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











Base de dados
Intervalo de ano de publicação
1.
Cell Genom ; 2(4)2022 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-35967079

RESUMO

Repeat elements can be dysregulated at a genome-wide scale in human diseases. For example, in Ewing sarcoma, hundreds of inert GGAA repeats can be converted into active enhancers when bound by EWS-FLI1. Here we show that fusions between EWS and GGAA-repeat-targeted engineered zinc finger arrays (ZFAs) can function at least as efficiently as EWS-FLI1 for converting hundreds of GGAA repeats into active enhancers in a Ewing sarcoma precursor cell model. Furthermore, a fusion of a KRAB domain to a ZFA can silence GGAA microsatellite enhancers genome wide in Ewing sarcoma cells, thereby reducing expression of EWS-FLI1-activated genes. Remarkably, this KRAB-ZFA fusion showed selective toxicity against Ewing sarcoma cells compared with non-Ewing cancer cells, consistent with its Ewing sarcoma-specific impact on the transcriptome. These findings demonstrate the value of ZFAs for functional annotation of repeats and illustrate how aberrant microsatellite activities might be regulated for potential therapeutic applications.

2.
Nat Cancer ; 3(8): 961-975, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35982179

RESUMO

Rhabdomyosarcoma (RMS) is a common childhood cancer that shares features with developing skeletal muscle. Yet, the conservation of cellular hierarchy with human muscle development and the identification of molecularly defined tumor-propagating cells has not been reported. Using single-cell RNA-sequencing, DNA-barcode cell fate mapping and functional stem cell assays, we uncovered shared tumor cell hierarchies in RMS and human muscle development. We also identified common developmental stages at which tumor cells become arrested. Fusion-negative RMS cells resemble early myogenic cells found in embryonic and fetal development, while fusion-positive RMS cells express a highly specific gene program found in muscle cells transiting from embryonic to fetal development at 7-7.75 weeks of age. Fusion-positive RMS cells also have neural pathway-enriched states, suggesting less-rigid adherence to muscle-lineage hierarchies. Finally, we identified a molecularly defined tumor-propagating subpopulation in fusion-negative RMS that shares remarkable similarity to bi-potent, muscle mesenchyme progenitors that can make both muscle and osteogenic cells.


Assuntos
Rabdomiossarcoma Embrionário , Rabdomiossarcoma , Criança , Humanos , Músculo Esquelético/patologia , Rabdomiossarcoma/genética , Análise de Célula Única , Células-Tronco/patologia
3.
Blood ; 140(17): 1891-1906, 2022 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-35544598

RESUMO

Relapse and refractory T-cell acute lymphoblastic leukemia (T-ALL) has a poor prognosis, and new combination therapies are sorely needed. Here, we used an ex vivo high-throughput screening platform to identify drug combinations that kill zebrafish T-ALL and then validated top drug combinations for preclinical efficacy in human disease. This work uncovered potent drug synergies between AKT/mTORC1 (mammalian target of rapamycin complex 1) inhibitors and the general tyrosine kinase inhibitor dasatinib. Importantly, these same drug combinations effectively killed a subset of relapse and dexamethasone-resistant zebrafish T-ALL. Clinical trials are currently underway using the combination of mTORC1 inhibitor temsirolimus and dasatinib in other pediatric cancer indications, leading us to prioritize this therapy for preclinical testing. This combination effectively curbed T-ALL growth in human cell lines and primary human T-ALL and was well tolerated and effective in suppressing leukemia growth in patient-derived xenografts (PDX) grown in mice. Mechanistically, dasatinib inhibited phosphorylation and activation of the lymphocyte-specific protein tyrosine kinase (LCK) to blunt the T-cell receptor (TCR) signaling pathway, and when complexed with mTORC1 inhibition, induced potent T-ALL cell killing through reducing MCL-1 protein expression. In total, our work uncovered unexpected roles for the LCK kinase and its regulation of downstream TCR signaling in suppressing apoptosis and driving continued leukemia growth. Analysis of a wide array of primary human T-ALLs and PDXs grown in mice suggest that combination of temsirolimus and dasatinib treatment will be efficacious for a large fraction of human T-ALLs.


Assuntos
Proteína Tirosina Quinase p56(lck) Linfócito-Específica , Leucemia-Linfoma Linfoblástico de Células T Precursoras , Criança , Humanos , Camundongos , Animais , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Dasatinibe/farmacologia , Dasatinibe/uso terapêutico , Peixe-Zebra/metabolismo , Tirosina , Linhagem Celular Tumoral , Transdução de Sinais , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Receptores de Antígenos de Linfócitos T/uso terapêutico , Linfócitos T/metabolismo , Recidiva , Mamíferos/metabolismo
4.
Life Sci Alliance ; 4(2)2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33361335

RESUMO

Synovial sarcoma (SyS) is an aggressive mesenchymal malignancy invariably associated with the chromosomal translocation t(X:18; p11:q11), which results in the in-frame fusion of the BAF complex gene SS18 to one of three SSX genes. Fusion of SS18 to SSX generates an aberrant transcriptional regulator, which, in permissive cells, drives tumor development by initiating major chromatin remodeling events that disrupt the balance between BAF-mediated gene activation and polycomb-dependent repression. Here, we developed SyS organoids and performed genome-wide epigenomic profiling of these models and mesenchymal precursors to define SyS-specific chromatin remodeling mechanisms and dependencies. We show that SS18-SSX induces broad BAF domains at its binding sites, which oppose polycomb repressor complex (PRC) 2 activity, while facilitating recruitment of a non-canonical (nc)PRC1 variant. Along with the uncoupling of polycomb complexes, we observed H3K27me3 eviction, H2AK119ub deposition and the establishment of de novo active regulatory elements that drive SyS identity. These alterations are completely reversible upon SS18-SSX depletion and are associated with vulnerability to USP7 loss, a core member of ncPRC1.1. Using the power of primary tumor organoids, our work helps define the mechanisms of epigenetic dysregulation on which SyS cells are dependent.


Assuntos
Montagem e Desmontagem da Cromatina , Cromatina/genética , Epigênese Genética , Regulação Neoplásica da Expressão Gênica , Sarcoma Sinovial/genética , Sítios de Ligação , Cromatina/metabolismo , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Perfilação da Expressão Gênica , Histonas/metabolismo , Humanos , Complexos Multiproteicos/metabolismo , Organoides , Ligação Proteica , Transporte Proteico , Sarcoma Sinovial/metabolismo , Transcriptoma
5.
Oncotarget ; 11(12): 1051-1074, 2020 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-32256978

RESUMO

Polycomb repressive complex 2 (PRC2) allows the deposition of H3K27me3. PRC2 facultative subunits modulate its activity and recruitment such as hPCL3/PHF19, a human ortholog of Drosophila Polycomb-like protein (PCL). These proteins contain a TUDOR domain binding H3K36me3, two PHD domains and a "Winged-helix" domain involved in GC-rich DNA binding. The human PCL3 locus encodes the full-length hPCL3L protein and a shorter isoform, hPCL3S containing the TUDOR and PHD1 domains only. In this study, we demonstrated by RT-qPCR analyses of 25 prostate tumors that hPCL3S is frequently up-regulated. In addition, hPCL3S is overexpressed in the androgen-independent DU145 and PC3 cells, but not in the androgen-dependent LNCaP cells. hPCL3S knockdown decreased the proliferation and migration of DU145 and PC3 whereas its forced expression into LNCaP increased these properties. A mutant hPCL3S unable to bind H3K36me3 (TUDOR-W50A) increased proliferation and migration of LNCaP similarly to wt hPCL3S whereas inactivation of its PHD1 domain decreased proliferation. These effects partially relied on the up-regulation of genes known to be important for the proliferation and/or migration of prostate cancer cells such as S100A16, PlexinA2, and Spondin1. Collectively, our results suggest hPCL3S as a new potential therapeutic target in castration resistant prostate cancers.

6.
Cell Death Dis ; 11(1): 75, 2020 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-32001672

RESUMO

The bivalent domain (BD) at promoter region is an unique epigenetic feature poised for activation or repression during cell differentiation in embryonic stem cell. However, the function of BDs in already differentiated cells remains exclusive. By profiling the epigenetic landscape of endothelial cells during VEGFA (vascular endothelial growth factor A) stimulation, we discovered that BDs are widespread in endothelial cells and preferentially marked genes responsive to VEGFA. The BDs responsive to VEGFA have more permissive chromatin environment comparing to other BDs. The initial activation of bivalent genes depends on RNAPII pausing release induced by EZH1 rather than removal of H3K27me3. The later suppression of bivalent gene expression depended on KDM5A recruitment by its interaction with PRC2. Importantly, EZH1 promoted both in vitro and in vivo angiogenesis by upregulating EGR3, whereas KDM5A dampened angiogenesis. Collectively, this study demonstrates a novel dual function of BDs in endothelial cells to control VEGF responsiveness and angiogenesis.


Assuntos
Células Endoteliais/metabolismo , Histonas/metabolismo , Neovascularização Fisiológica/genética , Regiões Promotoras Genéticas/genética , Fator A de Crescimento do Endotélio Vascular/farmacologia , Animais , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Cromatina/metabolismo , Sequenciamento de Cromatina por Imunoprecipitação , Proteína 3 de Resposta de Crescimento Precoce/metabolismo , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Células Endoteliais/efeitos dos fármacos , Epigênese Genética , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos , Complexo Repressor Polycomb 2/metabolismo , Domínios Proteicos/genética , RNA Polimerase II/metabolismo , RNA Interferente Pequeno , RNA-Seq , Proteína 2 de Ligação ao Retinoblastoma/metabolismo , Regulação para Cima
7.
Nat Commun ; 10(1): 4439, 2019 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-31570731

RESUMO

Adeno-associated virus (AAV) vectors have shown promising results in preclinical models, but the genomic consequences of transduction with AAV vectors encoding CRISPR-Cas nucleases is still being examined. In this study, we observe high levels of AAV integration (up to 47%) into Cas9-induced double-strand breaks (DSBs) in therapeutically relevant genes in cultured murine neurons, mouse brain, muscle and cochlea. Genome-wide AAV mapping in mouse brain shows no overall increase of AAV integration except at the CRISPR/Cas9 target site. To allow detailed characterization of integration events we engineer a miniature AAV encoding a 465 bp lambda bacteriophage DNA (AAV-λ465), enabling sequencing of the entire integrated vector genome. The integration profile of AAV-465λ in cultured cells display both full-length and fragmented AAV genomes at Cas9 on-target sites. Our data indicate that AAV integration should be recognized as a common outcome for applications that utilize AAV for genome editing.


Assuntos
Sistemas CRISPR-Cas , Quebras de DNA , Dependovirus/genética , Edição de Genes/métodos , Vetores Genéticos , Integração Viral/genética , Animais , Bacteriófago lambda/genética , Encéfalo , Linhagem Celular , Mapeamento Cromossômico , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Cóclea , Endonucleases , Marcação de Genes/métodos , Terapia Genética/métodos , Genoma , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Músculos , Neurônios/virologia , Reparo Gênico Alvo-Dirigido/métodos , Resultado do Tratamento
8.
Nat Med ; 25(7): 1123-1130, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31270503

RESUMO

Since most dominant human mutations are single nucleotide substitutions1,2, we explored gene editing strategies to disrupt dominant mutations efficiently and selectively without affecting wild-type alleles. However, single nucleotide discrimination can be difficult to achieve3 because commonly used endonucleases, such as Streptococcus pyogenes Cas9 (SpCas9), can tolerate up to seven mismatches between guide RNA (gRNA) and target DNA. Furthermore, the protospacer-adjacent motif (PAM) in some Cas9 enzymes can tolerate mismatches with the target DNA3,4. To circumvent these limitations, we screened 14 Cas9/gRNA combinations for specific and efficient disruption of a nucleotide substitution that causes the dominant progressive hearing loss, DFNA36. As a model for DFNA36, we used Beethoven mice5, which harbor a point mutation in Tmc1, a gene required for hearing that encodes a pore-forming subunit of mechanosensory transduction channels in inner-ear hair cells6. We identified a PAM variant of Staphylococcus aureus Cas9 (SaCas9-KKH) that selectively and efficiently disrupted the mutant allele, but not the wild-type Tmc1/TMC1 allele, in Beethoven mice and in a DFNA36 human cell line. Adeno-associated virus (AAV)-mediated SaCas9-KKH delivery prevented deafness in Beethoven mice up to one year post injection. Analysis of current ClinVar entries revealed that ~21% of dominant human mutations could be targeted using a similar approach.


Assuntos
Alelos , Edição de Genes , Perda Auditiva Neurossensorial/prevenção & controle , Proteínas de Membrana/genética , Animais , Proteína 9 Associada à CRISPR/fisiologia , Linhagem Celular , Células Cultivadas , Dependovirus/genética , Modelos Animais de Doenças , Perda Auditiva Neurossensorial/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL
9.
Nature ; 569(7756): 433-437, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30995674

RESUMO

CRISPR-Cas base-editor technology enables targeted nucleotide alterations, and is being increasingly used for research and potential therapeutic applications1,2. The most widely used cytosine base editors (CBEs) induce deamination of DNA cytosines using the rat APOBEC1 enzyme, which is targeted by a linked Cas protein-guide RNA complex3,4. Previous studies of the specificity of CBEs have identified off-target DNA edits in mammalian cells5,6. Here we show that a CBE with rat APOBEC1 can cause extensive transcriptome-wide deamination of RNA cytosines in human cells, inducing tens of thousands of C-to-U edits with frequencies ranging from 0.07% to 100% in 38-58% of expressed genes. CBE-induced RNA edits occur in both protein-coding and non-protein-coding sequences and generate missense, nonsense, splice site, and 5' and 3' untranslated region mutations. We engineered two CBE variants bearing mutations in rat APOBEC1 that substantially decreased the number of RNA edits (by more than 390-fold and more than 3,800-fold) in human cells. These variants also showed more precise on-target DNA editing than the wild-type CBE and, for most guide RNAs tested, no substantial reduction in editing efficiency. Finally, we show that an adenine base editor7 can also induce transcriptome-wide RNA edits. These results have implications for the use of base editors in both research and clinical settings, illustrate the feasibility of engineering improved variants with reduced RNA editing activities, and suggest the need to more fully define and characterize the RNA off-target effects of deaminase enzymes in base editor platforms.


Assuntos
Sistemas CRISPR-Cas/genética , Edição de Genes , Edição de RNA , Especificidade por Substrato/genética , Transcriptoma/genética , Desaminase APOBEC-1/química , Desaminase APOBEC-1/genética , Desaminase APOBEC-1/metabolismo , Animais , Sequência de Bases , Citosina/metabolismo , Desaminação , Células HEK293 , Células Hep G2 , Humanos , Mutação , RNA/química , RNA/metabolismo , Ratos
10.
Genome Res ; 29(2): 193-207, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30670628

RESUMO

Cell behaviors are dictated by epigenetic and transcriptional programs. Little is known about how extracellular stimuli modulate these programs to reshape gene expression and control cell behavioral responses. Here, we interrogated the epigenetic and transcriptional response of endothelial cells to VEGFA treatment and found rapid chromatin changes that mediate broad transcriptomic alterations. VEGFA-responsive genes were associated with active promoters, but changes in promoter histone marks were not tightly linked to gene expression changes. VEGFA altered transcription factor occupancy and the distal epigenetic landscape, which profoundly contributed to VEGFA-dependent changes in gene expression. Integration of gene expression, dynamic enhancer, and transcription factor occupancy changes induced by VEGFA yielded a VEGFA-regulated transcriptional regulatory network, which revealed that the small MAF transcription factors are master regulators of the VEGFA transcriptional program and angiogenesis. Collectively these results revealed that extracellular stimuli rapidly reconfigure the chromatin landscape to coordinately regulate biological responses.


Assuntos
Epigênese Genética , Neovascularização Fisiológica/genética , Transcrição Gênica , Fator A de Crescimento do Endotélio Vascular/fisiologia , Animais , Células Cultivadas , Cromatina/metabolismo , Elementos Facilitadores Genéticos , Humanos , Fatores de Transcrição Maf/metabolismo , Masculino , Camundongos , Camundongos Nus , Regiões Promotoras Genéticas , Fatores de Transcrição/metabolismo
12.
Elife ; 72018 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-30192230

RESUMO

The TP53 tumor-suppressor gene is mutated in >50% of human tumors and Li-Fraumeni patients with germ line inactivation are predisposed to developing cancer. Here, we generated tp53 deleted zebrafish that spontaneously develop malignant peripheral nerve-sheath tumors, angiosarcomas, germ cell tumors, and an aggressive Natural Killer cell-like leukemia for which no animal model has been developed. Because the tp53 deletion was generated in syngeneic zebrafish, engraftment of fluorescent-labeled tumors could be dynamically visualized over time. Importantly, engrafted tumors shared gene expression signatures with predicted cells of origin in human tissue. Finally, we showed that tp53del/del enhanced invasion and metastasis in kRASG12D-induced embryonal rhabdomyosarcoma (ERMS), but did not alter the overall frequency of cancer stem cells, suggesting novel pro-metastatic roles for TP53 loss-of-function in human muscle tumors. In summary, we have developed a Li-Fraumeni zebrafish model that is amenable to large-scale transplantation and direct visualization of tumor growth in live animals.


Assuntos
Rabdomiossarcoma Embrionário/metabolismo , Rabdomiossarcoma Embrionário/patologia , Proteína Supressora de Tumor p53/deficiência , Peixe-Zebra/metabolismo , Animais , Contagem de Células , Deleção de Genes , Regulação Neoplásica da Expressão Gênica , Hemangiossarcoma/metabolismo , Hemangiossarcoma/patologia , Homozigoto , Leucemia/metabolismo , Leucemia/patologia , Invasividade Neoplásica , Metástase Neoplásica , Transplante de Neoplasias , Neoplasias Embrionárias de Células Germinativas/metabolismo , Neoplasias Embrionárias de Células Germinativas/patologia , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Rabdomiossarcoma Embrionário/genética , Análise de Sobrevida , Proteína Supressora de Tumor p53/metabolismo , Peixe-Zebra/genética
14.
Cell Stem Cell ; 22(3): 414-427.e6, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29499154

RESUMO

Tumor growth and relapse are driven by tumor propagating cells (TPCs). However, mechanisms regulating TPC fate choices, maintenance, and self-renewal are not fully understood. Here, we show that Van Gogh-like 2 (Vangl2), a core regulator of the non-canonical Wnt/planar cell polarity (Wnt/PCP) pathway, affects TPC self-renewal in rhabdomyosarcoma (RMS)-a pediatric cancer of muscle. VANGL2 is expressed in a majority of human RMS and within early mononuclear progenitor cells. VANGL2 depletion inhibited cell proliferation, reduced TPC numbers, and induced differentiation of human RMS in vitro and in mouse xenografts. Using a zebrafish model of embryonal rhabdomyosarcoma (ERMS), we determined that Vangl2 expression enriches for TPCs and promotes their self-renewal. Expression of constitutively active and dominant-negative isoforms of RHOA revealed that it acts downstream of VANGL2 to regulate proliferation and maintenance of TPCs in human RMS. Our studies offer insights into pathways that control TPCs and identify new potential therapeutic targets.


Assuntos
Autorrenovação Celular , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Células-Tronco Neoplásicas/patologia , Rabdomiossarcoma/patologia , Transdução de Sinais , Proteínas de Peixe-Zebra/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Animais , Diferenciação Celular , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Camundongos , Células-Tronco Neoplásicas/metabolismo , Rabdomiossarcoma/genética , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Science ; 345(6204): 1251033, 2014 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-25258084

RESUMO

Blood cells derive from hematopoietic stem cells through stepwise fating events. To characterize gene expression programs driving lineage choice, we sequenced RNA from eight primary human hematopoietic progenitor populations representing the major myeloid commitment stages and the main lymphoid stage. We identified extensive cell type-specific expression changes: 6711 genes and 10,724 transcripts, enriched in non-protein-coding elements at early stages of differentiation. In addition, we found 7881 novel splice junctions and 2301 differentially used alternative splicing events, enriched in genes involved in regulatory processes. We demonstrated experimentally cell-specific isoform usage, identifying nuclear factor I/B (NFIB) as a regulator of megakaryocyte maturation-the platelet precursor. Our data highlight the complexity of fating events in closely related progenitor populations, the understanding of which is essential for the advancement of transplantation and regenerative medicine.


Assuntos
Processamento Alternativo , Linhagem da Célula/genética , Hematopoese/genética , Células-Tronco Hematopoéticas/citologia , Variação Genética , Células-Tronco Hematopoéticas/metabolismo , Humanos , Fatores de Transcrição NFI/genética , Fatores de Transcrição NFI/metabolismo , Proteínas de Ligação a RNA/metabolismo , Trombopoese/genética , Transcriptoma
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA