BACKGROUND AND AIMS: Gene therapy could provide curative therapies to many inherited monogenic liver diseases. Clinical trials have largely focused on adeno-associated viruses (AAVs) for liver gene delivery. These vectors, however, are limited by small packaging size, capsid immune responses, and inability to redose. As an alternative, nonviral, hydrodynamic injection through vascular routes can successfully deliver plasmid DNA (pDNA) into mouse liver but has achieved limited success in large animal models. METHODS: We explored hydrodynamic delivery of pDNA through the biliary system into the liver of pigs using ERCP and a power injector to supply hydrodynamic force. Human factor IX (hFIX), deficient in hemophilia B, was used as a model gene therapy. RESULTS: Biliary hydrodynamic injection was well tolerated without significant changes in vital signs, liver enzymes, hematology, or histology. No off-target pDNA delivery to other organs was detected by polymerase chain reaction. Immunohistochemistry revealed that 50.19% of the liver stained positive for hFIX after hydrodynamic injection at 5.5 mg pDNA, with every hepatic lobule in all liver lobes demonstrating hFIX expression. hFIX-positive hepatocytes were concentrated around the central vein, radiating outward across all 3 metabolic zones. Biliary hydrodynamic injection in pigs resulted in significantly higher transfection efficiency than mouse vascular hydrodynamic injection at matched pDNA per liver weight dose (32.7%-51.9% vs 18.9%, P < .0001). CONCLUSIONS: Biliary hydrodynamic injection using ERCP can achieve higher transfection efficiency into hepatocytes compared with AAVs at magnitudes of less cost in a clinically relevant human-sized large animal. This technology may serve as a platform for gene therapy of human liver diseases.


Assuntos
Sistema Biliar , Hidrodinâmica , Animais , Técnicas de Transferência de Genes , Terapia Genética , Fígado , Camundongos , Suínos
6.
J Cell Physiol ; 235(9): 6257-6267, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-31994198

RESUMO

Ischemic heart disease and congestive heart failure are major contributors to high morbidity and mortality. Approximately 1.5 million cases of myocardial infarction occur annually in the United States; the yearly incidence rate is approximately 600 cases per 100,000 people. Although significant progress to improve the survival rate has been made by medications and implantable medical devices, damaged cardiomyocytes are unable to be recovered by current treatment strategies. After almost two decades of research, stem cell therapy has become a very promising approach to generate new cardiomyocytes and enhance the function of the heart. Along with clinical trials with stem cells conducted in cardiac regeneration, concerns regarding safety and potential risks have emerged. One of the contentious issues is the electrical dysfunctions of cardiomyocytes and cardiac arrhythmia after stem cell therapy. In this review, we focus on the cell sources currently used for stem cell therapy and discuss related arrhythmogenic risk.


Assuntos
Arritmias Cardíacas/patologia , Doenças Cardiovasculares/terapia , Miócitos Cardíacos/transplante , Transplante de Células-Tronco/efeitos adversos , Arritmias Cardíacas/etiologia , Arritmias Cardíacas/terapia , Doenças Cardiovasculares/complicações , Doenças Cardiovasculares/patologia , Humanos , Fatores de Risco
7.
J Cell Physiol ; 234(9): 16136-16147, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30740687

RESUMO

Distinct regions of the primitive streak (PS) have diverse potential to differentiate into several tissues, including the hematopoietic lineage originated from the posterior region of PS. Although various signaling pathways have been identified to promote the development of PS and its mesoderm derivatives, there is a large gap in our understanding of signaling pathways that regulate the hematopoietic fate of PS. Here, we defined the roles of Wnt, activin, and bone morphogenetic protein (BMP) signaling pathways in generating hematopoietic-fated PS from human pluripotent stem cells (hPSCs). We found that the synergistic balance of these signaling pathways was crucial for controlling the PS fate determination towards hematopoietic lineage via mesodermal progenitors. Although the induction of PS depends largely on the Wnt and activin signaling, the PS generated without BMP4 lacks the hematopoietic potential, indicating that the BMP signaling is necessary for the PS to acquire hematopoietic property. Appropriate levels of Wnt signaling is crucial for the development of PS and its specification to the hematopoietic lineage. Although the development of PS is less sensitive to activin or BMP signaling, the fate of PS to mesoderm progenitors and subsequent hematopoietic lineage is determined by appropriate levels of activin or BMP signaling. Collectively, our study demonstrates that Wnt, activin, and BMP signaling pathways play cooperative and distinct roles in regulating the fate determination of PS for hematopoietic development. Our understanding of the regulatory networks of hematopoietic-fated PS would provide important insights into early hematopoietic patterning and possible guidance for generating functional hematopoietic cells from hPSCs in vitro.

8.
J Cell Physiol ; 232(12): 3261-3272, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28079253

RESUMO

The lineage transition between epithelium and mesenchyme is a process known as epithelial-mesenchymal transition (EMT), by which polarized epithelial cells lose their adhesion property and obtain mesenchymal cell phenotypes. EMT is a biological process that is often involved in embryogenesis and diseases, such as cancer invasion and metastasis. The EMT and the reverse process, mesenchymal-epithelial transition (MET), also play important roles in stem cell differentiation and de-differentiation (or reprogramming). In this review, we will discuss current research progress of EMT in embryonic development, cellular differentiation and reprogramming, and cancer progression, all of which are representative models for researches of stem cell biology in normal and in diseases. Understanding of EMT and MET may help to identify specific markers to distinguish normal stem cells from cancer stem cells in future.


Assuntos
Diferenciação Celular , Transformação Celular Neoplásica , Transição Epitelial-Mesenquimal , Células-Tronco/citologia , Animais , Reprogramação Celular , Humanos
9.
J Cell Physiol ; 231(5): 1065-76, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26395760

RESUMO

Generation of fully functional hematopoietic multipotent progenitor (MPP) cells from human pluripotent stem cells (hPSCs) has a great therapeutic potential to provide an unlimited cell source for treatment of hematological disorders. We previously demonstrated that CD34(+) CD31(+) CD144(+) population derived from hPSCs contain hemato-endothelial progenitors (HEPs) that give rise to hematopoietic and endothelial cells. Here, we report a differentiation system to generate definitive hematopoietic MPP cells from HEPs via endothelial monolayer. In the presence of angiogenic factors, HEPs formed an endothelial monolayer, from which hematopoietic clusters emerged through the process of endothelial-to-hematopoietic transition (EHT). EHT was significantly enhanced by hematopoietic growth factors. The definitive MPP cells generated from endothelial monolayer were capable of forming multilineage hematopoietic colonies, giving rise to T lymphoid cells, and differentiating into enucleated erythrocytes. Emergence of hematopoietic cells from endothelial monolayer occurred transiently. Hematopoietic potential was lost during prolonged culture of HEPs in endothelial growth conditions. Our study demonstrated that CD34(+) CD31(+) CD144(+) HEPs gave rise to hematopoietic MPP cells via hemogenic endothelial cells that exist transiently. The established differentiation system provides a platform for future investigation of regulatory factors involved in de novo generation of hematopoietic MPP cells and their applications in transplantation.


Assuntos
Células Endoteliais/citologia , Hematopoese , Células-Tronco Multipotentes/citologia , Células-Tronco Pluripotentes/citologia , Antígenos CD/metabolismo , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Linhagem da Célula/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Células Eritroides/citologia , Células Eritroides/efeitos dos fármacos , Hematopoese/efeitos dos fármacos , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Células-Tronco Multipotentes/efeitos dos fármacos , Células-Tronco Pluripotentes/efeitos dos fármacos
10.
J Cell Biochem ; 116(7): 1179-89, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25740540

RESUMO

Human pluripotent stem cells (hPSCs), including human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs), provide a new cell source for regenerative medicine, disease modeling, drug discovery, and preclinical toxicity screening. Understanding of the onset and the sequential process of hematopoietic cells from differentiated hPSCs will enable the achievement of personalized medicine and provide an in vitro platform for studying of human hematopoietic development and disease. During embryogenesis, hemogenic endothelial cells, a specified subset of endothelial cells in embryonic endothelium, are the primary source of multipotent hematopoietic stem cells. In this review, we discuss current status in the generation of multipotent hematopoietic stem and progenitor cells from hPSCs via hemogenic endothelial cells. We also review the achievements in direct reprogramming from non-hematopoietic cells to hematopoietic stem and progenitor cells. Further characterization of hematopoietic differentiation in hPSCs will improve our understanding of blood development and expedite the development of hPSC-derived blood products for therapeutic purpose.


Assuntos
Células-Tronco Hematopoéticas/citologia , Células-Tronco Embrionárias Humanas/citologia , Células-Tronco Multipotentes/citologia , Diferenciação Celular , Células-Tronco Hematopoéticas/metabolismo , Humanos , Células-Tronco Multipotentes/metabolismo , Células-Tronco Pluripotentes/citologia , Medicina de Precisão , Transdução de Sinais
11.
J Cell Biochem ; 116(3): 467-75, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25359705

RESUMO

Eph receptor (Eph)-ephrin signaling plays an important role in organ development and tissue regeneration. Bidirectional signaling of EphB4-ephrinB2 regulates cardiovascular development. To assess the role of EphB4-ephrinB2 signaling in cardiac lineage development, we utilized two GFP reporter systems in embryonic stem (ES) cells, in which the GFP transgenes were expressed in Nkx2.5(+) cardiac progenitor cells and in α-MHC(+) cardiomyocytes, respectively. We found that both EphB4 and ephrinB2 were expressed in Nkx2.5-GFP(+) cardiac progenitor cells, but not in α-MHC-GFP(+) cardiomyocytes during cardiac lineage differentiation of ES cells. An antagonist of EphB4, TNYL-RAW peptides, that block the binding of EphB4 and ephrinB2, impaired cardiac lineage development in ES cells. Inhibition of EphB4-ephrinB2 signaling at different time points during ES cell differentiation demonstrated that the interaction of EphB4 and ephrinB2 was required for the early stage of cardiac lineage development. Forced expression of human full-length EphB4 or intracellular domain-truncated EphB4 in EphB4-null ES cells was established to investigate the role of EphB4-forward signaling in ES cells. Interestingly, while full-length EphB4 was able to restore the cardiac lineage development in EphB4-null ES cells, the truncated EphB4 that lacks the intracellular domain of tyrosine kinase and PDZ motif failed to rescue the defect of cardiomyocyte development, suggesting that EphB4 intracellular domain is essential for the development of cardiomyocytes. Our study provides evidence that receptor-kinase-dependent EphB4-forward signaling plays a crucial role in the development of cardiac progenitor cells.


Assuntos
Células-Tronco Embrionárias/citologia , Miócitos Cardíacos/citologia , Receptor EphB4/metabolismo , Transdução de Sinais , Animais , Diferenciação Celular , Linhagem Celular , Corpos Embrioides/citologia , Corpos Embrioides/metabolismo , Efrina-B2/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Proteína Homeobox Nkx-2.5 , Proteínas de Homeodomínio/metabolismo , Humanos , Camundongos , Cadeias Pesadas de Miosina/metabolismo , Ligação Proteica , Reação em Cadeia da Polimerase em Tempo Real , Fatores de Transcrição/metabolismo
12.
J Cell Biochem ; 116(12): 2735-43, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26012423

RESUMO

Patient-specific human induced-pluripotent stem cells (hiPSCs) represent important cell sources to treat patients with acquired blood disorders. To realize the therapeutic potential of hiPSCs, it is crucial to understand signals that direct hiPSC differentiation to a hematopoietic lineage fate. Our previous study demonstrated that CD34(+)CD31(+) cells derived from human pluripotent stem cells (hPSCs) contain hemato-endothelial progenitors (HEPs) that give rise to hematopoietic cells and endothelial cells. Here, we established a serum-free and feeder-free system to induce the differentiation of hPSC-derived CD34(+)CD31(+) progenitor cells to erythroid cells. We show that extracellular matrix (ECM) proteins promote the differentiation of CD34(+)CD31(+) progenitor cells into CD235a(+) erythroid cells through CD41(+)CD235a(+) megakaryocyte-erythroid progenitors (MEP). Erythropoietin (EPO) is a predominant factor for CD34(+)CD31(+) progenitor differentiation to erythroid cells, whereas transforming growth factor beta (TGF-ß) inhibits the development of CD34(+)CD31(+) progenitor cells. Apoptosis of progenitor cells is induced by TGF-ß in early erythroid differentiation. Suppression of TGF-ß signaling by SB431542 at early stage of CD34(+)CD31(+) progenitor differentiation induces the erythroid cell generation. Together, these findings suggest that TGF-ß suppression and EPO stimulation promote erythropoiesis of CD34(+)CD31(+) progenitor cells derived from hPSCs.


Assuntos
Células Eritroides/efeitos dos fármacos , Eritropoese/efeitos dos fármacos , Eritropoetina/antagonistas & inibidores , Células-Tronco Pluripotentes/metabolismo , Fator de Crescimento Transformador beta/antagonistas & inibidores , Antígenos CD34/metabolismo , Benzamidas/administração & dosagem , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Linhagem da Célula/efeitos dos fármacos , Dioxóis/administração & dosagem , Células Eritroides/citologia , Eritropoetina/genética , Células-Tronco Hematopoéticas/citologia , Humanos , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Células-Tronco Pluripotentes/efeitos dos fármacos , Células-Tronco Pluripotentes/transplante , Fator de Crescimento Transformador beta/genética
13.
Stem Cells ; 32(5): 1161-72, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24307629

RESUMO

Embryonic stem cells (ESCs), characterized by their ability to both self-renew and differentiate into multiple cell lineages, are a powerful model for biomedical research and developmental biology. Human and mouse ESCs share many features, yet have distinctive aspects, including fundamental differences in the signaling pathways and cell cycle controls that support self-renewal. Here, we explore the molecular basis of human ESC self-renewal using Bayesian network machine learning to integrate cell-type-specific, high-throughput data for gene function discovery. We integrated high-throughput ESC data from 83 human studies (~1.8 million data points collected under 1,100 conditions) and 62 mouse studies (~2.4 million data points collected under 1,085 conditions) into separate human and mouse predictive networks focused on ESC self-renewal to analyze shared and distinct functional relationships among protein-coding gene orthologs. Computational evaluations show that these networks are highly accurate, literature validation confirms their biological relevance, and reverse transcriptase polymerase chain reaction (RT-PCR) validation supports our predictions. Our results reflect the importance of key regulatory genes known to be strongly associated with self-renewal and pluripotency in both species (e.g., POU5F1, SOX2, and NANOG), identify metabolic differences between species (e.g., threonine metabolism), clarify differences between human and mouse ESC developmental signaling pathways (e.g., leukemia inhibitory factor (LIF)-activated JAK/STAT in mouse; NODAL/ACTIVIN-A-activated fibroblast growth factor in human), and reveal many novel genes and pathways predicted to be functionally associated with self-renewal in each species. These interactive networks are available online at www.StemSight.org for stem cell researchers to develop new hypotheses, discover potential mechanisms involving sparsely annotated genes, and prioritize genes of interest for experimental validation.


Assuntos
Diferenciação Celular , Proliferação de Células , Células-Tronco Embrionárias/citologia , Biologia de Sistemas/métodos , Algoritmos , Animais , Teorema de Bayes , Linhagem da Célula , Biologia Computacional/métodos , Células-Tronco Embrionárias/metabolismo , Redes Reguladoras de Genes , Humanos , Camundongos , Reprodutibilidade dos Testes , Transdução de Sinais
15.
Biochem Biophys Res Commun ; 448(2): 212-7, 2014 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-24780395

RESUMO

Mesenchymal stem cells (MSCs) can be polarized into two distinct populations, MSC1 and MSC2, by activation of different Toll-like receptors (TLRs). TLR4-primed MSC1 expressed proinflammatory factors, whereas TLR3-primed MSC2 expressed suppressive factors. However, little is known about the function of TLRs on B lymphocyte-related immune modulation. In this study, we investigated the expression of B cell activating factor (BAFF), a member of the tumor necrosis factor ligand superfamily with notable stimulating activity on B cells, in human MSCs (hMSCs) and in murine MSCs (mMSCs) after activation of TLRs. BAFF was increasingly expressed in presence of TLR4 agonist (lipopolysaccharide, LPS), while TLR2 agonist (Zymosan) and TLR3-agonist (polyinocinic-polycytidykic acid, poly I:C) had no effect on BAFF expression. In addition, we demonstrated that signaling pathways of NF-κB, p38 MAPK, and JNK were involved in TLR4-primed BAFF expression. Our results suggested that TLR4 and downstream pathways in MSCs exert an important function in B lymphocyte-related immune regulation. Further defining a homogeneous population of MSCs should provide insight into MSC-based immune-modulating therapy.


Assuntos
Fator Ativador de Células B/metabolismo , Células-Tronco Mesenquimais/metabolismo , Receptor 4 Toll-Like/metabolismo , Adulto , Animais , Fator Ativador de Células B/genética , Fator Ativador de Células B/imunologia , Diferenciação Celular/imunologia , Células Cultivadas , Humanos , Lipopolissacarídeos/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Pessoa de Meia-Idade , NF-kappa B/metabolismo , Poli I-C/farmacologia , Transdução de Sinais , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/imunologia , Zimosan/farmacologia
16.
Blood ; 115(9): 1709-17, 2010 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-20032500

RESUMO

Both extrinsic and intrinsic mechanisms tightly govern hematopoietic stem cell (HSC) decisions of self-renewal and differentiation. However, transcription factors that can selectively regulate HSC self-renewal division after stress remain to be identified. Slug is an evolutionarily conserved zinc-finger transcription factor that is highly expressed in primitive hematopoietic cells and is critical for the radioprotection of these key cells. We studied the effect of Slug in the regulation of HSCs in Slug-deficient mice under normal and stress conditions using serial functional assays. Here, we show that Slug deficiency does not disturb hematopoiesis or alter HSC homeostasis and differentiation in bone marrow but increases the numbers of primitive hematopoietic cells in the extramedullary spleen site. Deletion of Slug enhances HSC repopulating potential but not its homing and differentiation ability. Furthermore, Slug deficiency increases HSC proliferation and repopulating potential in vivo after myelosuppression and accelerates HSC expansion during in vitro culture. Therefore, we propose that Slug is essential for controlling the transition of HSCs from relative quiescence under steady-state condition to rapid proliferation under stress conditions. Our data suggest that inhibition of Slug in HSCs may present a novel strategy for accelerating hematopoietic recovery, thus providing therapeutic benefits for patients after clinical myelosuppressive treatment.


Assuntos
Hematopoese/fisiologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Regeneração/fisiologia , Fatores de Transcrição/deficiência , Animais , Transplante de Medula Óssea , Diferenciação Celular , Proliferação de Células , Ensaio de Unidades Formadoras de Colônias , Fluoruracila/toxicidade , Hematopoese Extramedular/fisiologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fatores de Transcrição da Família Snail , Estresse Fisiológico , Fatores de Transcrição/genética , Fatores de Transcrição/fisiologia
17.
Blood ; 115(12): 2412-9, 2010 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-20061560

RESUMO

Eph receptors and their ephrin ligands are involved in normal hematopoietic development and tumorigenesis. Using methylated CpG island amplification/DNA promoter microarray, we identified several EPH receptor and EPHRIN genes as potential hypermethylation targets in acute lymphoblastic leukemia (ALL). We subsequently studied the DNA methylation status of the Eph/ephrin family by bisulfite pyrosequencing. Hypermethylation of EPHA2, -A4, -A5, -A6, -A7, -A10, EPHB1, -B2, -B3, -B4, EFNA1, -A3, -A5, and EFNB1 and -B2 genes was detected in leukemia cell lines and primary ALL bone marrow samples. Expression analysis of EPHB4, EFNB2, and EFNA5 genes demonstrated that DNA methylation was associated with gene silencing. We cloned the promoter region of EPHB4 and demonstrated that promoter hypermethylation can result in EPHB4 transcriptional silencing. Restoration of EPHB4 expression by lentiviral transduction resulted in reduced proliferation and apoptotic cell death in Raji cells in which EPHB4 is methylated and silenced. Finally, we demonstrated that phosphorylated Akt is down-regulated in Raji cells transduced with EPHB4. These results suggest that epigenetic silencing by hypermethylation of EPH/EPHRIN family genes contributes to ALL pathogenesis and that EPHB4 can function as a tumor suppressor in ALL.


Assuntos
Metilação de DNA , Efrina-B2/genética , Epigênese Genética , Regulação Leucêmica da Expressão Gênica , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Receptor EphB4/genética , Apoptose/fisiologia , Divisão Celular/fisiologia , Efrina-B2/metabolismo , Efrina-B2/farmacologia , Efrinas/genética , Efrinas/metabolismo , Efrinas/farmacologia , Inativação Gênica , Genes Supressores de Tumor , Humanos , Células Jurkat , Família Multigênica/fisiologia , Análise de Sequência com Séries de Oligonucleotídeos , Leucemia-Linfoma Linfoblástico de Células Precursoras/mortalidade , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Prevalência , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor EphB4/metabolismo , Receptores da Família Eph/genética , Receptores da Família Eph/metabolismo , Análise de Sobrevida , Transdução Genética
18.
Blood ; 115(23): 4707-14, 2010 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-20360471

RESUMO

Bone marrow injury is a major adverse side effect of radiation and chemotherapy. Attempts to limit such damage are warranted, but their success requires a better understanding of how radiation and anticancer drugs harm the bone marrow. Here, we report one pivotal role of the BH3-only protein Puma in the radiosensitivity of hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs). Puma deficiency in mice confers resistance to high-dose radiation in a hematopoietic cell-autonomous manner. Unexpectedly, loss of one Puma allele is sufficient to confer mice radioresistance. Interestingly, null mutation in Puma protects both primitive and differentiated hematopoietic cells from damage caused by low-dose radiation but selectively protects HSCs and HPCs against high-dose radiation, thereby accelerating hematopoietic regeneration. Consistent with these findings, Puma is required for radiation-induced apoptosis in HSCs and HPCs, and Puma is selectively induced by irradiation in primitive hematopoietic cells, and this induction is impaired in Puma-heterozygous cells. Together, our data indicate that selective targeting of p53 downstream apoptotic targets may represent a novel strategy to protecting HSCs and HPCs in patients undergoing intensive cancer radiotherapy and chemotherapy.


Assuntos
Proteínas Reguladoras de Apoptose , Apoptose/efeitos da radiação , Raios gama/efeitos adversos , Células-Tronco Hematopoéticas/metabolismo , Tolerância a Radiação/fisiologia , Proteínas Supressoras de Tumor , Animais , Apoptose/fisiologia , Relação Dose-Resposta à Radiação , Camundongos , Camundongos Knockout , Mutação , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
19.
PLoS One ; 16(4): e0249931, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33909609

RESUMO

The biliary system is routinely accessed for clinical purposes via endoscopic retrograde cholangiopancreatography (ERCP). We previously pioneered ERCP-mediated hydrodynamic injection in large animal models as an innovative gene delivery approach for monogenic liver diseases. However, the procedure poses potential safety concerns related mainly to liver or biliary tree injury. Here, we sought to further define biliary hydrodynamic injection parameters that are well-tolerated in a human-sized animal model. ERCP was performed in pigs, and hydrodynamic injection carried out using a novel protocol to reduce duct wall stress. Each pig was subjected to multiple repeated injections to expedite testing and judge tolerability. Different injection parameters (volume, flow rate) and injection port diameters were tested. Vital signs were monitored throughout the procedure, and liver enzyme panels were collected pre- and post-procedure. Pigs tolerated repeated biliary hydrodynamic injections with only occasional, mild, isolated elevation in aspartate aminotransferase (AST), which returned to normal levels within one day post-injection. All other liver tests remained unchanged. No upper limit of volume tolerance was reached, which suggests the biliary tree can readily transmit fluid into the vascular space. Flow rates up to 10 mL/sec were also tolerated with minimal disturbance to vital signs and no anatomic rupture of bile ducts. Measured intrabiliary pressure was up to 150 mmHg, and fluid-filled vesicles were induced in liver histology at high flow rates, mimicking the changes in histology observed in mouse liver after hydrodynamic tail vein injection. Overall, our investigations in a human-sized pig liver using standard clinical equipment suggest that ERCP-guided hydrodynamic injection will be safely tolerated in patients. Future investigations will interrogate if higher flow rates and pressure mediate higher DNA delivery efficiencies.


Assuntos
Sistema Biliar/fisiologia , DNA/administração & dosagem , Terapia Genética/métodos , Hidrodinâmica , Animais , Aspartato Aminotransferases/sangue , Bilirrubina/sangue , Pressão Sanguínea , Colangiopancreatografia Retrógrada Endoscópica , Frequência Cardíaca , Fígado/metabolismo , Fígado/patologia , Camundongos , Camundongos Endogâmicos C57BL , Suínos
20.
Cell Prolif ; 54(4): e13012, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33656760

RESUMO

OBJECTIVES: Vitronectin (VTN) has been widely used for the maintenance and expansion of human pluripotent stem cells (hPSCs) as feeder-free conditions. However, the effect of VTN on hPSC differentiation remains unclear. Here, we investigated the role of VTN in early haematopoietic development of hPSCs. MATERIALS AND METHODS: A chemically defined monolayer system was applied to study the role of different matrix or basement membrane proteins in haematopoietic development of hPSCs. The role of integrin signalling in VTN-mediated haematopoietic differentiation was investigated by integrin antagonists. Finally, small interfering RNA was used to knock down integrin gene expression in differentiated cells. RESULTS: We found that the haematopoietic differentiation of hPSCs on VTN was far more efficient than that on Matrigel that is also often used for hPSC culture. VTN promoted the fate determination of endothelial-haematopoietic lineage during mesoderm development to generate haemogenic endothelium (HE). Moreover, we demonstrated that the signals through αvß3 and αvß5 integrins were required for VTN-promoted haematopoietic differentiation. Blocking αvß3 and αvß5 integrins by the integrin antagonists impaired the development of HE, but not endothelial-to-haematopoietic transition (EHT). Finally, both αvß3 and αvß5 were confirmed acting synergistically for early haematopoietic differentiation by knockdown the expression of αv, ß3 or ß5. CONCLUSION: The established VTN-based monolayer system of haematopoietic differentiation of hPSCs presents a valuable platform for further investigating niche signals involved in human haematopoietic development.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Integrina alfaVbeta3/metabolismo , Receptores de Vitronectina/metabolismo , Vitronectina/farmacologia , Adesão Celular/efeitos dos fármacos , Linhagem Celular , Regulação da Expressão Gênica , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Integrina alfaVbeta3/antagonistas & inibidores , Integrina alfaVbeta3/genética , Mesoderma/citologia , Mesoderma/crescimento & desenvolvimento , Mesoderma/metabolismo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Receptores de Vitronectina/antagonistas & inibidores , Receptores de Vitronectina/genética , Transdução de Sinais/efeitos dos fármacos , Venenos de Serpentes/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA