Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biomaterials ; 301: 122255, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37651922

RESUMO

To better understand sodium channel (SCN5A)-related cardiomyopathies, we generated ventricular cardiomyocytes from induced pluripotent stem cells obtained from a dilated cardiomyopathy patient harbouring the R222Q mutation, which is only expressed in adult SCN5A isoforms. Because the adult SCN5A isoform was poorly expressed, without functional differences between R222Q and control in both embryoid bodies and cell sheet preparations (cultured for 29-35 days), we created heart-on-a-chip biowires which promote myocardial maturation. Indeed, biowires expressed primarily adult SCN5A with R222Q preparations displaying (arrhythmogenic) short action potentials, altered Na+ channel biophysical properties and lower contractility compared to corrected controls. Comprehensive RNA sequencing revealed differential gene regulation between R222Q and control biowires in cellular pathways related to sarcoplasmic reticulum and dystroglycan complex as well as biological processes related to calcium ion regulation and action potential. Additionally, R222Q biowires had marked reductions in actin expression accompanied by profound sarcoplasmic disarray, without differences in cell composition (fibroblast, endothelial cells, and cardiomyocytes) compared to corrected biowires. In conclusion, we demonstrate that in addition to altering cardiac electrophysiology and Na+ current, the R222Q mutation also causes profound sarcomere disruptions and mechanical destabilization. Possible mechanisms for these observations are discussed.


Assuntos
Cardiomiopatia Dilatada , Células-Tronco Pluripotentes Induzidas , Adulto , Humanos , Miócitos Cardíacos , Cardiomiopatia Dilatada/genética , Células Endoteliais , Dispositivos Lab-On-A-Chip
2.
Sci Transl Med ; 12(562)2020 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-32967972

RESUMO

Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) offer an unprecedented opportunity to remuscularize infarcted human hearts. However, studies have shown that most hiPSC-CMs do not survive after transplantation into the ischemic myocardial environment, limiting their regenerative potential and clinical application. We established a method to improve hiPSC-CM survival by cotransplanting ready-made microvessels obtained from adipose tissue. Ready-made microvessels promoted a sixfold increase in hiPSC-CM survival and superior functional recovery when compared to hiPSC-CMs transplanted alone or cotransplanted with a suspension of dissociated endothelial cells in infarcted rat hearts. Microvessels showed unprecedented persistence and integration at both early (~80%, week 1) and late (~60%, week 4) time points, resulting in increased vessel density and graft perfusion, and improved hiPSC-CM maturation. These findings provide an approach to cell-based therapies for myocardial infarction, whereby incorporation of ready-made microvessels can improve functional outcomes in cell replacement therapies.


Assuntos
Células-Tronco Pluripotentes Induzidas , Infarto do Miocárdio , Animais , Diferenciação Celular , Células Endoteliais , Humanos , Microvasos , Infarto do Miocárdio/terapia , Miócitos Cardíacos , Ratos
3.
Sci Adv ; 6(12): eaay7629, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32440533

RESUMO

The ability to detect rare human pluripotent stem cells (hPSCs) in differentiated populations is critical for safeguarding the clinical translation of cell therapy, as these undifferentiated cells have the capacity to form teratomas in vivo. The detection of hPSCs must be performed using an approach compatible with traceable manufacturing of therapeutic cell products. Here, we report a novel microfluidic approach, stem cell quantitative cytometry (SCQC), for the quantification of rare hPSCs in hPSC-derived cardiomyocyte (CM) populations. This approach enables the ultrasensitive capture, profiling, and enumeration of trace levels of hPSCs labeled with magnetic nanoparticles in a low-cost, manufacturable microfluidic chip. We deploy SCQC to assess the tumorigenic risk of hPSC-derived CM populations in vivo. In addition, we isolate rare hPSCs from the differentiated populations using SCQC and characterize their pluripotency.


Assuntos
Miócitos Cardíacos , Células-Tronco Pluripotentes , Diferenciação Celular , Terapia Baseada em Transplante de Células e Tecidos , Humanos
4.
Sci Rep ; 10(1): 6919, 2020 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-32332814

RESUMO

To accelerate the cardiac drug discovery pipeline, we set out to develop a platform that would be capable of quantifying tissue-level functions such as contractile force and be amenable to standard multiwell-plate manipulations. We report a 96-well-based array of 3D human pluripotent stem cell (hPSC)-derived cardiac microtissues - termed Cardiac MicroRings (CaMiRi) - in custom 3D-print-molded multiwell plates capable of contractile force measurement. Within each well, two elastomeric microcantilevers are situated above a circumferential ramp. The wells are seeded with cell-laden collagen, which, in response to the gradual slope of the circumferential ramp, self-organizes around tip-gated microcantilevers to form contracting CaMiRi. The contractile force exerted by the CaMiRi is measured and calculated using the deflection of the cantilevers. Platform responses were robust and comparable across wells, and we used it to determine an optimal tissue formulation. We validated the contractile force response of CaMiRi using selected cardiotropic compounds with known effects. Additionally, we developed automated protocols for CaMiRi seeding, image acquisition, and analysis to enable the measurement of contractile force with increased throughput. The unique tissue fabrication properties of the platform, and the consequent effects on tissue function, were demonstrated upon adding hPSC-derived epicardial cells to the system. This platform represents an open-source contractile force screening system useful for drug screening and tissue engineering applications.


Assuntos
Células-Tronco Pluripotentes/citologia , Engenharia Tecidual/métodos , Animais , Automação , Cardiotônicos/farmacologia , Células Cultivadas , Coração/efeitos dos fármacos , Coração/fisiologia , Humanos , Camundongos , Contração Miocárdica/efeitos dos fármacos , Células-Tronco Pluripotentes/efeitos dos fármacos , Impressão Tridimensional
5.
ACS Biomater Sci Eng ; 3(9): 1911-1921, 2017 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-33440549

RESUMO

Animal models have been instrumental in providing insight into the molecular basis of disease. While such information has been successfully applied to the study of human disease, this translation would be significantly strengthened by the availability of models based on human cells. This would be particularly important for cardiovascular disease, as the physiology of human cardiomyocytes (CMs) differs significantly from rodents. Here, we have generated a three-dimensional human engineered cardiac tissue, termed biowire, from human embryonic stem cell-derived CMs to investigate the effects of chronic (7 day) treatment with isoproterenol, endothelin-1, or angiotensin II. We show that biowires chronically treated with either isoproterenol, endothelin-1, or angiotensin II have disrupted myofibril alignment and significantly reduced force of contraction. Isoproterenol-treated biowires have upregulated brain natriuretic peptide and atrial natriuretic peptide gene expression. Endothelin-1 and angiotensin II-treated biowires demonstrated a significantly increased cell size. Endothelin-1-treated biowires exhibited increased cardiac troponin secretion into the culture media. This demonstrates that human biowires treated for 7 days with isoproterenol, angiotensin II, or endothelin-1 exhibit some changes compatible with hypertrophic cardiomyopathy.

6.
Nat Commun ; 7: 10312, 2016 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-26785135

RESUMO

The therapeutic success of human stem cell-derived cardiomyocytes critically depends on their ability to respond to and integrate with the surrounding electromechanical environment. Currently, the immaturity of human cardiomyocytes derived from stem cells limits their utility for regenerative medicine and biological research. We hypothesize that biomimetic electrical signals regulate the intrinsic beating properties of cardiomyocytes. Here we show that electrical conditioning of human stem cell-derived cardiomyocytes in three-dimensional culture promotes cardiomyocyte maturation, alters their automaticity and enhances connexin expression. Cardiomyocytes adapt their autonomous beating rate to the frequency at which they were stimulated, an effect mediated by the emergence of a rapidly depolarizing cell population, and the expression of hERG. This rate-adaptive behaviour is long lasting and transferable to the surrounding cardiomyocytes. Thus, electrical conditioning may be used to promote cardiomyocyte maturation and establish their automaticity, with implications for cell-based reduction of arrhythmia during heart regeneration.


Assuntos
Canais de Potássio Éter-A-Go-Go/metabolismo , Miócitos Cardíacos/citologia , Diferenciação Celular/fisiologia , Conexinas/metabolismo , Canal de Potássio ERG1 , Estimulação Elétrica , Canais de Potássio Éter-A-Go-Go/genética , Humanos , Microscopia Eletrônica de Transmissão , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/ultraestrutura , Células-Tronco Pluripotentes/citologia
7.
J Cell Physiol ; 230(7): 1475-88, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25514832

RESUMO

Nutrient depletion triggers a series of adaptive processes as part of the unfolded protein response or UPR. These processes reduce stress to the endoplasmic reticulum by enhancing its protein folding capacity or ability to promote the degradation of dysfunctional proteins. Failure to restore ER homeostasis causes the activation of lethal pathways. The expression of a dominant negative mutant of C/EBPß (Δ184-C/EBPß) alters this balance in chicken embryo fibroblasts (CEF). As a result, CEF display enhanced survival upon prolonged nutrient depletion. Starved Δ184-C/EBPß-expressing CEF display pronounced features of autophagy characterized by the appearance of large vesicles containing amorphous material, the formation of smaller double-membrane vesicles (autophagosomes) and processing of LC3 and GABARAP. However, there were marked differences in the expression and processing of these proteins. In both normal and Δ184-C/EBPß expressing CEF, the lipidated form of LC3 (form II) accumulated during starvation but was detectable even when cells were actively dividing in complete medium. In contrast, GABARAP expression and lipidation were strongly stimulated in response to starvation. Inhibition of LC3 expression by RNA interference led to apoptosis in normal CEF even in the absence of starvation but stable and near complete repression of GABARAP was tolerated. Moreover, the inhibition of GABARAP enhanced CEF survival and abolished the expression of the pro-apoptotic CHOP factor in conditions of starvation, suggesting a reduced level of ER stress. Therefore, GABARAP is a determinant of apoptosis in CEF subjected to prolonged nutrient depletion.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Apoptose/fisiologia , Fibroblastos/citologia , Fibroblastos/fisiologia , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Sequência de Aminoácidos , Animais , Proteínas Estimuladoras de Ligação a CCAAT/genética , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Técnicas de Cultura de Células , Proliferação de Células , Embrião de Galinha , Galinhas , Regulação da Expressão Gênica/fisiologia , Proteínas Associadas aos Microtúbulos/genética , Mutação , Fator de Transcrição CHOP/genética , Fator de Transcrição CHOP/metabolismo
8.
Biomaterials ; 35(9): 2798-808, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24424206

RESUMO

The goal of cardiac tissue engineering is to restore function to the damaged myocardium with regenerative constructs. Human embryonic stem cell-derived cardiomyocytes (hESC-CMs) can produce viable, contractile, three-dimensional grafts that function in vivo. We sought to enhance the viability and functional maturation of cardiac tissue constructs by cyclical stretch. hESC-CMs seeded onto gelatin-based scaffolds underwent cyclical stretching. Histological analysis demonstrated a greater proportion of cardiac troponin T-expressing cells in stretched than non-stretched constructs, and flow sorting demonstrated a higher proportion of cardiomyocytes. Ultrastructural assessment showed that cells in stretched constructs had a more mature phenotype, characterized by greater cell elongation, increased gap junction expression, and better contractile elements. Real-time PCR revealed enhanced mRNA expression of genes associated with cardiac maturation as well as genes encoding cardiac ion channels. Calcium imaging confirmed that stretched constructs contracted more frequently, with shorter calcium cycle duration. Epicardial implantation of constructs onto ischemic rat hearts demonstrated the feasibility of this platform, with enhanced survival and engraftment of transplanted cells in the stretched constructs. This uniaxial stretching system may serve as a platform for the production of cardiac tissue-engineered constructs for translational applications.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/citologia , Miócitos Cardíacos/citologia , Estresse Mecânico , Engenharia Tecidual/métodos , Animais , Cálcio/metabolismo , Diferenciação Celular/genética , Linhagem Celular , Conexina 43/metabolismo , Modelos Animais de Doenças , Células-Tronco Embrionárias/ultraestrutura , Estudos de Viabilidade , Regulação da Expressão Gênica , Humanos , Traumatismo por Reperfusão Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/terapia , Miocárdio/patologia , Ratos , Ratos Nus , Alicerces Teciduais/química , Cicatrização
9.
Lab Chip ; 14(5): 869-82, 2014 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-24352498

RESUMO

Tissue engineering enables the generation of three-dimensional (3D) functional cardiac tissue for pre-clinical testing in vitro, which is critical for new drug development. However, current tissue engineering methods poorly recapitulate the architecture of oriented cardiac bundles with supporting capillaries. In this study, we designed a microfabricated bioreactor to generate 3D micro-tissues, termed biowires, using both primary neonatal rat cardiomyocytes and human embryonic stem cell (hESC) derived cardiomyocytes. Perfusable cardiac biowires were generated with polytetrafluoroethylene (PTFE) tubing template, and were integrated with electrical field stimulation using carbon rod electrodes. To demonstrate the feasibility of this platform for pharmaceutical testing, nitric oxide (NO) was released from perfused sodium nitroprusside (SNP) solution and diffused through the tubing. The NO treatment slowed down the spontaneous beating of cardiac biowires based on hESC derived cardiomyocytes and degraded the myofibrillar cytoskeleton of the cardiomyocytes within the biowires. The biowires were also integrated with electrical stimulation using carbon rod electrodes to further improve phenotype of cardiomyocytes, as indicated by organized contractile apparatus, higher Young's modulus, and improved electrical properties. This microfabricated platform provides a unique opportunity to assess pharmacological effects on cardiac tissue in vitro by perfusion in a cardiac bundle model, which could provide improved physiological relevance.


Assuntos
Materiais Biocompatíveis/metabolismo , Técnicas Analíticas Microfluídicas/métodos , Engenharia Tecidual , Animais , Materiais Biocompatíveis/química , Reatores Biológicos , Células Cultivadas , Módulo de Elasticidade , Estimulação Elétrica , Eletrodos , Células-Tronco Embrionárias/citologia , Géis/química , Humanos , Técnicas Analíticas Microfluídicas/instrumentação , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Óxido Nítrico/química , Óxido Nítrico/metabolismo , Perfusão , Politetrafluoretileno/química , Ratos
10.
Am J Pathol ; 183(3): 720-34, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23871585

RESUMO

Hypoplastic left heart syndrome (HLHS) is a severe cardiac malformation characterized by left ventricle (LV) hypoplasia and abnormal LV perfusion and oxygenation. We studied hypoxia-associated injury in fetal HLHS and human pluripotent stem cells during cardiac differentiation to assess the effect of microenvironmental perturbations on fetal cardiac reprogramming. We studied LV myocardial samples from 32 HLHS and 17 structurally normal midgestation fetuses. Compared with controls, the LV in fetal HLHS samples had higher nuclear expression of hypoxia-inducible factor-1α but lower angiogenic growth factor expression, higher expression of oncogenes and transforming growth factor (TGF)-ß1, more DNA damage and senescence with cell cycle arrest, fewer cardiac progenitors, myocytes and endothelial lineages, and increased myofibroblast population (P < 0.05 versus controls). Smooth muscle cells (SMCs) had less DNA damage compared with endothelial cells and myocytes. We recapitulated the fetal phenotype by subjecting human pluripotent stem cells to hypoxia during cardiac differentiation. DNA damage was prevented by treatment with a TGF-ß1 inhibitor (P < 0.05 versus nonhypoxic cells). The hypoplastic LV in fetal HLHS samples demonstrates hypoxia-inducible factor-1α up-regulation, oncogene-associated cellular senescence, TGF-ß1-associated fibrosis and impaired vasculogenesis. The phenotype is recapitulated by subjecting human pluripotent stem cells to hypoxia during cardiac differentiation and rescued by inhibition of TGF-ß1. This finding suggests that hypoxia may reprogram the immature heart and affect differentiation and development.


Assuntos
Reprogramação Celular , Senescência Celular , Feto/patologia , Síndrome do Coração Esquerdo Hipoplásico/embriologia , Síndrome do Coração Esquerdo Hipoplásico/patologia , Morfogênese , Miocárdio/patologia , Células-Tronco Pluripotentes/patologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular Transformada , Linhagem da Célula/efeitos dos fármacos , Reprogramação Celular/efeitos dos fármacos , Senescência Celular/efeitos dos fármacos , Dano ao DNA , Corpos Embrioides/efeitos dos fármacos , Corpos Embrioides/patologia , Feto/efeitos dos fármacos , Feto/metabolismo , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/patologia , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/embriologia , Ventrículos do Coração/patologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/patologia , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Morfogênese/efeitos dos fármacos , Mutagênicos/toxicidade , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Fenótipo , Células-Tronco Pluripotentes/efeitos dos fármacos , Células-Tronco Pluripotentes/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Regulação para Cima/efeitos dos fármacos
11.
Nat Methods ; 10(8): 781-7, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23793239

RESUMO

Directed differentiation protocols enable derivation of cardiomyocytes from human pluripotent stem cells (hPSCs) and permit engineering of human myocardium in vitro. However, hPSC-derived cardiomyocytes are reflective of very early human development, limiting their utility in the generation of in vitro models of mature myocardium. Here we describe a platform that combines three-dimensional cell cultivation with electrical stimulation to mature hPSC-derived cardiac tissues. We used quantitative structural, molecular and electrophysiological analyses to explain the responses of immature human myocardium to electrical stimulation and pacing. We demonstrated that the engineered platform allows for the generation of three-dimensional, aligned cardiac tissues (biowires) with frequent striations. Biowires submitted to electrical stimulation had markedly increased myofibril ultrastructural organization, elevated conduction velocity and improved both electrophysiological and Ca(2+) handling properties compared to nonstimulated controls. These changes were in agreement with cardiomyocyte maturation and were dependent on the stimulation rate.


Assuntos
Técnicas de Cultura de Células/métodos , Células-Tronco Pluripotentes Induzidas/citologia , Miocárdio/citologia , Miócitos Cardíacos/citologia , Engenharia Tecidual/métodos , Diferenciação Celular/fisiologia , Estimulação Elétrica , Fenômenos Eletrofisiológicos , Humanos , Microscopia Eletrônica de Transmissão , Miocárdio/ultraestrutura
12.
Cardiovasc Res ; 98(1): 125-35, 2013 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-23213107

RESUMO

AIMS: Despite the diverse developmental origins of vascular smooth muscle cells (VSMCs), recent attempts to generate VSMCs from human embryonic stem cells (hESCs) differentiated along various lineages did not yield distinct cell phenotypes. The aim of this study was to derive and characterize functional coronary-like VSMCs from hESCs using serum-free cardiac-directed differentiation. METHODS AND RESULTS: Embryoid bodies (EBs) from three pluripotent stem cell lines subjected to cardiac-directed differentiation in defined media were characterized over 30 days for VSMC-specific gene expression by qRT-PCR, immunofluorescence microscopy and fluorescence-activated cell sorting (FACS). EBs composed of cardiomyocytes, endothelial cells (ECs), fibroblasts, and VSMCs underwent FACS on d28 to reveal that the VSMCs form a distinct subpopulation, which migrate with ECs in an in vitro angiogenesis assay. To enrich for VSMCs, d28 EBs were dissociated and cultured as monolayers. Over several passages, mRNA and protein levels of cardiomyocyte, endothelial, and fibroblast markers were abolished, whereas those of mature VSMCs were unchanged. Vascular endothelial growth factor and basic fibroblast growth factor were critical for the separation of the cardiac and VSMC lineages in EBs, and for the enrichment of functional VSMCs in monolayer cultures. Calcium cycling and cell shortening responses to vasoconstrictors in hESC-derived VSMCs in vitro were indistinguishable from primary human coronary artery SMCs, and distinct from bladder and aorta SMCs. VSMCs identically derived from green fluorescent protein -expressing hESCs integrated in and contributed to new vessel formation in vivo. CONCLUSION: The ability to generate hESC-derived functional human coronary-like VSMCs in serum-free conditions has implications for disease modelling, drug screening, and regenerative therapies.


Assuntos
Diferenciação Celular , Vasos Coronários/citologia , Células-Tronco Embrionárias/citologia , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/citologia , Actinas/análise , Animais , Cálcio/metabolismo , Células Cultivadas , Meios de Cultura Livres de Soro , Fator 2 de Crescimento de Fibroblastos/fisiologia , Humanos , Camundongos , Camundongos SCID , Neovascularização Fisiológica , RNA Mensageiro/análise , Fator A de Crescimento do Endotélio Vascular/fisiologia , Vasoconstrição/efeitos dos fármacos
13.
Cell Stem Cell ; 8(2): 228-40, 2011 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-21295278

RESUMO

Efficient differentiation of embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) to a variety of lineages requires step-wise approaches replicating the key commitment stages found during embryonic development. Here we show that expression of PdgfR-α segregates mouse ESC-derived Flk-1 mesoderm into Flk-1(+)PdgfR-α(+) cardiac and Flk-1(+)PdgfR-α(-) hematopoietic subpopulations. By monitoring Flk-1 and PdgfR-α expression, we found that specification of cardiac mesoderm and cardiomyocytes is determined by remarkably small changes in levels of Activin/Nodal and BMP signaling. Translation to human ESCs and iPSCs revealed that the emergence of cardiac mesoderm could also be monitored by coexpression of KDR and PDGFR-α and that this process was similarly dependent on optimal levels of Activin/Nodal and BMP signaling. Importantly, we found that individual mouse and human pluripotent stem cell lines require optimization of these signaling pathways for efficient cardiac differentiation, illustrating a principle that may well apply in other contexts.


Assuntos
Ativinas/metabolismo , Proteínas Morfogenéticas Ósseas/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Proteína Nodal/metabolismo , Células-Tronco Pluripotentes/citologia , Animais , Diferenciação Celular/fisiologia , Citometria de Fluxo , Humanos , Camundongos , Células-Tronco Pluripotentes/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
PLoS Genet ; 3(8): e134, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17696614

RESUMO

Genome instability is a hallmark of cancer cells. One class of genome aberrations prevalent in tumor cells is termed gross chromosomal rearrangements (GCRs). GCRs comprise chromosome translocations, amplifications, inversions, deletion of whole chromosome arms, and interstitial deletions. Here, we report the results of a genome-wide screen in Saccharomyces cerevisiae aimed at identifying novel suppressors of GCR formation. The most potent novel GCR suppressor identified is BUD16, the gene coding for yeast pyridoxal kinase (Pdxk), a key enzyme in the metabolism of pyridoxal 5' phosphate (PLP), the biologically active form of vitamin B6. We show that Pdxk potently suppresses GCR events by curtailing the appearance of DNA lesions during the cell cycle. We also show that pharmacological inhibition of Pdxk in human cells leads to the production of DSBs and activation of the DNA damage checkpoint. Finally, our evidence suggests that PLP deficiency threatens genome integrity, most likely via its role in dTMP biosynthesis, as Pdxk-deficient cells accumulate uracil in their nuclear DNA and are sensitive to inhibition of ribonucleotide reductase. Since Pdxk links diet to genome stability, our work supports the hypothesis that dietary micronutrients reduce cancer risk by curtailing the accumulation of DNA damage and suggests that micronutrient depletion could be part of a defense mechanism against hyperproliferation.


Assuntos
Aberrações Cromossômicas , Cromossomos Fúngicos , Dano ao DNA , Genes Supressores , Fosfato de Piridoxal/fisiologia , Saccharomyces cerevisiae/genética , Quebras de DNA de Cadeia Dupla , Genes Supressores/fisiologia , Genes cdc , Técnicas Genéticas , Genoma Fúngico , Instabilidade Genômica , Células HeLa , Humanos , Modelos Biológicos , Piridoxal Quinase/genética , Piridoxal Quinase/fisiologia , Fosfato de Piridoxal/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/fisiologia , Supressão Genética
15.
J Biol Chem ; 278(44): 43846-54, 2003 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-12896981

RESUMO

Chicken embryo fibroblasts (CEF) express several growth arrest-specific (GAS) gene products in G0. In contact-inhibited cells, the expression of the most abundant of these proteins, the p20K lipocalin, is activated at the transcriptional level by C/EBPbeta. In this report, we describe the role of C/EBPbeta in CEF proliferation. We show that the expression of a dominant negative mutant of C/EBPbeta (designated Delta184-C/EBPbeta) completely inhibited p20K expression at confluence and stimulated the proliferation of CEF without inducing transformation. Mouse embryo fibroblasts nullizygous for C/EBPbeta had a proliferative advantage over cells with one or two functional copies of this gene. C/EBP inhibition enhanced the expression of the three major components of AP-1 in cycling CEF, namely c-Jun, JunD, and Fra-2, and stimulated AP-1 activity. In contrast, the over-expression of C/EBPbeta caused a dramatic reduction in the levels of AP-1 proteins. Therefore, C/EBPbeta is a negative regulator of AP-1 expression and activity in CEF. The expression of cyclin D1 and cell proliferation were stimulated by the dominant negative mutant of C/EBPbeta but not in the presence of TAM67, a dominant negative mutant of c-Jun and AP-1. CEF over-expressing c-Jun, and to a lesser extent JunD and Fra-2, did not growth arrest at high cell density and did not express p20K. Therefore, AP-1 interfered with the action of C/EBPbeta at high cell density, indicating that these factors play opposing roles in the control of GAS gene expression and CEF proliferation.


Assuntos
Proteínas Sanguíneas/fisiologia , Proteína beta Intensificadora de Ligação a CCAAT/fisiologia , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Fator de Transcrição AP-1/fisiologia , Animais , Proteínas Aviárias , Proteínas Sanguíneas/metabolismo , Western Blotting , Divisão Celular , Células Cultivadas , Embrião de Galinha , Ciclina D1/metabolismo , Fragmentação do DNA , Genes Dominantes , Lipocalinas , Mutação , Fatores de Tempo , Fator de Transcrição AP-1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...