Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
PLoS Pathog ; 14(11): e1007445, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30422992

RESUMO

Wolbachia is an intracellular bacterium that infects a remarkable range of insect hosts. Insects such as mosquitos act as vectors for many devastating human viruses such as Dengue, West Nile, and Zika. Remarkably, Wolbachia infection provides insect hosts with resistance to many arboviruses thereby rendering the insects ineffective as vectors. To utilize Wolbachia effectively as a tool against vector-borne viruses a better understanding of the host-Wolbachia relationship is needed. To investigate Wolbachia-insect interactions we used the Wolbachia/Drosophila model that provides a genetically tractable system for studying host-pathogen interactions. We coupled genome-wide RNAi screening with a novel high-throughput fluorescence in situ hybridization (FISH) assay to detect changes in Wolbachia levels in a Wolbachia-infected Drosophila cell line JW18. 1117 genes altered Wolbachia levels when knocked down by RNAi of which 329 genes increased and 788 genes decreased the level of Wolbachia. Validation of hits included in depth secondary screening using in vitro RNAi, Drosophila mutants, and Wolbachia-detection by DNA qPCR. A diverse set of host gene networks was identified to regulate Wolbachia levels and unexpectedly revealed that perturbations of host translation components such as the ribosome and translation initiation factors results in increased Wolbachia levels both in vitro using RNAi and in vivo using mutants and a chemical-based translation inhibition assay. This work provides evidence for Wolbachia-host translation interaction and strengthens our general understanding of the Wolbachia-host intracellular relationship.


Assuntos
Drosophila melanogaster/genética , Interações entre Hospedeiro e Microrganismos/genética , Wolbachia/genética , Animais , Culicidae , Drosophila/genética , Drosophila/microbiologia , Drosophila melanogaster/microbiologia , Genoma , Interações Hospedeiro-Patógeno/genética , Humanos , Hibridização in Situ Fluorescente/métodos , Mosquitos Vetores , Interferência de RNA , Simbiose , Vírus/genética , Sequenciamento Completo do Genoma/métodos
2.
Nature ; 493(7434): 632-7, 2013 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-23254936

RESUMO

Mitochondrial DNA mutations transmitted maternally within the oocyte cytoplasm often cause life-threatening disorders. Here we explore the use of nuclear genome transfer between unfertilized oocytes of two donors to prevent the transmission of mitochondrial mutations. Nuclear genome transfer did not reduce developmental efficiency to the blastocyst stage, and genome integrity was maintained provided that spontaneous oocyte activation was avoided through the transfer of incompletely assembled spindle-chromosome complexes. Mitochondrial DNA transferred with the nuclear genome was initially detected at levels below 1%, decreasing in blastocysts and stem-cell lines to undetectable levels, and remained undetectable after passaging for more than one year, clonal expansion, differentiation into neurons, cardiomyocytes or ß-cells, and after cellular reprogramming. Stem cells and differentiated cells had mitochondrial respiratory chain enzyme activities and oxygen consumption rates indistinguishable from controls. These results demonstrate the potential of nuclear genome transfer to prevent the transmission of mitochondrial disorders in humans.


Assuntos
DNA Mitocondrial/genética , Técnicas de Transferência Nuclear/normas , Oócitos , Linhagem Celular , Células Cultivadas , Criopreservação , Desenvolvimento Embrionário , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Genótipo , Humanos , Mitocôndrias/genética , Mitocôndrias/metabolismo , Oócitos/citologia , Oócitos/metabolismo
3.
Nat Methods ; 12(9): 885-92, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26237226

RESUMO

Induced pluripotent stem cells (iPSCs) are an essential tool for modeling how causal genetic variants impact cellular function in disease, as well as an emerging source of tissue for regenerative medicine. The preparation of somatic cells, their reprogramming and the subsequent verification of iPSC pluripotency are laborious, manual processes limiting the scale and reproducibility of this technology. Here we describe a modular, robotic platform for iPSC reprogramming enabling automated, high-throughput conversion of skin biopsies into iPSCs and differentiated cells with minimal manual intervention. We demonstrate that automated reprogramming and the pooled selection of polyclonal pluripotent cells results in high-quality, stable iPSCs. These lines display less line-to-line variation than either manually produced lines or lines produced through automation followed by single-colony subcloning. The robotic platform we describe will enable the application of iPSCs to population-scale biomedical problems including the study of complex genetic diseases and the development of personalized medicines.


Assuntos
Técnicas de Cultura Celular por Lotes/instrumentação , Separação Celular/instrumentação , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Técnicas Analíticas Microfluídicas/instrumentação , Robótica/instrumentação , Diferenciação Celular/fisiologia , Células Cultivadas , Desenho de Equipamento , Análise de Falha de Equipamento , Fibroblastos/citologia , Fibroblastos/fisiologia , Humanos
4.
Proc Natl Acad Sci U S A ; 107(23): 10644-8, 2010 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-20498068

RESUMO

A discrete population of splenocytes with attributes of dendritic cells (DCs) and coexpressing the B-cell marker CD19 is uniquely competent to express the T-cell regulatory enzyme indoleamine 2,3-dioxygenase (IDO) in mice treated with TLR9 ligands (CpGs). Here we show that IDO-competent cells express the B-lineage commitment factor Pax5 and surface immunoglobulins. CD19 ablation abrogated IDO-dependent T-cell suppression by DCs, even though cells with phenotypic attributes matching IDO-competent cells developed normally and expressed IDO in response to interferon gamma. Consequently, DCs and regulatory T cells (Tregs) did not acquire T-cell regulatory functions after TLR9 ligation, providing an alternative perspective on the known T-cell regulatory defects of CD19-deficient mice. DCs from B-cell-deficient mice expressed IDO and mediated T-cell suppression after TLR9 ligation, indicating that B-cell attributes were not essential for B-lymphoid IDO-competent cells to regulate T cells. Thus, IDO-competent cells constitute a distinctive B-lymphoid cell type with quintessential T-cell regulatory attributes and phenotypic features of both B cells and DCs.


Assuntos
Linfócitos B/imunologia , Células Dendríticas/imunologia , Indolamina-Pirrol 2,3,-Dioxigenase/imunologia , Linfócitos T/imunologia , Animais , Antígenos CD19/imunologia , Linfócitos B/citologia , Linfócitos B/enzimologia , Linhagem da Célula , Células Dendríticas/enzimologia , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Camundongos , Camundongos Knockout , Fator de Transcrição PAX5/metabolismo , Baço/imunologia , Linfócitos T/enzimologia , Receptor Toll-Like 9/imunologia
5.
Nat Commun ; 14(1): 1867, 2023 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-37015919

RESUMO

Metastatic melanoma develops once transformed melanocytic cells begin to de-differentiate into migratory and invasive melanoma cells with neural crest cell (NCC)-like and epithelial-to-mesenchymal transition (EMT)-like features. However, it is still unclear how transformed melanocytes assume a metastatic melanoma cell state. Here, we define DNA methylation changes that accompany metastatic progression in melanoma patients and discover Nuclear Receptor Subfamily 2 Group F, Member 2 - isoform 2 (NR2F2-Iso2) as an epigenetically regulated metastasis driver. NR2F2-Iso2 is transcribed from an alternative transcriptional start site (TSS) and it is truncated at the N-terminal end which encodes the NR2F2 DNA-binding domain. We find that NR2F2-Iso2 expression is turned off by DNA methylation when NCCs differentiate into melanocytes. Conversely, this process is reversed during metastatic melanoma progression, when NR2F2-Iso2 becomes increasingly hypomethylated and re-expressed. Our functional and molecular studies suggest that NR2F2-Iso2 drives metastatic melanoma progression by modulating the activity of full-length NR2F2 (Isoform 1) over EMT- and NCC-associated target genes. Our findings indicate that DNA methylation changes play a crucial role during metastatic melanoma progression, and their control of NR2F2 activity allows transformed melanocytes to acquire NCC-like and EMT-like features. This epigenetically regulated transcriptional plasticity facilitates cell state transitions and metastatic spread.


Assuntos
Melanoma , Neoplasias Cutâneas , Humanos , Linhagem Celular Tumoral , Melanoma/patologia , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo , Transição Epitelial-Mesenquimal/genética , Epigênese Genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Regulação Neoplásica da Expressão Gênica , Fator II de Transcrição COUP/metabolismo
6.
Proc Natl Acad Sci U S A ; 105(44): 17073-8, 2008 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-18952840

RESUMO

Topical application of phorbol myristate acetate (PMA) elicits intense local inflammation that facilitates outgrowth of premalignant lesions in skin after carcinogen exposure. The inflammatory response to PMA treatment activates immune stimulatory mechanisms. However, we show here that PMA exposure also induces plasmacytoid dendritic cells (pDCs) in local draining lymph nodes (dLNs) to express indoleamine 2,3 dioxygenase (IDO), which confers T cell suppressor activity on pDCs. The induced IDO-mediated inhibitory activity in this subset of pDCs was potent, dominantly suppressing the T cell stimulatory activity of other DCs that comprise the major fraction of dLN DCs. IDO induction in pDCs depended on inflammatory signaling by means of IFN type I and II receptors, the TLR/IL-1 signaling adaptor MyD88, and on cellular stress responses to amino acid withdrawal by means of the integrated stress response kinase GCN2. Consistent with the hypothesis that T cell suppressive, IDO(+) pDCs elicited by PMA exposure create local immune privilege that favors tumor development, IDO-deficient mice exhibited a robust tumor-resistant phenotype in the standard DMBA/PMA 2-stage carcinogenesis model of skin papilloma formation. Thus, IDO is a key immunosuppressive factor that facilitates tumor progression in this setting of chronic inflammation driven by repeated topical PMA exposure.


Assuntos
Dermatite de Contato/enzimologia , Tolerância Imunológica/imunologia , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Papiloma/imunologia , Neoplasias Cutâneas/imunologia , Animais , Células Dendríticas/citologia , Células Dendríticas/enzimologia , Células Dendríticas/imunologia , Dermatite de Contato/imunologia , Dermatite de Contato/patologia , Progressão da Doença , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Linfonodos/enzimologia , Linfonodos/imunologia , Linfonodos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Papiloma/patologia , Transdução de Sinais/imunologia , Neoplasias Cutâneas/patologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Acetato de Tetradecanoilforbol/farmacologia
7.
Mol Biol Cell ; 32(2): 169-185, 2021 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-33206585

RESUMO

We performed a high-throughput whole-genome RNAi screen to identify novel inhibitors of ciliogenesis in normal and basal breast cancer cells. Our screen uncovered a previously undisclosed, extensive network of genes linking integrin signaling and cellular adhesion to the extracellular matrix (ECM) with inhibition of ciliation in both normal and cancer cells. Surprisingly, a cohort of genes encoding ECM proteins was also identified. We characterized several ciliation inhibitory genes and showed that their silencing was accompanied by altered cytoskeletal organization and induction of ciliation, which restricts cell growth and migration in normal and breast cancer cells. Conversely, supplying an integrin ligand, vitronectin, to the ECM rescued the enhanced ciliation observed on silencing this gene. Aberrant ciliation could also be suppressed through hyperactivation of the YAP/TAZ pathway, indicating a potential mechanistic basis for our findings. Our findings suggest an unanticipated reciprocal relationship between ciliation and cellular adhesion to the ECM and provide a resource that could vastly expand our understanding of controls involving "outside-in" and "inside-out" signaling that restrain cilium assembly.


Assuntos
Cílios/genética , Testes Genéticos , Genoma Humano , Organogênese/genética , Transdução de Sinais/genética , Actinas/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Adesão Celular/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Matriz Extracelular/metabolismo , Feminino , Adesões Focais/metabolismo , Inativação Gênica , Estudos de Associação Genética , Humanos , Integrinas/metabolismo , Ligantes , RNA Interferente Pequeno/metabolismo , Supressão Genética
8.
Nat Struct Mol Biol ; 27(2): 179-191, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32042152

RESUMO

Long interspersed element-1 (LINE-1, or L1) is the only autonomous retrotransposon that is active in human cells. Different host factors have been shown to influence L1 mobility; however, systematic analyses of these factors are limited. Here, we developed a high-throughput microscopy-based retrotransposition assay that identified the double-stranded break (DSB) repair and Fanconi anemia (FA) factors active in the S/G2 phase as potent inhibitors and regulators of L1 activity. In particular, BRCA1, an E3 ubiquitin ligase with a key role in several DNA repair pathways, directly affects L1 retrotransposition frequency and structure and plays a distinct role in controlling L1 ORF2 protein translation through L1 mRNA binding. These results suggest the existence of a 'battleground' at the DNA replication fork between homologous recombination (HR) factors and L1 retrotransposons and reveal a potential role for L1 in the genotypic evolution of tumors characterized by BRCA1 and HR repair deficiencies.


Assuntos
Proteína BRCA1/metabolismo , Reparo do DNA , Elementos Nucleotídeos Longos e Dispersos , Fase S , Proteína BRCA1/genética , Sistemas CRISPR-Cas , Linhagem Celular , Quebras de DNA de Cadeia Dupla , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Deleção de Genes , Células HEK293 , Células HeLa , Recombinação Homóloga , Humanos , Microscopia
9.
Handb Exp Pharmacol ; (188): 165-96, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19031026

RESUMO

Mature dendritic cells (DCs) are potent stimulators of T cells that recognize antigens presented by the DCs. In this chapter we describe mature DCs that suppress T cell responses to antigens they present due to expression of the intracellular enzyme indoleamine 2,3 dioxygenase (IDO). IDO-competent DCs are a subset of plasmacytoid DCs that can be induced to express IDO under certain inflammatory conditions in humans and mice. Though rare, IDO-expressing DCs acquire potent T cell suppressor activity that may predominate over the T cell stimulatory functions of all other antigen-presenting cells in physiologic environments due in part, to cooperation with regulatory T cells. Thus, IDO-expressing DCs are critical regulators of adaptive immunity that contribute to a wide range of inflammatory disease processes. As such, manipulating IDO expression in DCs using IDO inhibitors or IDO inducers offers considerable opportunities to improve immunotherapies in a range of clinically-significant disease syndromes.


Assuntos
Células Dendríticas/enzimologia , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Inflamação/enzimologia , Linfócitos T Reguladores/imunologia , Animais , Apresentação de Antígeno , Diferenciação Celular , Células Dendríticas/imunologia , Indução Enzimática , Humanos , Imunocompetência , Imunoterapia/métodos , Indolamina-Pirrol 2,3,-Dioxigenase/biossíntese , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Inflamação/imunologia , Inflamação/terapia , Ativação Linfocitária
10.
Genetics ; 213(4): 1401-1414, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31666291

RESUMO

Long Interspersed Nuclear Element-1 (LINE-1, L1) is the only autonomous active transposable element in the human genome. The L1-encoded proteins ORF1p and ORF2p enable the element to jump from one locus to another via a "copy-and-paste" mechanism. ORF1p is an RNA-binding protein, and ORF2p has endonuclease and reverse transcriptase activities. The huge number of truncated L1 remnants in the human genome suggests that the host has likely evolved mechanisms to prevent full L1 replication, and thereby decrease the proliferation of active elements and reduce the mutagenic potential of L1. In turn, L1 appears to have a minimized length to increase the probability of successful full-length replication. This streamlining would be expected to lead to high information density. Here, we describe the construction and initial characterization of a library of 538 consecutive trialanine substitutions that scan along ORF1p and ORF2p to identify functionally important regions. In accordance with the streamlining hypothesis, retrotransposition was overall very sensitive to mutations in ORF1p and ORF2p; only 16% of trialanine mutants retained near-wild-type (WT) activity. All ORF1p mutants formed near-WT levels of mRNA transcripts and 75% formed near-WT levels of protein. Two ORF1p mutants presented a unique nucleolar-relocalization phenotype. Regions of ORF2p that are sensitive to mutagenesis but lack phylogenetic conservation were also identified. We provide comprehensive information on the regions most critical to retrotransposition. This resource will guide future studies of intermolecular interactions that form with RNA, proteins, and target DNA throughout the L1 life cycle.


Assuntos
Elementos Nucleotídeos Longos e Dispersos/genética , Mutagênese/genética , Motivos de Nucleotídeos/genética , Retroelementos/genética , Sequência de Aminoácidos , Sequência de Bases , Nucléolo Celular/metabolismo , Sequência Conservada , Células HeLa , Humanos , Modelos Moleculares , Mutação/genética , Fases de Leitura Aberta , Domínios Proteicos
11.
Oncogene ; 38(29): 5766-5777, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31239516

RESUMO

We previously showed that KLF4, a gene highly expressed in murine prostate stem cells, blocks the progression of indolent intraepithelial prostatic lesions into aggressive and rapidly growing tumors. Here, we show that the anti-tumorigenic effect of KLF4 extends to PC3 human prostate cancer cells growing in the bone. We compared KLF4 null cells with cells transduced with a DOX-inducible KLF4 expression system, and find KLF4 function inhibits PC3 growth in monolayer and soft agar cultures. Furthermore, KLF4 null cells proliferate rapidly, forming large, invasive, and osteolytic tumors when injected into mouse femurs, whereas KLF4 re-expression immediately after their intra-femoral inoculation blocks tumor development and preserves a normal bone architecture. KLF4 re-expression in established KLF4 null bone tumors inhibits their osteolytic effects, preventing bone fractures and inducing an osteogenic response with new bone formation. In addition to these profound biological changes, KLF4 also induces a transcriptional shift from an osteolytic program in KLF4 null cells to an osteogenic program. Importantly, bioinformatic analysis shows that genes regulated by KLF4 overlap significantly with those expressed in metastatic prostate cancer patients and in three individual cohorts with bone metastases, strengthening the clinical relevance of the findings in our xenograft model.


Assuntos
Neoplasias Ósseas/secundário , Fatores de Transcrição Kruppel-Like/fisiologia , Osteólise/fisiopatologia , Neoplasias da Próstata/patologia , Animais , Linhagem Celular Tumoral , Proliferação de Células/genética , Estudos de Coortes , Xenoenxertos , Humanos , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Masculino , Camundongos , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo
12.
G3 (Bethesda) ; 8(10): 3293-3309, 2018 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-30126834

RESUMO

The proper accumulation and maintenance of stem cells is critical for organ development and homeostasis. The Notch signaling pathway maintains stem cells in diverse organisms and organ systems. In Caenorhabditis elegans, GLP-1/Notch activity prevents germline stem cell (GSC) differentiation. Other signaling mechanisms also influence the maintenance of GSCs, including the highly-conserved TOR substrate ribosomal protein S6 kinase (S6K). Although C. elegans bearing either a null mutation in rsks-1/S6K or a reduction-of-function (rf) mutation in glp-1/Notch produce half the normal number of adult germline progenitors, virtually all these single mutant animals are fertile. However, glp-1(rf) rsks-1(null) double mutant animals are all sterile, and in about half of their gonads, all GSCs differentiate, a distinctive phenotype associated with a significant reduction or loss of GLP-1 signaling. How rsks-1/S6K promotes GSC fate is unknown. Here, we determine that rsks-1/S6K acts germline-autonomously to maintain GSCs, and that it does not act through Cyclin-E or MAP kinase in this role. We found that interfering with translation also enhances glp-1(rf), but that regulation through rsks-1 cannot fully account for this effect. In a genome-scale RNAi screen for genes that act similarly to rsks-1/S6K, we identified 56 RNAi enhancers of glp-1(rf) sterility, many of which were previously not known to interact functionally with Notch. Further investigation revealed at least six candidates that, by genetic criteria, act linearly with rsks-1/S6K. These include genes encoding translation-related proteins, cacn-1/Cactin, an RNA exosome component, and a Hedgehog-related ligand. We found that additional Hedgehog-related ligands may share functional relationships with glp-1/Notch and rsks-1/S6K in maintaining germline progenitors.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Fertilidade/genética , Células Germinativas/metabolismo , Receptores Notch/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Células-Tronco/metabolismo , Animais , Proteínas de Caenorhabditis elegans/genética , Biologia Computacional/métodos , Ciclina E/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Ligação Proteica , Interferência de RNA
13.
Elife ; 72018 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-29309036

RESUMO

LINE-1/L1 retrotransposon sequences comprise 17% of the human genome. Among the many classes of mobile genetic elements, L1 is the only autonomous retrotransposon that still drives human genomic plasticity today. Through its co-evolution with the human genome, L1 has intertwined itself with host cell biology. However, a clear understanding of L1's lifecycle and the processes involved in restricting its insertion and intragenomic spread remains elusive. Here we identify modes of L1 proteins' entrance into the nucleus, a necessary step for L1 proliferation. Using functional, biochemical, and imaging approaches, we also show a clear cell cycle bias for L1 retrotransposition that peaks during the S phase. Our observations provide a basis for novel interpretations about the nature of nuclear and cytoplasmic L1 ribonucleoproteins (RNPs) and the potential role of DNA replication in L1 retrotransposition.


Assuntos
Ciclo Celular , Núcleo Celular/metabolismo , Ribonucleoproteínas/metabolismo , Humanos , Elementos Nucleotídeos Longos e Dispersos , Transporte Proteico
14.
Diabetes ; 63(3): 923-33, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24227685

RESUMO

Wolfram syndrome is an autosomal recessive disorder caused by mutations in WFS1 and is characterized by insulin-dependent diabetes mellitus, optic atrophy, and deafness. To investigate the cause of ß-cell failure, we used induced pluripotent stem cells to create insulin-producing cells from individuals with Wolfram syndrome. WFS1-deficient ß-cells showed increased levels of endoplasmic reticulum (ER) stress molecules and decreased insulin content. Upon exposure to experimental ER stress, Wolfram ß-cells showed impaired insulin processing and failed to increase insulin secretion in response to glucose and other secretagogues. Importantly, 4-phenyl butyric acid, a chemical protein folding and trafficking chaperone, restored normal insulin synthesis and the ability to upregulate insulin secretion. These studies show that ER stress plays a central role in ß-cell failure in Wolfram syndrome and indicate that chemical chaperones might have therapeutic relevance under conditions of ER stress in Wolfram syndrome and other forms of diabetes.


Assuntos
Estresse do Retículo Endoplasmático , Células-Tronco Pluripotentes Induzidas/citologia , Células Secretoras de Insulina/fisiologia , Síndrome de Wolfram/patologia , Animais , Cálcio/metabolismo , Diferenciação Celular , Humanos , Insulina/biossíntese , Insulina/metabolismo , Secreção de Insulina , Proteínas de Membrana/genética , Camundongos , Fenilbutiratos/farmacologia , Síndrome de Wolfram/genética
15.
PLoS One ; 9(1): e84547, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24416243

RESUMO

Presenilin 1 (PSEN1) encodes the catalytic subunit of γ-secretase, and PSEN1 mutations are the most common cause of early onset familial Alzheimer's disease (FAD). In order to elucidate pathways downstream of PSEN1, we characterized neural progenitor cells (NPCs) derived from FAD mutant PSEN1 subjects. Thus, we generated induced pluripotent stem cells (iPSCs) from affected and unaffected individuals from two families carrying PSEN1 mutations. PSEN1 mutant fibroblasts, and NPCs produced greater ratios of Aß42 to Aß40 relative to their control counterparts, with the elevated ratio even more apparent in PSEN1 NPCs than in fibroblasts. Molecular profiling identified 14 genes differentially-regulated in PSEN1 NPCs relative to control NPCs. Five of these targets showed differential expression in late onset AD/Intermediate AD pathology brains. Therefore, in our PSEN1 iPSC model, we have reconstituted an essential feature in the molecular pathogenesis of FAD, increased generation of Aß42/40, and have characterized novel expression changes.


Assuntos
Doença de Alzheimer/patologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Neurais/metabolismo , Presenilina-1/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Peptídeos beta-Amiloides/biossíntese , Animais , Apolipoproteínas E/genética , Proteínas Reguladoras de Apoptose , Sequência de Bases , Encéfalo/citologia , Encéfalo/patologia , Diferenciação Celular , Linhagem Celular , Proteínas do Olho/genética , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Genótipo , Humanos , Mutação , Proteínas do Tecido Nervoso/genética , Neurônios/citologia , Neurônios/patologia , Fragmentos de Peptídeos/biossíntese , Presenilina-1/genética , Ratos , Proteínas Supressoras da Sinalização de Citocina/genética
16.
PLoS One ; 8(3): e59867, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23555815

RESUMO

Current methods to derive induced pluripotent stem cell (iPSC) lines from human dermal fibroblasts by viral infection rely on expensive and lengthy protocols. One major factor contributing to the time required to derive lines is the ability of researchers to identify fully reprogrammed unique candidate clones from a mixed cell population containing transformed or partially reprogrammed cells and fibroblasts at an early time point post infection. Failure to select high quality colonies early in the derivation process results in cell lines that require increased maintenance and unreliable experimental outcomes. Here, we describe an improved method for the derivation of iPSC lines using fluorescence activated cell sorting (FACS) to isolate single cells expressing the cell surface marker signature CD13(NEG)SSEA4(POS)Tra-1-60(POS) on day 7-10 after infection. This technique prospectively isolates fully reprogrammed iPSCs, and depletes both parental and "contaminating" partially reprogrammed fibroblasts, thereby substantially reducing the time and reagents required to generate iPSC lines without the use of defined small molecule cocktails. FACS derived iPSC lines express common markers of pluripotency, and possess spontaneous differentiation potential in vitro and in vivo. To demonstrate the suitability of FACS for high-throughput iPSC generation, we derived 228 individual iPSC lines using either integrating (retroviral) or non- integrating (Sendai virus) reprogramming vectors and performed extensive characterization on a subset of those lines. The iPSC lines used in this study were derived from 76 unique samples from a variety of tissue sources, including fresh or frozen fibroblasts generated from biopsies harvested from healthy or disease patients.


Assuntos
Reprogramação Celular , Fibroblastos/citologia , Citometria de Fluxo , Células-Tronco Pluripotentes Induzidas/citologia , Pele/citologia , Adulto , Animais , Biópsia , Diferenciação Celular , Separação Celular , Células Cultivadas , Feminino , Humanos , Cariotipagem , Masculino , Camundongos , Pessoa de Meia-Idade , Pele/patologia , Teratoma/patologia , Fatores de Tempo
17.
Nat Biotechnol ; 29(3): 279-86, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21293464

RESUMO

Human induced pluripotent stem cells (iPSCs) present exciting opportunities for studying development and for in vitro disease modeling. However, reported variability in the behavior of iPSCs has called their utility into question. We established a test set of 16 iPSC lines from seven individuals of varying age, sex and health status, and extensively characterized the lines with respect to pluripotency and the ability to terminally differentiate. Under standardized procedures in two independent laboratories, 13 of the iPSC lines gave rise to functional motor neurons with a range of efficiencies similar to that of human embryonic stem cells (ESCs). Although three iPSC lines were resistant to neural differentiation, early neuralization rescued their performance. Therefore, all 16 iPSC lines passed a stringent test of differentiation capacity despite variations in karyotype and in the expression of early pluripotency markers and transgenes. This iPSC and ESC test set is a robust resource for those interested in the basic biology of stem cells and their applications.


Assuntos
Técnicas de Cultura de Células/métodos , Fibroblastos/citologia , Células-Tronco Pluripotentes/citologia , Pele/citologia , Engenharia Tecidual/métodos , Diferenciação Celular , Células Cultivadas , Humanos
18.
Eur J Immunol ; 37(4): 1064-71, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17343295

RESUMO

Following CD80/86 (B7) and TLR9 ligation, small subsets of splenic dendritic cells expressing CD19 (CD19(+) DC) acquire potent T cell regulatory functions due to induced expression of the intracellular enzyme indoleamine 2,3-dioxygenase (IDO), which catabolizes tryptophan. In CD19(+) DC, IFN type I (IFN-alpha) is the obligate inducer of IDO. We now report that IFN-alpha production needed to stimulate high-level expression of IDO following B7 ligation is itself dependent on basal levels of IDO activity. Genetic and pharmacologic ablation of IDO completely abrogated IFN-alpha production by CD19(+) DC after B7 ligation. In contrast, IDO ablation did not block IFN-alpha production by CD19(+) DC after TLR9 ligation. IDO-mediated control of IFN-alpha production depended on tryptophan depletion as adding excess tryptophan also blocked IFN-alpha expression after B7 ligation. Consistent with this, DC from mice deficient in general control of non-derepressible-2 (GCN2)-kinase, a component of the cellular stress response to amino acid withdrawal, did not produce IFN-alpha following B7 ligation, but produced IFN-alpha after TLR9 ligation. Thus, B7 and TLR9 ligands stimulate IFN-alpha expression in CD19(+) DC via distinct signaling pathways. In the case of B7 ligation, IDO activates cell-autonomous signals essential for IFN-alpha production, most likely by activating the GCN2-kinase-dependent stress response.


Assuntos
Antígenos CD19/biossíntese , Células Dendríticas/enzimologia , Regulação da Expressão Gênica/fisiologia , Indolamina-Pirrol 2,3,-Dioxigenase/fisiologia , Interferon-alfa/genética , Animais , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenase/deficiência , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Interferon-alfa/biossíntese , Camundongos , Camundongos Endogâmicos CBA , Camundongos Knockout , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
19.
Blood ; 110(4): 1199-206, 2007 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-17463170

RESUMO

Scurfy mice develop CD4 T-cell-mediated lymphoproliferative disease leading to death within 4 weeks of age. The scurfy mutation causes loss of function of the foxp3 gene (foxp3(sf)), which is essential for development and maintenance of naturally occurring regulatory CD4 T cells (nTregs). In humans, mutations of the foxp3 gene cause immune dysregulation, polyendocrinopathy, enteropathy, and X-linked syndrome (IPEX). In most patients with IPEX and also in scurfy mice, T cells show hyperreactivity and levels of Th1- and Th2-associated cytokines are substantially elevated. We report that removal of CD28 expression rescued scurfy mice from early death. Longer-term surviving CD28-deficient scurfy mice still had lymphoproliferative disorder, but their CD4 T cells showed decreased interferon-gamma and no sign of interleukin-4 or interleukin-10 hyperproduction. Furthermore, injection of CTLA4-Ig to block CD28-B7 interactions substantially improved the survival of scurfy mice by blocking effector T-cell differentiation. These data support the hypothesis that CD28-B7 interactions play a critical role in the etiology of lethal autoimmune disease in scurfy mice by stimulating the differentiation of antigen-activated naive T cells into effector T cells.


Assuntos
Doenças Autoimunes/terapia , Antígenos CD28/fisiologia , Fatores de Transcrição Forkhead/genética , Genes Letais , Transtornos Linfoproliferativos/terapia , Animais , Antígenos CD/metabolismo , Antígenos de Diferenciação/metabolismo , Doenças Autoimunes/imunologia , Doenças Autoimunes/mortalidade , Antígenos CD28/genética , Linfócitos T CD4-Positivos/metabolismo , Antígeno CTLA-4 , Proliferação de Células , Citocinas/metabolismo , Feminino , Humanos , Imunoglobulina G/metabolismo , Interferon gama/metabolismo , Interleucina-10/metabolismo , Interleucina-4/metabolismo , Transtornos Linfoproliferativos/imunologia , Transtornos Linfoproliferativos/mortalidade , Masculino , Camundongos , Camundongos Knockout , Camundongos Mutantes , Poliendocrinopatias Autoimunes/imunologia , Poliendocrinopatias Autoimunes/mortalidade , Poliendocrinopatias Autoimunes/terapia , Linfócitos T Reguladores/imunologia
20.
J Immunol ; 175(9): 5601-5, 2005 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-16237046

RESUMO

CpG oligodeoxynucleotides (CpG-ODNs) stimulate innate and adaptive immunity by binding to TLR9 molecules. Paradoxically, expression of the immunoregulatory enzyme indoleamine 2,3-dioxygenase (IDO) is induced following i.v. CpG-ODN administration to mice. CpG-ODNs induced selective IDO expression by a minor population of splenic CD19+ dendritic cells (DCs) that did not express the plasmacytoid DC marker 120G8. Following CpG-ODN treatment, CD19+ DCs acquired potent IDO-dependent T cell suppressive functions. Signaling through IFN type I receptors was essential for IDO up-regulation, and CpG-ODNs induced selective activation of STAT-1 in CD19+ DCs. Thus, CpG-ODNs delivered systemically at relatively high doses elicited potent T cell regulatory responses by acting on a discrete, minor population of splenic DCs. The ability of CpG-ODNs to induce both stimulatory and regulatory responses offers novel opportunities for using them as immunomodulatory reagents but may complicate therapeutic use of CpG-ODNs to stimulate antitumor immunity in cancer patients.


Assuntos
Adjuvantes Imunológicos/farmacologia , Antígenos CD19/análise , Células Dendríticas/efeitos dos fármacos , Indolamina-Pirrol 2,3,-Dioxigenase/fisiologia , Interferon-alfa/fisiologia , Oligodesoxirribonucleotídeos/farmacologia , Transdução de Sinais/fisiologia , Baço/imunologia , Linfócitos T Reguladores/fisiologia , Animais , Antígeno CD11c/análise , Células Dendríticas/enzimologia , Células Dendríticas/fisiologia , Indolamina-Pirrol 2,3,-Dioxigenase/biossíntese , Camundongos , Fator de Transcrição STAT1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA