Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Biochem Biophys Res Commun ; 635: 244-251, 2022 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-36283337

RESUMO

Neural tube closure is a dynamic morphogenic event in early embryonic development. Perturbations of this process through either environmental or genetic factors induce the severe congenital malformations known collectively as neural tube defects (NTDs). Deficiencies in maternal folate intake have long been associated with NTDs, as have mutations in critical neurulation genes that include the Grainyhead-like 3 (Grhl3) gene. Mice lacking this gene exhibit fully penetrant thoraco-lumbo-sacral spina bifida and a low incidence of exencephaly. Previous studies have shown that exposure of pregnant mice carrying hypomorphic Grhl3 alleles to exogenous retinoic acid (RA) increases the incidence and severity of NTDs in their offspring. Here, we demonstrate that inhibition of RA signaling using a high affinity pan-RA receptor antagonist administered to pregnant mice at E7.5 induces fully penetrant exencephaly and more severe spina bifida in Grhl3-null mice. Later administration, although prior to neural tube closure has no effect. Similarly, blockade of RA in the context of reduced expression of Grhl2, a related gene known to induce NTDs, has no effect. Taken together, these findings provide new insights into the complexities of the interplay between RA signaling and Grhl3-induced neurulation.


Assuntos
Defeitos do Tubo Neural , Disrafismo Espinal , Gravidez , Feminino , Camundongos , Animais , Fatores de Transcrição/metabolismo , Neurulação/genética , Tubo Neural/metabolismo , Tretinoína/farmacologia , Tretinoína/metabolismo , Defeitos do Tubo Neural/metabolismo , Camundongos Knockout , Coluna Vertebral/metabolismo , Proteínas de Ligação a DNA/metabolismo
2.
Cell Rep ; 37(8): 110058, 2021 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-34818538

RESUMO

Mouse hematopoietic tissues contain abundant tissue-resident macrophages that support immunity, hematopoiesis, and bone homeostasis. A systematic strategy to characterize macrophage subsets in mouse bone marrow (BM), spleen, and lymph node unexpectedly reveals that macrophage surface marker staining emanates from membrane-bound subcellular remnants associated with unrelated cells. Intact macrophages are not present within these cell preparations. The macrophage remnant binding profile reflects interactions between macrophages and other cell types in vivo. Depletion of CD169+ macrophages in vivo eliminates F4/80+ remnant attachment. Remnant-restricted macrophage-specific membrane markers, cytoplasmic fluorescent reporters, and mRNA are all detected in non-macrophage cells including isolated stem and progenitor cells. Analysis of RNA sequencing (RNA-seq) data, including publicly available datasets, indicates that macrophage fragmentation is a general phenomenon that confounds bulk and single-cell analysis of disaggregated hematopoietic tissues. Hematopoietic tissue macrophage fragmentation undermines the accuracy of macrophage ex vivo molecular profiling and creates opportunity for misattribution of macrophage-expressed genes to non-macrophage cells.


Assuntos
Separação Celular/métodos , Macrófagos/citologia , Análise de Célula Única/métodos , Animais , Medula Óssea/metabolismo , Hematopoese , Células-Tronco Hematopoéticas/citologia , Homeostase , Camundongos
3.
Nat Commun ; 11(1): 3021, 2020 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-32541670

RESUMO

The caudal-related homeobox transcription factor CDX2 is expressed in leukemic cells but not during normal blood formation. Retroviral overexpression of Cdx2 induces AML in mice, however the developmental stage at which CDX2 exerts its effect is unknown. We developed a conditionally inducible Cdx2 mouse model to determine the effects of in vivo, inducible Cdx2 expression in hematopoietic stem and progenitor cells (HSPCs). Cdx2-transgenic mice develop myelodysplastic syndrome with progression to acute leukemia associated with acquisition of additional driver mutations. Cdx2-expressing HSPCs demonstrate enrichment of hematopoietic-specific enhancers associated with pro-differentiation transcription factors. Furthermore, treatment of Cdx2 AML with azacitidine decreases leukemic burden. Extended scheduling of low-dose azacitidine shows greater efficacy in comparison to intermittent higher-dose azacitidine, linked to more specific epigenetic modulation. Conditional Cdx2 expression in HSPCs is an inducible model of de novo leukemic transformation and can be used to optimize treatment in high-risk AML.


Assuntos
Fator de Transcrição CDX2/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Leucemia Mieloide Aguda/metabolismo , Síndromes Mielodisplásicas/metabolismo , Animais , Fator de Transcrição CDX2/genética , Transformação Celular Neoplásica , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Síndromes Mielodisplásicas/genética , Síndromes Mielodisplásicas/fisiopatologia
4.
BMC Genomics ; 20(1): 417, 2019 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-31126231

RESUMO

BACKGROUND: Mutations in the transcription factor, KLF1, are common within certain populations of the world. Heterozygous missense mutations in KLF1 mostly lead to benign phenotypes, but a heterozygous mutation in a DNA-binding residue (E325K in human) results in severe Congenital Dyserythropoietic Anemia type IV (CDA IV); i.e. an autosomal-dominant disorder characterized by neonatal hemolysis. RESULTS: To investigate the biochemical and genetic mechanism of CDA IV, we generated murine erythroid cell lines that harbor tamoxifen-inducible (ER™) versions of wild type and mutant KLF1 on a Klf1-/- genetic background. Nuclear translocation of wild type KLF1 results in terminal erythroid differentiation, whereas mutant KLF1 results in hemolysis without differentiation. The E to K variant binds poorly to the canonical 9 bp recognition motif (NGG-GYG-KGG) genome-wide but binds at high affinity to a corrupted motif (NGG-GRG-KGG). We confirmed altered DNA-binding specificity by quantitative in vitro binding assays of recombinant zinc-finger domains. Our results are consistent with previously reported structural data of KLF-DNA interactions. We employed 4sU-RNA-seq to show that a corrupted transcriptome is a direct consequence of aberrant DNA binding. CONCLUSIONS: Since all KLF/SP family proteins bind DNA in an identical fashion, these results are likely to be generally applicable to mutations in all family members. Importantly, they explain how certain mutations in the DNA-binding domain of transcription factors can generate neomorphic functions that result in autosomal dominant disease.


Assuntos
Anemia Diseritropoética Congênita/genética , DNA/metabolismo , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Mutação Puntual , Animais , Linhagem Celular , DNA/química , Regulação da Expressão Gênica , Camundongos , Motivos de Nucleotídeos , Ligação Proteica , Transcrição Gênica
5.
Blood Adv ; 3(3): 406-418, 2019 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-30733301

RESUMO

In normoxia, hypoxia-inducible transcription factors (HIFs) are rapidly degraded within the cytoplasm as a consequence of their prolyl hydroxylation by oxygen-dependent prolyl hydroxylase domain (PHD) enzymes. We have previously shown that hematopoietic stem and progenitor cells (HSPCs) require HIF-1 for effective mobilization in response to granulocyte colony-stimulating factor (G-CSF) and CXCR4 antagonist AMD3100/plerixafor. Conversely, HIF PHD inhibitors that stabilize HIF-1 protein in vivo enhance HSPC mobilization in response to G-CSF or AMD3100 in a cell-intrinsic manner. We now show that extrinsic mechanisms involving vascular endothelial growth factor receptor-2 (VEGFR2), via bone marrow (BM) endothelial cells, are also at play. PTK787/vatalanib, a tyrosine kinase inhibitor selective for VEGFR1 and VEGFR2, and neutralizing anti-VEGFR2 monoclonal antibody DC101 blocked enhancement of HSPC mobilization by FG-4497. VEGFR2 was absent on mesenchymal and hematopoietic cells and was detected only in Sca1+ endothelial cells in the BM. We propose that HIF PHD inhibitor FG-4497 enhances HSPC mobilization by stabilizing HIF-1α in HSPCs as previously demonstrated, as well as by activating VEGFR2 signaling in BM endothelial cells, which facilitates HSPC egress from the BM into the circulation.


Assuntos
Mobilização de Células-Tronco Hematopoéticas/métodos , Isoquinolinas/farmacologia , Inibidores de Prolil-Hidrolase/farmacologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ftalazinas/farmacologia , Piridinas/farmacologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
7.
PLoS One ; 12(7): e0180922, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28732065

RESUMO

Erythropoietin (EPO) acts through the dimeric erythropoietin receptor to stimulate proliferation, survival, differentiation and enucleation of erythroid progenitor cells. We undertook two complimentary approaches to find EPO-dependent pSTAT5 target genes in murine erythroid cells: RNA-seq of newly transcribed (4sU-labelled) RNA, and ChIP-seq for pSTAT5 30 minutes after EPO stimulation. We found 302 pSTAT5-occupied sites: ~15% of these reside in promoters while the rest reside within intronic enhancers or intergenic regions, some >100kb from the nearest TSS. The majority of pSTAT5 peaks contain a central palindromic GAS element, TTCYXRGAA. There was significant enrichment for GATA motifs and CACCC-box motifs within the neighbourhood of pSTAT5-bound peaks, and GATA1 and/or KLF1 co-occupancy at many sites. Using 4sU-RNA-seq we determined the EPO-induced transcriptome and validated differentially expressed genes using dynamic CAGE data and qRT-PCR. We identified known direct pSTAT5 target genes such as Bcl2l1, Pim1 and Cish, and many new targets likely to be involved in driving erythroid cell differentiation including those involved in mRNA splicing (Rbm25), epigenetic regulation (Suv420h2), and EpoR turnover (Clint1/EpsinR). Some of these new EpoR-JAK2-pSTAT5 target genes could be used as biomarkers for monitoring disease activity in polycythaemia vera, and for monitoring responses to JAK inhibitors.


Assuntos
Eritropoese/fisiologia , Eritropoetina/metabolismo , Fator de Transcrição STAT5/metabolismo , Animais , Western Blotting , Linhagem Celular , Imunoprecipitação da Cromatina , Eritropoese/genética , Eritropoetina/genética , Retroalimentação Fisiológica , Camundongos , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Fator de Transcrição STAT5/genética , Transdução de Sinais , Transcriptoma
9.
Nucleic Acids Res ; 45(11): 6572-6588, 2017 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-28541545

RESUMO

Krüppel-like factors (KLFs) are a family of 17 transcription factors characterized by a conserved DNA-binding domain of three zinc fingers and a variable N-terminal domain responsible for recruiting cofactors. KLFs have diverse functions in stem cell biology, embryo patterning, and tissue homoeostasis. KLF1 and related family members function as transcriptional activators via recruitment of co-activators such as EP300, whereas KLF3 and related members act as transcriptional repressors via recruitment of C-terminal Binding Proteins. KLF1 directly activates the Klf3 gene via an erythroid-specific promoter. Herein, we show KLF1 and KLF3 bind common as well as unique sites within the erythroid cell genome by ChIP-seq. We show KLF3 can displace KLF1 from key erythroid gene promoters and enhancers in vivo. Using 4sU RNA labelling and RNA-seq, we show this competition results in reciprocal transcriptional outputs for >50 important genes. Furthermore, Klf3-/- mice displayed exaggerated recovery from anemic stress and persistent cell cycling consistent with a role for KLF3 in dampening KLF1-driven proliferation. We suggest this study provides a paradigm for how KLFs work in incoherent feed-forward loops or networks to fine-tune transcription and thereby control diverse biological processes such as cell proliferation.


Assuntos
Elementos Facilitadores Genéticos , Fatores de Transcrição Kruppel-Like/metabolismo , Regiões Promotoras Genéticas , Ativação Transcricional , Animais , Linhagem Celular , Técnicas de Cocultura , Células Eritroides/metabolismo , Eritropoese , Camundongos , Transcrição Gênica
10.
Nucleic Acids Res ; 45(3): 1130-1143, 2017 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-28180284

RESUMO

The rules of engagement between zinc finger transcription factors and DNA have been partly defined by in vitro DNA-binding and structural studies, but less is known about how these rules apply in vivo. Here, we demonstrate how a missense mutation in the second zinc finger of Krüppel-like factor-1 (KLF1) leads to degenerate DNA-binding specificity in vivo, resulting in ectopic transcription and anemia in the Nan mouse model. We employed ChIP-seq and 4sU-RNA-seq to identify aberrant DNA-binding events genome wide and ectopic transcriptional consequences of this binding. We confirmed novel sequence specificity of the mutant recombinant zinc finger domain by performing biophysical measurements of in vitro DNA-binding affinity. Together, these results shed new light on the mechanisms by which missense mutations in DNA-binding domains of transcription factors can lead to autosomal dominant diseases.


Assuntos
DNA/metabolismo , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Transcriptoma/genética , Dedos de Zinco/genética , Animais , Linhagem Celular , Sobrevivência Celular/genética , Células Eritroides/metabolismo , Eritropoese/genética , Humanos , Fatores de Transcrição Kruppel-Like/química , Camundongos , Modelos Genéticos , Modelos Moleculares , Proteínas Mutantes/química , Mutação de Sentido Incorreto , Ligação Proteica
11.
J Mol Diagn ; 18(5): 707-718, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27449473

RESUMO

Myeloproliferative neoplasms (MPNs) are a heterogeneous group of blood disorders characterized by excess production of mature blood cells and an increased risk of late transformation to acute myeloid leukemia or primary myelofibrosis. Approximately 15% of MPN cases do not carry mutations in JAK2, CALR, or MPL and are thus often referred to as triple-negative cases. These are caused by a diverse set of rare mutations in cytokine receptors, JAK-STAT signaling pathway components, or epigenetic modifiers. In addition, some cases diagnosed as MPN are reactive rather than clonal disorders, so a negative result from a genetic screen can be informative. To obtain a comprehensive rapid molecular diagnosis for most MPNs, we developed an assay to detect genetic mutations (single nucleotide variants and/or small insertions/deletions) in 86 genes using targeted exon resequencing (AmpliSeq) and a bench-top semiconductor machine (Ion Torrent Personal Genome Machine). Our assay reliably detects well characterized mutations in JAK2, CALR, and MPL, but also rarer mutations in ASXL1, TET2, SH2B3, and other genes. Some of these mutations are novel. We find multiple mutations in advanced cases, suggesting co-operation between Janus kinase-STAT pathway mutations and epigenetic mutations in disease progression. This assay can be used to follow molecular progression, clonal heterogeneity, and drug resistance in MPNs.


Assuntos
Epigênese Genética , Éxons , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Transtornos Mieloproliferativos/diagnóstico , Transtornos Mieloproliferativos/genética , Transcriptoma , Adulto , Idoso , Idoso de 80 Anos ou mais , Alelos , Biomarcadores , Biologia Computacional/métodos , Feminino , Perfilação da Expressão Gênica/métodos , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Humanos , Janus Quinases/metabolismo , Masculino , Pessoa de Meia-Idade , Anotação de Sequência Molecular , Mutação , Transtornos Mieloproliferativos/metabolismo , Reprodutibilidade dos Testes , Fatores de Transcrição STAT/metabolismo , Sensibilidade e Especificidade , Transdução de Sinais
12.
Sci Rep ; 6: 28990, 2016 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-27350069

RESUMO

Prolonged high fat diets (HFD) induce low-grade chronic intestinal inflammation in mice, and diets high in saturated fat are a risk factor for the development of human inflammatory bowel diseases. We hypothesized that HFD-induced endoplasmic reticulum (ER)/oxidative stress occur in intestinal secretory goblet cells, triggering inflammatory signaling and reducing synthesis/secretion of proteins that form the protective mucus barrier. In cultured intestinal cells non-esterified long-chain saturated fatty acids directly increased oxidative/ER stress leading to protein misfolding. A prolonged HFD elevated the intestinal inflammatory cytokine signature, alongside compromised mucosal barrier integrity with a decrease in goblet cell differentiation and Muc2, a loss in the tight junction protein, claudin-1 and increased serum endotoxin levels. In Winnie mice, that develop spontaneous colitis, HFD-feeding increased ER stress, further compromised the mucosal barrier and increased the severity of colitis. In obese mice IL-22 reduced ER/oxidative stress and improved the integrity of the mucosal barrier, and reversed microbial changes associated with obesity with an increase in Akkermansia muciniphila. Consistent with epidemiological studies, our experiments suggest that HFDs are likely to impair intestinal barrier function, particularly in early life, which partially involves direct effects of free-fatty acids on intestinal cells, and this can be reversed by IL-22 therapy.


Assuntos
Colo/efeitos dos fármacos , Dieta Hiperlipídica , Células Epiteliais/efeitos dos fármacos , Inflamação/induzido quimicamente , Interleucinas/metabolismo , Mucosa Intestinal/efeitos dos fármacos , Estresse Fisiológico , Animais , Células Cultivadas , Colo/fisiologia , Citocinas/metabolismo , Células Epiteliais/fisiologia , Mucosa Intestinal/fisiologia , Camundongos Obesos , Muco/metabolismo , Interleucina 22
13.
Sci Rep ; 6: 26657, 2016 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-27226347

RESUMO

Thousands of sense-antisense mRNA-lncRNA gene pairs occur in the mammalian genome. While there is usually little doubt about the function of the coding transcript, the function of the lncRNA partner is mostly untested. Here we examine the function of the homeotic Evx1-Evx1as gene locus. Expression is tightly co-regulated in posterior mesoderm of mouse embryos and in embryoid bodies. Expression of both genes is enhanced by BMP4 and WNT3A, and reduced by Activin. We generated a suite of deletions in the locus by CRISPR-Cas9 editing. We show EVX1 is a critical downstream effector of BMP4 and WNT3A with respect to patterning of posterior mesoderm. The lncRNA, Evx1as arises from alternative promoters and is difficult to fully abrogate by gene editing or siRNA approaches. Nevertheless, we were able to generate a large 2.6 kb deletion encompassing the shared promoter with Evx1 and multiple additional exons of Evx1as. This led to an identical dorsal-ventral patterning defect to that generated by micro-deletion in the DNA-binding domain of EVX1. Thus, Evx1as has no function independent of EVX1, and is therefore unlikely to act in trans. We predict many antisense lncRNAs have no specific trans function, possibly only regulating the linked coding genes in cis.


Assuntos
Padronização Corporal/fisiologia , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/fisiologia , Embrião de Mamíferos/embriologia , Gastrulação/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas de Homeodomínio/biossíntese , RNA Longo não Codificante/biossíntese , Animais , Proteína Morfogenética Óssea 4/genética , Proteína Morfogenética Óssea 4/metabolismo , Sistemas CRISPR-Cas , Edição de Genes , Proteínas de Homeodomínio/genética , Camundongos , RNA Longo não Codificante/genética , Proteína Wnt3A/genética , Proteína Wnt3A/metabolismo
14.
Growth Factors ; 34(5-6): 210-216, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-28209092

RESUMO

Fibroblast growth factor-1 (FGF-1) promotes differentiation of human preadipocytes into mature adipocytes via modulation of a BMP and Activin Membrane-Bound Inhibitor (BAMBI)/Peroxisome proliferator-activated receptor (PPARγ)-dependent network. Here, we combined transcriptomic and functional investigations to identify novel downstream effectors aligned with complementary analyses of gene expression in human adipose tissue to explore relationships with insulin sensitivity. RNA-Seq and qRT-PCR analysis revealed significant down-regulation of carboxypeptidase A4 (CPA4) following FGF-1 treatment or induction of differentiation of human preadipocytes in a BAMBI/PPARγ-independent manner. siRNA-mediated knockdown of CPA4 resulted in enhanced differentiation of human preadipocytes. Furthermore, expression of CPA4 in subcutaneous adipose tissue correlated negatively with indices of local and systemic (liver and muscle) insulin sensitivity. These results identify CPA4 as a negative regulator of adipogenesis that is down-regulated by FGF-1 and a putative deleterious modulator of local and systemic insulin sensitivity. Further investigations are required to define the molecular mechanism(s) involved and potential therapeutic opportunities.


Assuntos
Adipócitos/metabolismo , Adipogenia , Carboxipeptidases A/metabolismo , Fator 1 de Crescimento de Fibroblastos/farmacologia , Resistência à Insulina , Adipócitos/citologia , Adipócitos/efeitos dos fármacos , Adulto , Carboxipeptidases A/genética , Células Cultivadas , Regulação para Baixo , Humanos , Fígado/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Pessoa de Meia-Idade , Músculo Esquelético/metabolismo , PPAR gama/metabolismo
15.
Blood ; 125(15): 2405-17, 2015 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-25724378

RESUMO

We describe a case of severe neonatal anemia with kernicterus caused by compound heterozygosity for null mutations in KLF1, each inherited from asymptomatic parents. One of the mutations is novel. This is the first described case of a KLF1-null human. The phenotype of severe nonspherocytic hemolytic anemia, jaundice, hepatosplenomegaly, and marked erythroblastosis is more severe than that present in congenital dyserythropoietic anemia type IV as a result of dominant mutations in the second zinc-finger of KLF1. There was a very high level of HbF expression into childhood (>70%), consistent with a key role for KLF1 in human hemoglobin switching. We performed RNA-seq on circulating erythroblasts and found that human KLF1 acts like mouse Klf1 to coordinate expression of many genes required to build a red cell including those encoding globins, cytoskeletal components, AHSP, heme synthesis enzymes, cell-cycle regulators, and blood group antigens. We identify novel KLF1 target genes including KIF23 and KIF11 which are required for proper cytokinesis. We also identify new roles for KLF1 in autophagy, global transcriptional control, and RNA splicing. We suggest loss of KLF1 should be considered in otherwise unexplained cases of severe neonatal NSHA or hydrops fetalis.


Assuntos
Anemia Neonatal/genética , Anemia Neonatal/patologia , Deleção de Genes , Hidropisia Fetal/genética , Hidropisia Fetal/patologia , Fatores de Transcrição Kruppel-Like/genética , Transcriptoma , Anemia Neonatal/sangue , Anemia Neonatal/complicações , Autofagia , Eritroblastos/metabolismo , Eritroblastos/patologia , Eritropoese , Feminino , Regulação da Expressão Gênica , Humanos , Hidropisia Fetal/sangue , Recém-Nascido , Fatores de Transcrição Kruppel-Like/metabolismo , Masculino
16.
Genomics ; 105(2): 116-22, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25451176

RESUMO

Position-effect variegation of transgene expression is sensitive to the chromatin state. We previously reported a forward genetic screen in mice carrying a variegated α-globin GFP transgene to find novel genes encoding epigenetic regulators. We named the phenovariant strains "Mommes" for modifiers of murine metastable epialleles. Here we report positional cloning of mutations in two Momme strains which result in suppression of variegation. Both strains harbour point mutations in the erythroid transcription factor, Klf1. One (D11) generates a stop codon in the zinc finger domain and a homozygous null phenotype. The other (D45) generates an amino acid transversion (H350R) within a conserved linker between zinc fingers two and three. Homozygous MommeD45 mice have chronic microcytic anaemia which models the phenotype in a recently described family. This is the first genetic evidence that the linkers between the zinc fingers of transcription factors have a function beyond that of a simple spacer.


Assuntos
Efeitos da Posição Cromossômica , Fatores de Transcrição Kruppel-Like/genética , Mutação , alfa-Globinas/genética , Anemia/genética , Animais , Testes Genéticos/métodos , Camundongos , Camundongos Transgênicos/embriologia , Camundongos Transgênicos/genética , Esplenomegalia/genética , Dedos de Zinco/genética
17.
BMC Genomics ; 15: 1002, 2014 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-25409780

RESUMO

BACKGROUND: CRISPR-Cas9 is a revolutionary genome editing technique that allows for efficient and directed alterations of the eukaryotic genome. This relatively new technology has already been used in a large number of 'loss of function' experiments in cultured cells. Despite its simplicity and efficiency, screening for mutated clones remains time-consuming, laborious and/or expensive. RESULTS: Here we report a high-throughput screening strategy that allows parallel screening of up to 96 clones, using next-generation sequencing. As a proof of principle, we used CRISPR-Cas9 to disrupt the coding sequence of the homeobox gene, Evx1 in mouse embryonic stem cells. We screened 67 CRISPR-Cas9 transfected clones simultaneously by next-generation sequencing on the Ion Torrent PGM. We were able to identify both homozygous and heterozygous Evx1 mutants, as well as mixed clones, which must be identified to maintain the integrity of subsequent experiments. CONCLUSIONS: Our CRISPR-Cas9 screening strategy could be widely applied to screen for CRISPR-Cas9 mutants in a variety of contexts including the generation of mutant cell lines for in vitro research, the generation of transgenic organisms and for assessing the veracity of CRISPR-Cas9 homology directed repair. This technique is cost and time-effective, provides information on clonal heterogeneity and is adaptable for use on various sequencing platforms.


Assuntos
Sistemas CRISPR-Cas/genética , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Mutação/genética , Animais , Células Clonais , Células-Tronco Embrionárias/metabolismo , Proteínas de Homeodomínio/metabolismo , Camundongos
18.
Clin Vaccine Immunol ; 19(12): 1984-7, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23081813
19.
Genome Res ; 22(12): 2385-98, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22835905

RESUMO

KLF1 (formerly known as EKLF) regulates the development of erythroid cells from bi-potent progenitor cells via the transcriptional activation of a diverse set of genes. Mice lacking Klf1 die in utero prior to E15 from severe anemia due to the inadequate expression of genes controlling hemoglobin production, cell membrane and cytoskeletal integrity, and the cell cycle. We have recently described the full repertoire of KLF1 binding sites in vivo by performing KLF1 ChIP-seq in primary erythroid tissue (E14.5 fetal liver). Here we describe the KLF1-dependent erythroid transcriptome by comparing mRNA-seq from Klf1(+/+) and Klf1(-/-) erythroid tissue. This has revealed novel target genes not previously obtainable by traditional microarray technology, and provided novel insights into the function of KLF1 as a transcriptional activator. We define a cis-regulatory module bound by KLF1, GATA1, TAL1, and EP300 that coordinates a core set of erythroid genes. We also describe a novel set of erythroid-specific promoters that drive high-level expression of otherwise ubiquitously expressed genes in erythroid cells. Our study has identified two novel lncRNAs that are dynamically expressed during erythroid differentiation, and discovered a role for KLF1 in directing apoptotic gene expression to drive the terminal stages of erythroid maturation.


Assuntos
Eritropoese/genética , Regulação da Expressão Gênica no Desenvolvimento , Fatores de Transcrição Kruppel-Like/genética , RNA Mensageiro/genética , Transcriptoma , Animais , Apoptose , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Western Blotting , Diferenciação Celular , Mapeamento Cromossômico , Proteína p300 Associada a E1A/genética , Proteína p300 Associada a E1A/metabolismo , Células Eritroides/citologia , Células Eritroides/metabolismo , Fator de Transcrição GATA1/genética , Fator de Transcrição GATA1/metabolismo , Perfilação da Expressão Gênica , Marcação In Situ das Extremidades Cortadas , Fatores de Transcrição Kruppel-Like/metabolismo , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , RNA Mensageiro/metabolismo , Análise de Sequência de RNA/métodos , Proteína 1 de Leucemia Linfocítica Aguda de Células T
20.
J Mol Med (Berl) ; 90(10): 1197-207, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22527883

RESUMO

Streptococcus pyogenes (group A Streptococcus (GAS)) causes ∼700 million human infections each year, resulting in over 500,000 deaths. The development of a commercial GAS vaccine is hampered by the occurrence of many unique GAS serotypes, antigenic variation within the same serotype, differences in serotype geographical distribution, and the production of antibodies cross-reactive with human tissue that may lead to autoimmune disease. Several independent studies have documented a number of GAS cell wall-associated or secreted metabolic enzymes that contain neither N-terminal leader sequences nor C-terminal cell wall anchors. Here, we applied a proteomic analysis of serotype M1T1 GAS cell wall extracts for the purpose of vaccine development. This approach catalogued several anchorless proteins and identified two protective vaccine candidates, arginine deiminase and trigger factor. These surface-exposed enzymes are expressed across multiple GAS serotypes exhibiting ≥99% amino acid sequence identity. Vaccine safety concerns are alleviated by the observation that these vaccine candidates lack human homologs, while sera from human populations suffering repeated GAS infections and high levels of autoimmune complications do not recognize these enzymes. Our study demonstrates anchorless cell surface antigens as promising vaccine candidates for the prevention of GAS disease.


Assuntos
Proteínas de Bactérias/metabolismo , Parede Celular/metabolismo , Hidrolases/metabolismo , Peptidilprolil Isomerase/metabolismo , Infecções Estreptocócicas/prevenção & controle , Vacinas Estreptocócicas/metabolismo , Streptococcus pyogenes/imunologia , Adolescente , Animais , Proteínas de Bactérias/imunologia , Parede Celular/imunologia , Criança , Feminino , Humanos , Hidrolases/imunologia , Soros Imunes/imunologia , Imunidade Ativa , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Microscopia Imunoeletrônica , Peptidilprolil Isomerase/imunologia , Proteoma/imunologia , Proteoma/metabolismo , Coelhos , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Infecções Estreptocócicas/imunologia , Vacinas Estreptocócicas/administração & dosagem , Vacinas Estreptocócicas/imunologia , Streptococcus pyogenes/enzimologia , Streptococcus pyogenes/ultraestrutura , Vacinação , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...