Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Ther Methods Clin Dev ; 10: 313-326, 2018 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-30182035

RESUMO

In the present report, we carried out clinical-scale editing in adult mobilized CD34+ hematopoietic stem and progenitor cells (HSPCs) using zinc-finger nuclease-mediated disruption of BCL11a to upregulate the expression of γ-globin (fetal hemoglobin). In these cells, disruption of the erythroid-specific enhancer of the BCL11A gene increased endogenous γ-globin expression to levels that reached or exceeded those observed following knockout of the BCL11A coding region without negatively affecting survival or in vivo long-term proliferation of edited HSPCs and other lineages. In addition, BCL11A enhancer modification in mobilized CD34+ cells from patients with ß-thalassemia major resulted in a readily detectable γ-globin increase with a preferential increase in G-gamma, leading to an improved phenotype and, likely, a survival advantage for maturing erythroid cells after editing. Furthermore, we documented that both normal and ß-thalassemia HSPCs not only can be efficiently expanded ex vivo after editing but can also be successfully edited post-expansion, resulting in enhanced early in vivo engraftment compared with unexpanded cells. Overall, this work highlights a novel and effective treatment strategy for correcting the ß-thalassemia phenotype by genome editing.

2.
Cell Rep ; 18(13): 3117-3128, 2017 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-28355564

RESUMO

Histone citrullination regulates diverse cellular processes. Here, we report that SMARCAD1 preferentially associates with H3 arginine 26 citrullination (H3R26Cit) peptides present on arrays composed of 384 histone peptides harboring distinct post-transcriptional modifications. Among ten histone modifications assayed by ChIP-seq, H3R26Cit exhibited the most extensive genomewide co-localization with SMARCAD1 binding. Increased Smarcad1 expression correlated with naive pluripotency in pre-implantation embryos. In the presence of LIF, Smarcad1 knockdown (KD) embryonic stem cells lost naive state phenotypes but remained pluripotent, as suggested by morphology, gene expression, histone modifications, alkaline phosphatase activity, energy metabolism, embryoid bodies, teratoma, and chimeras. The majority of H3R26Cit ChIP-seq peaks occupied by SMARCAD1 were associated with increased levels of H3K9me3 in Smarcad1 KD cells. Inhibition of H3Cit induced H3K9me3 at the overlapping regions of H3R26Cit peaks and SMARCAD1 peaks. These data suggest a model in which SMARCAD1 regulates naive pluripotency by interacting with H3R26Cit and suppressing heterochromatin formation.


Assuntos
Citrulinação , Histonas/metabolismo , Proteínas Nucleares/metabolismo , Células-Tronco Pluripotentes/metabolismo , Animais , Sequência de Bases , Sítios de Ligação , Células Cultivadas , Cromatina/metabolismo , DNA Helicases , Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário , Células-Tronco Embrionárias/metabolismo , Epigênese Genética , Feminino , Técnicas de Silenciamento de Genes , Genoma , Lisina/metabolismo , Masculino , Metilação , Camundongos , Fenótipo , Ligação Proteica , Processamento de Proteína Pós-Traducional , Transcriptoma/genética
3.
Cancer Res ; 75(3): 594-604, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25589346

RESUMO

Ibrutinib (Imbruvica), a small-drug inhibitor of Bruton tyrosine kinase (BTK), is currently undergoing clinical testing in patients with multiple myeloma, yet important questions on the role of BTK in myeloma biology and treatment are outstanding. Using flow-sorted side population cells from human myeloma cell lines and multiple myeloma primary samples as surrogate for the elusive multiple myeloma stem cell, we found that elevated expression of BTK in myeloma cells leads to AKT/WNT/ß-catenin-dependent upregulation of key stemness genes (OCT4, SOX2, NANOG, and MYC) and enhanced self-renewal. Enforced transgenic expression of BTK in myeloma cells increased features of cancer stemness, including clonogenicity and resistance to widely used myeloma drugs, whereas inducible knockdown of BTK abolished them. Furthermore, overexpression of BTK in myeloma cells promoted tumor growth in laboratory mice and rendered side population-derived tumors that contained high levels of BTK more sensitive to the selective, second-generation BTK inhibitor, CGI1746, than side population-derived tumors that harbored low levels of BTK. Taken together, these findings implicate BTK as a positive regulator of myeloma stemness and provide additional support for the clinical testing of BTK-targeted therapies in patients with myeloma.


Assuntos
Mieloma Múltiplo/metabolismo , Células-Tronco Neoplásicas/citologia , Proteínas Tirosina Quinases/metabolismo , Adenina/análogos & derivados , Tirosina Quinase da Agamaglobulinemia , Animais , Antineoplásicos/química , Células da Medula Óssea/citologia , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Sobrevivência Celular , Humanos , Lentivirus/genética , Camundongos , Piperidinas , Pirazóis , Pirimidinas , Células da Side Population/citologia , Transdução de Sinais , beta Catenina/metabolismo
4.
Invest Ophthalmol Vis Sci ; 56(13): 8258-67, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26720480

RESUMO

PURPOSE: Age-related macular degeneration (AMD), the most common cause of incurable blindness in the western world, is characterized by the dysfunction and eventual death of choroidal endothelial (CECs), RPE, and photoreceptor cells. Stem cell-based treatment strategies designed to replace photoreceptor and RPE cells currently are a major scientific focus. However, the success of these approaches likely also will require replacement of the underlying, supportive choroidal vasculature. The purpose of this study was to generate stem cell-derived CECs to develop efficient differentiation and transplantation protocols. METHODS: Dermal fibroblasts from the Tie2-GFP mouse were isolated and reprogrammed into two independent induced pluripotent stem cell (iPSC) lines via viral transduction of the transcription factors Oct4, Sox2, Klf4, and c-Myc. Tie2-GFP iPSCs were differentiated into CECs using a coculture method with either the RF6A CEC line or primary mouse CECs. Induced pluripotent stem cell-derived CECs were characterized via RT-PCR and immunocytochemistry for EC- and CEC-specific markers. RESULTS: Induced pluripotent stem cells generated from mice expressing green fluorescent protein (GFP) under control of the endothelial Tie2 promoter display classic pluripotency markers and stem cell morphology. Induced pluripotent stem cell-derived CECs express carbonic anhydrase IV, eNOS, FOXA2, PLVAP, CD31, CD34, ICAM-1, Tie2, TTR, VE-cadherin, and vWF. CONCLUSIONS: Induced pluripotent stem cell-derived CECs will be a valuable tool for modeling of choriocapillaris-specific insults in AMD and for use in future choroidal endothelial cell replacement approaches.


Assuntos
Corioide/patologia , Células Endoteliais/patologia , Células-Tronco Pluripotentes Induzidas/citologia , Degeneração Macular/metabolismo , Animais , Animais Recém-Nascidos , Diferenciação Celular , Células Cultivadas , Células Endoteliais/metabolismo , Imuno-Histoquímica , Células-Tronco Pluripotentes Induzidas/metabolismo , Fator 4 Semelhante a Kruppel , Degeneração Macular/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
5.
Exp Hematol ; 42(9): 761-72.e1-10, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24852660

RESUMO

The development of a hematopoietic reporter is crucial for determining the fate of lineages derived from cell-based therapies. A marking system will enable safer embryonic stem and induced pluripotent stem cell-based derivation of blood lineages and facilitate the development of efficient cellular reprogramming strategies based on direct fibroblast conversion. Here we report that the protein tyrosine phosphatase CD45 is an ideal candidate gene on which to base a hematopoietic reporter. CD45 regulatory elements were discovered by analyzing transcription factor chromatin occupancy (ChIP-seq) and promoter nuclease sensitivity (DNase-seq) to identify minimally sufficient sequences required for expression. After cloning the CD45 regulatory elements into an attenuated lentiviral backbone, we found that two transcriptional initiation regions were essential for high-level expression. Expressing CD45 promoters containing these regions and tethered to green fluorescent protein (GFP) in a primary B-cell differentiation assay and a transplantation model resulted in high levels of GFP in lymphoid, myeloid, and nucleated erythroid cells in mouse and human blood cell lineages. Moreover, GFP levels remained high 5 months after secondary transplantation, indicating persistence of the reporter. No CD45-driven GFP expression is observed after fibroblast or embryonic stem cell transduction. The GFP reporter is seen only after embryonic stem cells differentiate into hematopoietic cell progenitors and lineages, suggesting that this hematopoietic reporter system could be useful in validating potential autologous blood cell therapies.


Assuntos
Genes Reporter , Transplante de Células-Tronco Hematopoéticas , Lentivirus , Antígenos Comuns de Leucócito , Elementos de Resposta , Transdução Genética , Animais , Proteínas de Fluorescência Verde/biossíntese , Humanos , Células Jurkat , Células K562 , Antígenos Comuns de Leucócito/genética , Antígenos Comuns de Leucócito/metabolismo , Células Progenitoras Linfoides/citologia , Células Progenitoras Linfoides/metabolismo , Camundongos , Células Progenitoras Mieloides/citologia , Células Progenitoras Mieloides/metabolismo
6.
PLoS One ; 8(12): e83624, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24349537

RESUMO

To develop stem/progenitor cell-based therapy for cystic fibrosis (CF) lung disease, it is first necessary to identify markers of human lung epithelial progenitor/stem cells and to better understand the potential for differentiation into distinct lineages. Here we investigated integrin α6ß4 as an epithelial progenitor cell marker in the human distal lung. We identified a subpopulation of α6ß4(+) cells that localized in distal small airways and alveolar walls and were devoid of pro-surfactant protein C expression. The α6ß4(+) epithelial cells demonstrated key properties of stem cells ex vivo as compared to α6ß4(-) epithelial cells, including higher colony forming efficiency, expression of stem cell-specific transcription factor Nanog, and the potential to differentiate into multiple distinct lineages including basal and Clara cells. Co-culture of α6ß4(+) epithelial cells with endothelial cells enhanced proliferation. We identified a subset of adeno-associated virus (AAVs) serotypes, AAV2 and AAV8, capable of transducing α6ß4(+) cells. In addition, reconstitution of bronchi epithelial cells from CF patients with only 5% normal α6ß4(+) epithelial cells significantly rescued defects in Cl(-) transport. Therefore, targeting the α6ß4(+) epithelial population via either gene delivery or progenitor cell-based reconstitution represents a potential new strategy to treat CF lung disease.


Assuntos
Fibrose Cística/metabolismo , Células Epiteliais/metabolismo , Integrina alfa6beta4/metabolismo , Pulmão/metabolismo , Mucosa Respiratória/metabolismo , Células-Tronco/metabolismo , Células Cultivadas , Fibrose Cística/genética , Fibrose Cística/patologia , Fibrose Cística/terapia , Dependovirus , Células Epiteliais/patologia , Feminino , Terapia Genética , Humanos , Integrina alfa6beta4/genética , Pulmão/patologia , Masculino , Mucosa Respiratória/patologia , Células-Tronco/patologia , Transdução Genética
7.
Blood ; 122(8): 1437-47, 2013 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-23847194

RESUMO

We previously demonstrated that RARα2 expression is increased in CD138 selected plasma cells of relapsed multiple myelomas (MMs), and increased expression was linked to poor prognosis in newly diagnosed MM patients. In the present study, we demonstrate that increased RARα2 confers myeloma stem cell features. Higher expression of RARα2 was identified in the multiple myeloma stem cell (MMSC) fraction. Overexpression of RARα2 in bulk MM cell lines resulted in: 1) increased drug resistance; 2) increased clonogenic potential; 3) activation of both Wnt and Hedgehog (Hh) pathways; 4) increased side population and aldehyde dehydrogenase levels; and 5) increased expression of embryonic stem cell genes. The opposite effects were seen with RARα2 knockdown. We demonstrate that RARα2 induces drug resistance by activating the drug efflux pump gene ABCC3 and anti-apoptotic Bcl-2 family members. Inhibition of Wnt signaling or ABCC3 function could overcome drug resistance in RARα2 overexpressing MM cells. We also showed that in the 5TGM1 mouse model, targeting of the Wnt and Hh pathways using CAY10404, cyclopamine, or itraconazole significantly reduced the myeloma tumor burden and increased survival. Targeting RARα2 or its downstream signaling pathways provides a potential strategy to eliminate MMSC.


Assuntos
Regulação Neoplásica da Expressão Gênica , Mieloma Múltiplo/metabolismo , Células-Tronco Neoplásicas/citologia , Receptores do Ácido Retinoico/metabolismo , Animais , Linhagem Celular Tumoral , Sobrevivência Celular , Perfilação da Expressão Gênica , Proteínas Hedgehog/metabolismo , Humanos , Isoxazóis/farmacologia , Itraconazol/farmacologia , Camundongos , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Plasmócitos/metabolismo , Prognóstico , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Receptor alfa de Ácido Retinoico , Transdução de Sinais , Sulfonas/farmacologia , Sindecana-1/metabolismo , Alcaloides de Veratrum/farmacologia , Proteínas Wnt/metabolismo
8.
Methods Mol Biol ; 1029: 191-203, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23756950

RESUMO

Embryonic stem cells (ESCs) are defined by their simultaneous capacity for limitless self-renewal and the ability to specify cells borne of all germ layers. The regulation of ESC pluripotency is governed by a set of core transcription factors that regulate transcription by interfacing with nuclear proteins that include the RNA polymerase II core transcriptional machinery, histone modification enzymes, and chromatin remodeling protein complexes. The growing adoption of systems biological approaches used in stem cell biology over last few years has contributed significantly to our understanding of pluripotency. Multilayered approaches coupling transcriptome profiling and proteomics (Nanog-, Oct4-, and Sox2-centered protein interaction networks or "interactomes") with transcription factor chromatin occupancy and epigenetic footprint measurements have enabled a more comprehensive understanding of ESC pluripotency and self-renewal. Together with the genetic and biochemical characterization of promising pluripotency modifying proteins, these systems biological approaches will continue to clarify the molecular underpinnings of the ESC state. This will most certainly contribute to the improvement of current methodologies for the derivation of pluripotent cells from adult tissues.


Assuntos
Linhagem da Célula/genética , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica , Transcrição Gênica , Animais , Humanos , MicroRNAs/metabolismo , Mapas de Interação de Proteínas/genética
9.
Nature ; 495(7441): 370-4, 2013 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-23395962

RESUMO

Molecular control of the pluripotent state is thought to reside in a core circuitry of master transcription factors including the homeodomain-containing protein NANOG, which has an essential role in establishing ground state pluripotency during somatic cell reprogramming. Whereas the genomic occupancy of NANOG has been extensively investigated, comparatively little is known about NANOG-associated proteins and their contribution to the NANOG-mediated reprogramming process. Using enhanced purification techniques and a stringent computational algorithm, we identify 27 high-confidence protein interaction partners of NANOG in mouse embryonic stem cells. These consist of 19 previously unknown partners of NANOG that have not been reported before, including the ten-eleven translocation (TET) family methylcytosine hydroxylase TET1. We confirm physical association of NANOG with TET1, and demonstrate that TET1, in synergy with NANOG, enhances the efficiency of reprogramming. We also find physical association and reprogramming synergy of TET2 with NANOG, and demonstrate that knockdown of TET2 abolishes the reprogramming synergy of NANOG with a catalytically deficient mutant of TET1. These results indicate that the physical interaction between NANOG and TET1/TET2 proteins facilitates reprogramming in a manner that is dependent on the catalytic activity of TET1/TET2. TET1 and NANOG co-occupy genomic loci of genes associated with both maintenance of pluripotency and lineage commitment in embryonic stem cells, and TET1 binding is reduced upon NANOG depletion. Co-expression of NANOG and TET1 increases 5-hydroxymethylcytosine levels at the top-ranked common target loci Esrrb and Oct4 (also called Pou5f1), resulting in priming of their expression before reprogramming to naive pluripotency. We propose that TET1 is recruited by NANOG to enhance the expression of a subset of key reprogramming target genes. These results provide an insight into the reprogramming mechanism of NANOG and uncover a new role for 5-methylcytosine hydroxylases in the establishment of naive pluripotency.


Assuntos
Reprogramação Celular/fisiologia , Proteínas de Ligação a DNA/metabolismo , Proteínas de Homeodomínio/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Animais , Proteínas de Ligação a DNA/genética , Dioxigenases , Células-Tronco Embrionárias , Regulação da Expressão Gênica no Desenvolvimento , Genoma , Proteínas de Homeodomínio/genética , Camundongos , Proteína Homeobox Nanog , Ligação Proteica , Proteínas Proto-Oncogênicas/genética
10.
J Stem Cell Res Ther ; Suppl 10: 009, 2012 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-25077040

RESUMO

INTRODUCTION: The potential of pluripotent stem cells to be used for cell therapy depends on a comprehensive understanding of the molecular mechanisms underlying their unique ability to specify cells of all germ layers while undergoing unlimited self-renewal. Alternative splicing and alternate promoter selection contribute to this mechanism by increasing the number of transcripts generated from a single gene locus and thus enabling expression of novel protein variants which may differ in their biological role. The homeodomain-containing transcription factor NANOG plays a critical role in maintaining the pluripotency of Embryonic Stem Cells (ESC). Therefore, a thorough understanding of the transcriptional regulation of the NANOG locus in ESCs is necessary. METHODS: Regulatory footprints and transcription levels were identified for NANOG in human embryonic stem cells from data obtained using high-throughput sequencing methodologies. Quantitative real-time PCR following reverse transcription of RNA extracted human ESCs was used to validate the expression of transcripts from a region that extends upstream of the annotated NANOG transcriptional start. Promoter identification and characterization were performed using promoter reporter and electrophoretic mobility shift assays. RESULTS: Transcriptionally active chromatin marking and transcription factor binding site enrichment were observed at a region upstream of the known transcriptional start site of NANOG. Expression of novel transcripts from this transcriptionally active region confirmed the existence of NANOG alternative splicing in human ESCs. We identified an alternate NANOG promoter of significant strength at this upstream region. We also discovered that NANOG autoregulates its expression by binding to its proximal downstream promoter. CONCLUSION: Our study reveals novel transcript expression from NANOG in human ESCs, indicating that alternative splicing increases the diversity of transcripts originating from the NANOG locus and that these transcripts are expressed by an alternate promoter. Alternative splicing and alternate promoter usage collaborate to regulate NANOG, enabling its function in the maintenance of ESCs.

11.
J Biol Chem ; 286(49): 42690-42703, 2011 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-21969378

RESUMO

Embryonic stem (ES) cells are distinguished by their ability to undergo unlimited self-renewal although retaining pluripotency, the capacity to specify cells of all germ layers. Alternative splicing contributes to these biological processes by vastly increasing the protein coding repertoire, enabling genes to code for novel variants that may confer different biological functions. The homeodomain transcription factor Nanog acts collaboratively with core factors Oct4 and Sox2 to govern the maintenance of pluripotency. We have discovered that Nanog is regulated by alternative splicing. Two novel exons and six subexons have been identified that extend the known Nanog gene structure and protein coding capacity. Alternative splicing results in two novel Nanog protein variants with attenuated capacities for self-renewal and pluripotency in ES cells. Our previous results have implicated the C-terminal domain, including the tryptophan-rich (WR) domain of Nanog, to be important for the function of Nanog (Wang, J., Levasseur, D. N., and Orkin, S. H. (2008) Proc. Natl. Acad. Sci. U.S.A. 105, 6326-6331). Using point mutation analyses, serine 2 (Ser-2) of Nanog has been identified as critical for ES cell self-renewal and for stabilizing a pluripotent gene signature. An inducible conditional knock-out was created to test the ability of new Nanog variants to genetically complement Nanog null ES cells. These results reveal for the first time an expanded Nanog protein coding capacity. We further reveal that a short region of the N-terminal domain and a single phosphorylatable Ser-2 is essential for the maintenance of self-renewal and pluripotency, demonstrating that this region of the protein is highly regulated.


Assuntos
Processamento Alternativo , Proteínas de Homeodomínio/genética , Animais , Diferenciação Celular , Linhagem Celular , RNA Polimerases Dirigidas por DNA/metabolismo , Células-Tronco Embrionárias/citologia , Variação Genética , Proteínas de Homeodomínio/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Proteína Homeobox Nanog , Células-Tronco Pluripotentes/citologia , Estrutura Terciária de Proteína , RNA/metabolismo , Células-Tronco/citologia , Transcrição Gênica
12.
Proc Natl Acad Sci U S A ; 105(17): 6326-31, 2008 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-18436640

RESUMO

Pluripotency of embryonic stem (ES) cells is maintained by transcription factors that form a highly interconnected protein interaction network surrounding the homeobox protein Nanog. Enforced expression of Nanog in mouse ES (mES) cells promotes self-renewal and alleviates their requirement for leukemia inhibitory factor (LIF). Understanding molecular mechanisms by which Nanog functions should illuminate fundamental properties of stem cells and the process of cellular reprogramming. Previously, we showed that Nanog forms multiple protein complexes in mES cells. Here, we demonstrate that Nanog dimerizes through its C-terminal domain rather than the homeodomain. Dimerization is required for interaction with other pluripotency network proteins. We also show that enforced expression of the Nanog dimer, but not the monomer, functionally replaces wild-type Nanog to sustain LIF-independent self-renewal of ES cells. Our results demonstrate that Nanog-Nanog homodimerization is a critical aspect of its function promoting stem cell pluripotency.


Assuntos
Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Proteínas de Homeodomínio/química , Proteínas de Homeodomínio/metabolismo , Células-Tronco Pluripotentes/citologia , Animais , Células COS , Proliferação de Células , Chlorocebus aethiops , DNA/metabolismo , Dimerização , Células-Tronco Embrionárias/metabolismo , Humanos , Fator Inibidor de Leucemia/metabolismo , Camundongos , Proteínas Mutantes/metabolismo , Proteína Homeobox Nanog , Ligação Proteica , Estrutura Terciária de Proteína
13.
Genes Dev ; 22(5): 575-80, 2008 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-18283123

RESUMO

Embryonic stem (ES) cells offer insight into early developmental fate decisions, and their controlled differentiation may yield vast regenerative potential. The molecular determinants supporting ES cell self-renewal are incompletely understood. The homeodomain proteins Nanog and Oct4 are essential for mouse ES cell self-renewal. Using a high-throughput approach, we discovered DNaseI hypersensitive sites and potential regulatory elements along a 160-kb region of the genome that includes GDF3, Dppa3, and Nanog. We analyzed gene expression, chromatin occupancy, and higher-order chromatin structure throughout this gene locus and found that expression of the reprogramming factor Oct4 is required to maintain its integrity.


Assuntos
Cromatina/metabolismo , Proteínas de Ligação a DNA/genética , Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Animais , Fator de Ligação a CCCTC , Diferenciação Celular/genética , Linhagem Celular , Cromatina/química , Cromatina/ultraestrutura , Proteínas Cromossômicas não Histona , Proteínas de Ligação a DNA/metabolismo , Desoxirribonuclease I/química , Células-Tronco Embrionárias/citologia , Fator 3 de Diferenciação de Crescimento , Elementos Isolantes , Peptídeos e Proteínas de Sinalização Intercelular/genética , Camundongos , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero/genética , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Fatores de Transcrição/metabolismo
14.
Nature ; 444(7117): 364-8, 2006 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-17093407

RESUMO

Embryonic stem (ES) cells are pluripotent and of therapeutic potential in regenerative medicine. Understanding pluripotency at the molecular level should illuminate fundamental properties of stem cells and the process of cellular reprogramming. Through cell fusion the embryonic cell phenotype can be imposed on somatic cells, a process promoted by the homeodomain protein Nanog, which is central to the maintenance of ES cell pluripotency. Nanog is thought to function in concert with other factors such as Oct4 (ref. 8) and Sox2 (ref. 9) to establish ES cell identity. Here we explore the protein network in which Nanog operates in mouse ES cells. Using affinity purification of Nanog under native conditions followed by mass spectrometry, we have identified physically associated proteins. In an iterative fashion we also identified partners of several Nanog-associated proteins (including Oct4), validated the functional relevance of selected newly identified components and constructed a protein interaction network. The network is highly enriched for nuclear factors that are individually critical for maintenance of the ES cell state and co-regulated on differentiation. The network is linked to multiple co-repressor pathways and is composed of numerous proteins whose encoding genes are putative direct transcriptional targets of its members. This tight protein network seems to function as a cellular module dedicated to pluripotency.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Células-Tronco Embrionárias/metabolismo , Proteínas de Homeodomínio/metabolismo , Células-Tronco Pluripotentes/metabolismo , Animais , Humanos , Imunoprecipitação , Camundongos , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Proteína Homeobox Nanog , Ligação Proteica , Interferência de RNA , Reprodutibilidade dos Testes
15.
Front Biosci ; 11: 1998-2006, 2006 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-16368574

RESUMO

MicroRNAs (miRNAs) have been suggested as suppressors of numerous target genes in human cells. In this report, we present gene chip array data indicating that in the absence of miRNA sequences, complete human introns are similarly capable of coordinating expression of large numbers of gene products at spatially diverse sites within the genome. The expression of selected intronic sequences (6a, 14b and 23) derived from the cystic fibrosis transmembrane conductance regulator (CFTR) gene caused extensive and specific transcriptional changes in epithelial cells (HeLa) that do not normally express this gene product. Each intron initiated a distinctive pattern of gene transcription. Affected genes such as FOXF1, sucrase-isomaltase, collagen, interferon, complement and thrombospondin 1 have previously been linked to CFTR function or are known to contribute to the related processes of epithelial differentiation and repair. A possible regulatory function of this nature has not been demonstrated previously for non-coding sequences within eukaryotic DNA. The results are consistent with the observation that splicesomal introns are found only in eukaryotic organisms and that the number of introns increases with phylogenetic complexity.


Assuntos
Regulação da Expressão Gênica , Íntrons , MicroRNAs/genética , Diferenciação Celular , Linhagem Celular , Biologia Computacional , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , DNA/química , Éxons , Genoma , Células HeLa , Humanos , Lentivirus/genética , Modelos Genéticos , Análise de Sequência com Séries de Oligonucleotídeos , Filogenia , RNA Mensageiro/metabolismo , Proteínas Recombinantes/química , Transdução de Sinais , Spliceossomos/metabolismo , Transcrição Gênica , Cicatrização
16.
Cancer Res ; 64(18): 6610-5, 2004 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-15374975

RESUMO

Escherichia coli purine nucleoside phosphorylase (PNP) expressed in tumors converts relatively nontoxic prodrugs into membrane-permeant cytotoxic compounds with high bystander activity. In the present study, we examined tumor regressions resulting from treatment with E. coli PNP and fludarabine phosphate (F-araAMP), a clinically approved compound used in the treatment of hematologic malignancies. We tested bystander killing with an adenoviral construct expressing E. coli PNP and then more formally examined thresholds for the bystander effect, using both MuLv and lentiviral vectoring. Because of the importance of understanding the mechanism of bystander action and the limits to this anticancer strategy, we also evaluated in vivo variables related to the expression of E. coli PNP (level of E. coli PNP activity in tumors, ectopic expression in liver, percentage of tumor cells transduced in situ, and accumulation of active metabolites in tumors). Our results indicate that F-araAMP confers excellent in vivo dose-dependent inhibition of bystander tumor cells, including strong responses in subcutaneous human glioma xenografts when 95 to 97.5% of the tumor mass is composed of bystander cells. These findings define levels of E. coli PNP expression necessary for antitumor activity with F-araAMP and demonstrate new potential for a clinically approved compound in solid tumor therapy.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Escherichia coli/genética , Terapia Genética/métodos , Purina-Núcleosídeo Fosforilase/genética , Fosfato de Vidarabina/análogos & derivados , Fosfato de Vidarabina/farmacologia , Adenoviridae/genética , Animais , Antimetabólitos Antineoplásicos/farmacocinética , Linhagem Celular Tumoral , Terapia Combinada , Relação Dose-Resposta a Droga , Escherichia coli/enzimologia , Vetores Genéticos/genética , Glioma/tratamento farmacológico , Glioma/enzimologia , Glioma/genética , Humanos , Lentivirus/genética , Camundongos , Camundongos Nus , Vírus da Leucemia Murina de Moloney/genética , Purina-Núcleosídeo Fosforilase/biossíntese , Purina-Núcleosídeo Fosforilase/metabolismo , Transfecção/métodos , Fosfato de Vidarabina/farmacocinética , Ensaios Antitumorais Modelo de Xenoenxerto
17.
J Biol Chem ; 279(26): 27518-24, 2004 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-15084588

RESUMO

A new recombinant, human anti-sickling beta-globin polypeptide designated beta(AS3) (betaGly(16) --> Asp/betaGlu(22) --> Ala/betaThr(87) --> Gln) was designed to increase affinity for alpha-globin. The amino acid substitutions at beta22 and beta87 are located at axial and lateral contacts of the sickle hemoglobin (HbS) polymers and strongly inhibit deoxy-HbS polymerization. The beta16 substitution confers the recombinant beta-globin subunit (beta(AS3)) with a competitive advantage over beta(S) for interaction with the alpha-globin polypeptide. Transgenic mouse lines that synthesize high levels of HbAS3 (alpha(2)beta(AS3)(2)) were established, and recombinant HbAS3 was purified from hemolysates and then characterized. HbAS3 binds oxygen cooperatively and has an oxygen affinity that is comparable with fetal hemoglobin. Delay time experiments demonstrate that HbAS3 is a potent inhibitor of HbS polymerization. Subunit competition studies confirm that beta(AS3) has a distinct advantage over beta(S) for dimerization with alpha-globin. When equal amounts of beta(S)- and beta(AS3)-globin monomers compete for limiting alpha-globin chains up to 82% of the tetramers formed is HbAS3. Knock-out transgenic mice that express exclusively human HbAS3 were produced. When these mice were bred with knock-out transgenic sickle mice the beta(AS3) polypeptides corrected all hematological parameters and organ pathology associated with the disease. Expression of beta(AS3)-globin should effectively lower the concentration of HbS in erythrocytes of patients with sickle cell disease, especially in the 30% percent of these individuals who coinherit alpha-thalassemia. Therefore, constructs expressing the beta(AS3)-globin gene may be suitable for future clinical trials for sickle cell disease.


Assuntos
Antidrepanocíticos/farmacologia , Hemoglobina Fetal/farmacologia , Globinas/genética , Hemoglobinas/farmacologia , Adulto , Anemia Falciforme/tratamento farmacológico , Anemia Falciforme/genética , Anemia Falciforme/patologia , Animais , Antidrepanocíticos/química , Modelos Animais de Doenças , Eritrócitos Anormais/citologia , Eritrócitos Anormais/metabolismo , Eritrócitos Anormais/ultraestrutura , Testes Hematológicos , Hemoglobinas/química , Hemoglobinas/genética , Humanos , Rim/metabolismo , Rim/patologia , Rim/ultraestrutura , Fígado/metabolismo , Fígado/patologia , Fígado/ultraestrutura , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Estrutura Quaternária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacologia , Baço/metabolismo , Baço/patologia , Baço/ultraestrutura
18.
Blood ; 102(13): 4312-9, 2003 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-12933581

RESUMO

Although sickle cell anemia was the first hereditary disease to be understood at the molecular level, there is still no adequate long-term treatment. Allogeneic bone marrow transplantation is the only available cure, but this procedure is limited to a minority of patients with an available, histocompatible donor. Autologous transplantation of bone marrow stem cells that are transduced with a stably expressed, antisickling globin gene would benefit a majority of patients with sickle cell disease. Therefore, the development of a gene therapy protocol that corrects the disease in an animal model and is directly translatable to human patients is critical. A method is described in which unmobilized, highly purified bone marrow stem cells are transduced with a minimum amount of self-inactivating (SIN) lentiviral vector containing a potent antisickling beta-globin gene. These cells, which were transduced in the absence of cytokine stimulation, fully reconstitute irradiated recipients and correct the hemolytic anemia and organ pathology that characterize the disease in humans. The mean increase of hemoglobin concentration was 46 g/L (4.6 g/dL) and the average lentiviral copy number was 2.2; therefore, a 21-g/L /vector copy increase (2.1-g/dL) was achieved. This transduction protocol may be directly translatable to patients with sickle cell disease who cannot tolerate current bone marrow mobilization procedures and may not safely be exposed to large viral loads.


Assuntos
Anemia Falciforme/terapia , Vetores Genéticos/uso terapêutico , Globinas/genética , Transplante de Células-Tronco Hematopoéticas , Lentivirus/genética , Anemia Falciforme/genética , Anemia Falciforme/patologia , Animais , Animais Congênicos , Separação Celular , Vírus Defeituosos/genética , Modelos Animais de Doenças , Feminino , Globinas/deficiência , Rim/patologia , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Quimera por Radiação , Baço/patologia , Transdução Genética , Talassemia beta/genética
19.
Chem Biol ; 10(12): 1173-81, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14700625

RESUMO

Activation of prodrugs by Escherichia coli purine nucleoside phosphorylase (PNP) provides a method for selectively killing tumor cells expressing a transfected PNP gene. This gene therapy approach requires matching a prodrug and a known enzymatic activity present only in tumor cells. The specificity of the method relies on avoiding prodrug cleavage by enzymes already present in the host cells or the intestinal flora. Using crystallographic and computer modeling methods as guides, we have redesigned E. coli PNP to cleave new prodrug substrates more efficiently than does the wild-type enzyme. In particular, the M64V PNP mutant cleaves 9-(6-deoxy-alpha-L-talofuranosyl)-6-methylpurine with a kcat/Km over 100 times greater than for native E. coli PNP. In a xenograft tumor experiment, this compound caused regression of tumors expressing the M64V PNP gene.


Assuntos
Escherichia coli/enzimologia , Terapia Genética/métodos , Pró-Fármacos/metabolismo , Pró-Fármacos/uso terapêutico , Engenharia de Proteínas , Purina-Núcleosídeo Fosforilase/química , Purina-Núcleosídeo Fosforilase/metabolismo , Animais , Sítios de Ligação , Linhagem Celular Tumoral , Simulação por Computador , Desenho Assistido por Computador , Cristalografia por Raios X , Escherichia coli/genética , Feminino , Humanos , Cinética , Camundongos , Modelos Moleculares , Estrutura Molecular , Mutação/genética , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/patologia , Especificidade de Órgãos , Pró-Fármacos/efeitos adversos , Conformação Proteica , Purina-Núcleosídeo Fosforilase/genética , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...