Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Bioorg Med Chem Lett ; 40: 127925, 2021 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-33705909

RESUMO

Our research group has been studying the design of intracellular delivery peptides based on cationic lytic peptides. By placing negatively charged amino acids on potentially hydrophobic faces of the peptides, membrane lytic activity is attenuated on the cell surface, whereas it recovers in endosomes, enabling cytosolic delivery of proteins including antibodies. These lytic peptides generally contain multiple lysines, facilitating cell surface interaction and membrane perturbation. This study evaluated the effect of lysine-to-homoarginine substitution using HAad as a model delivery peptide. The resulting peptide had a comparable or better delivery efficacy for Cre recombinase, antibodies, and the Cas9/sgRNA complex with one-quarter of the concentration of HAad, implying that a subtle structural difference can affect delivery activity.


Assuntos
Portadores de Fármacos/química , Endossomos/metabolismo , Homoarginina/química , Membranas Intracelulares/metabolismo , Peptídeos/química , Sequência de Aminoácidos , Proteína 9 Associada à CRISPR/farmacologia , Dextranos/química , Portadores de Fármacos/toxicidade , Liberação Controlada de Fármacos , Fluoresceínas/química , Corantes Fluorescentes/química , Células HeLa , Humanos , Imunoglobulina G/farmacologia , Integrases/farmacologia , Lipossomos/química , Peptídeos/toxicidade , RNA Guia de Cinetoplastídeos/farmacologia , Ácidos Sulfônicos/química
2.
Cancer Res ; 81(5): 1332-1346, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33372040

RESUMO

Although the cyclin-dependent kinases CDK4 and CDK6 play fundamental roles in cancer, the specific pathways and downstream targets by which they exert their tumorigenic effects remain elusive. In this study, we uncover distinct and novel functions for these kinases in regulating tumor formation and metastatic colonization in various solid tumors, including those of the breast, prostate, and pancreas. Combining in vivo CRISPR-based CDK4 and CDK6 gene editing with pharmacologic inhibition approaches in orthotopic transplantation and patient-derived xenograft preclinical models, we defined clear functions for CDK4 and CDK6 in facilitating tumor growth and progression in metastatic cancers. Transcriptomic profiling of CDK4/6 CRISPR knockouts in breast cancer revealed these two kinases to regulate cancer progression through distinct mechanisms. CDK4 regulated prometastatic inflammatory cytokine signaling, whereas CDK6 mainly controlled DNA replication and repair processes. Inhibition of CDK6 but not CDK4 resulted in defective DNA repair and increased DNA damage. Multiple CDK6 DNA replication/repair genes were not only associated with cancer subtype, grades, and poor clinical outcomes, but also facilitated primary tumor growth and metastasis in vivo. CRISPR-based genomic deletion of CDK6 efficiently blocked tumor formation and progression in preestablished cell- and patient-derived xenograft preclinical models of breast cancer, providing a potential novel targeted therapy for these deadly tumors. SIGNIFICANCE: In-depth transcriptomic analysis identifies cyclin-dependent kinases CDK4 and CDK6 as regulators of metastasis through distinct signaling pathways and reveals the DNA replication/repair pathway as central in promoting these effects.


Assuntos
Quinase 4 Dependente de Ciclina/genética , Quinase 6 Dependente de Ciclina/genética , Reparo do DNA/fisiologia , Replicação do DNA/fisiologia , Neoplasias/patologia , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Quinase 4 Dependente de Ciclina/metabolismo , Quinase 6 Dependente de Ciclina/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Técnicas de Inativação de Genes , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/secundário , Masculino , Camundongos SCID , Neoplasias/genética , RNA Guia de Cinetoplastídeos/administração & dosagem , RNA Guia de Cinetoplastídeos/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
3.
CRISPR J ; 3(6): 512-522, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33346704

RESUMO

Allele-specific genomic targeting by CRISPR is a versatile strategy that has been increasingly exploited not only in treating inherited dominant diseases and mutation-driven cancers, but also in other important fields such as genome imprinting, haploinsufficiency, and genome loci imaging. Despite its tremendous utilities, few bioinformatic tools have been implemented for the allele-specific purpose of CRISPR. We thus developed AsCRISPR (Allele-specific CRISPR), a comprehensive web tool to aid the design of short-guide RNA (sgRNA) sequences that can discriminate between alleles. AsCRISPR allows users to analyze both their own identified variants and heterozygous single nucleotide polymorphisms and, importantly, output the candidate sgRNAs and their quality control information. To facilitate targeting dominant diseases, AsCRISPR analyzed dominant single nucleotide variants (SNVs) retrieved from ClinVar and OMIM databases, and generated a dominant database of candidate-discriminating sgRNAs that may specifically target the alternative allele for each dominant SNV site. Moreover, a validated database was established, which manually curated the discriminating sgRNAs that were experimentally validated in the mounting literature for multiple allele-specific purposes.


Assuntos
Edição de Genes/métodos , Medicina de Precisão/métodos , RNA Guia de Cinetoplastídeos/genética , Algoritmos , Alelos , Sistemas CRISPR-Cas/genética , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Biologia Computacional/métodos , Computadores , Genômica/métodos , Humanos , Polimorfismo de Nucleotídeo Único/genética , RNA Guia de Cinetoplastídeos/farmacologia , Software
4.
Leuk Res ; 99: 106464, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33130330

RESUMO

The role of interferon-gamma (IFN-γ) in Chronic Myelogenous/Myeloid Leukemia (CML) and in the treatment of CML remains unclear; specifically, the effect of IFN-γ on apoptosis. There is reported interplay between IFN-γ and glycogen synthase kinase-3 (GSK-3), a kinase which has been implicated in both cell death and, conversely, cell survival. Thus, we utilized the CML-derived HAP1 cell line and a mutant HAP1 GSK-3ß knocked-down cell line (GSK-3ß 31bp) to investigate whether GSK-3 modulates IFN-γ's action on CML cells. Significantly less GSK-3ß 31bp cells, relative to HAP1 cells, were present after 48 h treatment with IFN-γ. IFN-γ treatment significantly decreased GSK-3ß 31bp substrate adhesiveness (relative to HAP1 cells); an observation often correlated with cell death. Fluorescence microscopy revealed that IFN-γ induces a modest level of apoptosis in the HAP1 cells and that IFN-γ induced apoptosis is significantly enhanced in GSK-3ß 31bp cells. Utilizing a complementary GSK-3ß knocked-down cell line (8bp) we found, via flow cytometric analysis, that IFN-γ induced apoptosis is significantly enhanced in GSK-3ß 8bp cells relative to HAP1 cells. Combined, our findings suggest that IFN-γ induces apoptosis of CML cells and that loss of GSK-3ß significantly augments IFN-γ-induced apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Glicogênio Sintase Quinase 3 beta/antagonistas & inibidores , Interferon gama/farmacologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Proteínas de Neoplasias/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Antineoplásicos/farmacologia , Sistemas CRISPR-Cas , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral/efeitos dos fármacos , Códon sem Sentido , Interações Medicamentosas , Citometria de Fluxo , Mutação da Fase de Leitura , Proteínas de Fusão bcr-abl/antagonistas & inibidores , Técnicas de Silenciamento de Genes , Glicogênio Sintase Quinase 3 beta/genética , Humanos , Interferon gama/uso terapêutico , Leucemia Mielogênica Crônica BCR-ABL Positiva/enzimologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Masculino , RNA Guia de Cinetoplastídeos/genética , RNA Guia de Cinetoplastídeos/farmacologia , Espectrometria de Fluorescência
5.
Mol Biol Rep ; 47(8): 6091-6103, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32761301

RESUMO

Cervical cancer is the leading cause of cancer-related death among women worldwide. Identifying an effective treatment with fewer side effects is imperative, because all of the current treatments have unique disadvantages. Aldo-keto reductase family 1 member B1 (AKR1B1) is highly expressed in various cancers and is associated with tumor development, but has not been studied in cervical cancer. In the current study, we used CRISPR/Cas9 technology to establish a stable HeLa cell line with AKR1B1 knockout. In vitro, AKR1B1 knockout inhibited the proliferation, migration and invasion of HeLa cells, providing evidence that AKR1B1 is an innovative therapeutic target. Notably, the clinically used epalrestat, an inhibitor of aldose reductases, including AKR1B1, had the same effect as AKR1B1 knockout on HeLa cells. This result suggests that epalrestat could be used in the clinical treatment of cervical cancer, a prospect that undoubtedly requires further research. Moreover, aiming to determine the underlying regulatory mechanism of AKR1B1, we screened a series of differentially regulated genes (DEGs) by RNA sequencing and verified selected DEGs by quantitative RT-PCR. In addition, gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses of the DEGs revealed a correlation between AKR1B1 and cancer. In summary, epalrestat inhibits the progression of cervical cancer by inhibiting AKR1B1, and thus may be a new drug for the clinical treatment of cervical cancer.


Assuntos
Aldeído Redutase/fisiologia , Inibidores Enzimáticos/farmacologia , Proteínas de Neoplasias/fisiologia , Rodanina/análogos & derivados , Tiazolidinas/farmacologia , Neoplasias do Colo do Útero/tratamento farmacológico , Aldeído Redutase/antagonistas & inibidores , Aldeído Redutase/genética , Divisão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Ontologia Genética , Células HeLa , Humanos , Invasividade Neoplásica , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , RNA Guia de Cinetoplastídeos/genética , RNA Guia de Cinetoplastídeos/farmacologia , Rodanina/farmacologia , Ensaio Tumoral de Célula-Tronco , Neoplasias do Colo do Útero/patologia
6.
Sci Rep ; 10(1): 10919, 2020 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-32616820

RESUMO

Marek's disease virus (MDV) is a highly cell-associated alphaherpesvirus that causes deadly lymphomas in chickens. While vaccination protects against clinical symptoms, MDV field strains can still circulate in vaccinated flocks and continuously evolve towards greater virulence. MDV vaccines do not provide sterilizing immunity, allowing the virus to overcome vaccine protection, and has increased the need for more potent vaccines or alternative interventions. In this study, we addressed if the CRISPR/Cas9 system can protect cells from MDV replication. We first screened a number of guide RNAs (gRNAs) targeting essential MDV genes for their ability to prevent virus replication. Single gRNAs significantly inhibited virus replication, but could result in the emergence of escape mutants. Strikingly, combining two or more gRNAs completely abrogated virus replication and no escape mutants were observed upon serial passaging. Our study provides the first proof-of-concept, demonstrating that the CRISPR/Cas9 system can be efficiently used to block MDV replication. The presented findings lay the foundation for future research to completely protect chickens from this deadly pathogen.


Assuntos
Sistemas CRISPR-Cas , Mardivirus/efeitos dos fármacos , RNA Guia de Cinetoplastídeos/farmacologia , Replicação Viral/efeitos dos fármacos , Animais , Embrião de Galinha , Galinhas , Patos , Genes Virais , Células HEK293 , Humanos , Mardivirus/genética , Mardivirus/fisiologia , Doença de Marek/prevenção & controle , Vacinas contra Doença de Marek , Mutação , Estudo de Prova de Conceito , RNA Guia de Cinetoplastídeos/genética , Organismos Livres de Patógenos Específicos , Replicação Viral/genética
7.
Nat Plants ; 6(6): 620-624, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32483329

RESUMO

An in planta gene editing approach was developed wherein Cas9 transgenic plants are infected with an RNA virus that expresses single guide RNAs (sgRNAs). The sgRNAs are augmented with sequences that promote cell-to-cell mobility. Mutant progeny are recovered in the next generation at frequencies ranging from 65 to 100%; up to 30% of progeny derived from plants infected with a virus expressing three sgRNAs have mutations in all three targeted loci.


Assuntos
Edição de Genes/métodos , Nicotiana/genética , Plantas Geneticamente Modificadas/genética , Vírus de RNA/genética , RNA Guia de Cinetoplastídeos/farmacologia , RNA Viral/farmacologia , Agrobacterium tumefaciens
8.
Sci Rep ; 10(1): 766, 2020 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-31964975

RESUMO

The proteasome inhibitor bortezomib is the most successfully applied chemotherapeutic drug for treating multiple myeloma. However, its clinical efficacy reduced due to resistance development. The underlying molecular mechanisms of bortezomib resistance are poorly understood. In this study, by combining in silico analysis and sgRNA library based drug resistance screening assay, we identified SENP2 (Sentrin/SUMO-specific proteases-2) as a bortezomib sensitive gene and found its expression highly downregulated in bortezomib resistant multiple myeloma patient's samples. Furthermore, down regulation of SENP2 in multiple myeloma cell line RPMI8226 alleviated bortezomib induced cell proliferation inhibition and apoptosis, whereas, overexpression of SENP2 sensitized these cells to bortezomib treatment. We further demonstrate that knockdown of SENP2 in RPMI8226 cells increased SUMO2 conjugated IκBα that resulted in the activation of NF-κB. Taken together, we report that silencing of SENP2 and consequent activation of NF-κB through the modulation of IκBα sumoylation as a novel mechanism inducing bortezomib resistance in multiple myeloma.


Assuntos
Bortezomib/farmacologia , Cisteína Endopeptidases/genética , Regulação para Baixo , Resistencia a Medicamentos Antineoplásicos , Mieloma Múltiplo/genética , Inibidor de NF-kappaB alfa/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Simulação por Computador , Regulação Neoplásica da Expressão Gênica , Humanos , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/metabolismo , NF-kappa B/metabolismo , RNA Guia de Cinetoplastídeos/farmacologia , Transdução de Sinais , Sumoilação
9.
Methods Mol Biol ; 2111: 59-70, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31933198

RESUMO

T-cell-based cancer immunotherapies have emerged as a promising approach for cancer treatment, highlighting the importance of understanding the regulation of T-cell function. However, the molecular mechanisms underlying T-cell activation are not fully understood. The CRISPR/Cas9 system can serve as a robust method to systematically study signaling pathways. In this chapter, we describe details of using the CRISPR screen to identify regulators in TCR signaling, from the sgRNA library construction to genomic DNA sequencing. We also add some notes to further help readers performing the CRISPR screen. This approach can be readily adapted to study the activation of other immune cells, including B cells and dendritic cells.


Assuntos
Redes Reguladoras de Genes , RNA Guia de Cinetoplastídeos/farmacologia , Análise de Sequência de DNA/métodos , Linfócitos T/imunologia , Sistemas CRISPR-Cas , Edição de Genes , Testes Genéticos , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Células Jurkat , Lentivirus/genética , Ativação Linfocitária , Transdução de Sinais , Transdução Genética
10.
Mol Syst Biol ; 15(12): e8983, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31885201

RESUMO

Arrayed CRISPR-based screens emerge as a powerful alternative to pooled screens making it possible to investigate a wide range of cellular phenotypes that are typically not amenable to pooled screens. Here, we describe a solid-phase transfection platform that enables CRISPR-based genetic screens in arrayed format with flexible readouts. We demonstrate efficient gene knockout upon delivery of guide RNAs and Cas9/guide RNA ribonucleoprotein complexes into untransformed and cancer cell lines. In addition, we provide evidence that our platform can be easily adapted to high-throughput screens and we use this approach to study oncogene addiction in tumor cells. Finally demonstrating that the human primary cells can also be edited using this method, we pave the way for rapid testing of potential targeted therapies.


Assuntos
Edição de Genes/instrumentação , Neoplasias/genética , RNA Guia de Cinetoplastídeos/farmacologia , Sistemas CRISPR-Cas , Linhagem Celular Tumoral , Predisposição Genética para Doença , Ensaios de Triagem em Larga Escala , Humanos , Fenótipo , Transfecção
11.
PLoS One ; 14(10): e0224113, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31658298

RESUMO

Inactive fusion variants of the CRISPR-Cas9 system are increasingly being used as standard methodology to study transcription regulation. Their ability to readily manipulate the native genomic loci is particularly advantageous. In this work, we serendipitously uncover the key cytokine IL6 as an off-target of the activating derivative of CRISPR (CRISPRa) while studying RP11-326A19.4, a novel long-non coding RNA (lncRNA). Increasing RP11-326A19.4 expression in HEK293T cells via CRISPRa-mediated activation of its promoter region induced genome-wide transcriptional changes, including upregulation of IL6, an important cytokine. IL6 was increased in response to distinct sgRNA targeting the RP11-326A19.4 promoter region, suggesting specificity. Loss of the cognate sgRNA recognition sites failed to abolish CRISPRa mediated activation of IL6 however, pointing to off-target effects. Bioinformatic approaches did not reveal predicted off-target binding sites. Off-target activation of IL6 was sustained and involved low level activation of known IL6 regulators. Increased IL6 remained sensitive to further activation by TNFα, consistent with the existence of independent mechanisms. This study provides experimental evidence that CRISPRa has discrete, unpredictable off-targeting limitations that must be considered when using this emerging technology.


Assuntos
Perfilação da Expressão Gênica/métodos , Interleucina-6/genética , RNA Guia de Cinetoplastídeos/farmacologia , RNA Longo não Codificante/genética , Sítios de Ligação , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Regulação da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Células HeLa , Humanos , Regiões Promotoras Genéticas , Ativação Transcricional , Regulação para Cima
12.
Mol Ther ; 27(10): 1737-1748, 2019 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-31383454

RESUMO

Cystic fibrosis (CF) is caused by mutations in the CF transmembrane conductance regulator (CFTR) gene. The majority of CFTR mutations result in impaired chloride channel function as only a fraction of the mutated CFTR reaches the plasma membrane. The development of a therapeutic approach that facilitates increased cell-surface expression of CFTR could prove clinically relevant. Here, we evaluate and contrast two molecular approaches to activate CFTR expression. We find that an RNA-guided nuclease null Cas9 (dCas9) fused with a tripartite activator, VP64-p65-Rta can activate endogenous CFTR in cultured human nasal epithelial cells from CF patients. We also find that targeting BGas, a long non-coding RNA involved in transcriptionally modulating CFTR expression with a gapmer, induced both strong knockdown of BGas and concordant activation of CFTR. Notably, the gapmer can be delivered to target cells when generated as electrostatic particles with recombinant HIV-Tat cell penetrating peptide (CPP), when packaged into exosomes, or when loaded into lipid nanoparticles (LNPs). Treatment of patient-derived human nasal epithelial cells containing F508del with gapmer-CPP, gapmer-exosomes, or LNPs resulted in increased expression and function of CFTR. Collectively, these observations suggest that CRISPR/dCas-VPR (CRISPR) and BGas-gapmer approaches can target and specifically activate CFTR.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Fibrose Cística/genética , Terapia de Alvo Molecular/métodos , Mucosa Nasal/metabolismo , Proteína 9 Associada à CRISPR/metabolismo , Linhagem Celular , Membrana Celular/metabolismo , Peptídeos Penetradores de Células/genética , Fibrose Cística/metabolismo , Fibrose Cística/terapia , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Humanos , Nanopartículas/química , Mucosa Nasal/citologia , RNA Guia de Cinetoplastídeos/farmacologia , RNA Longo não Codificante/genética , Ativação Transcricional , Produtos do Gene tat do Vírus da Imunodeficiência Humana/genética
13.
Acc Chem Res ; 52(6): 1555-1564, 2019 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-31099553

RESUMO

The discovery of CRISPR/Cas has revolutionized the field of genome editing. CRIPSR/Cas components are part of the bacterial immune system and are able to induce double-strand DNA breaks in the genome, which are resolved by endogenous DNA repair mechanisms. The most relevant of these are the error-prone nonhomologous end joining and homology directed repair pathways. The former can lead to gene knockout by introduction of insertions and deletions at the cut site, while the latter can be used for gene correction based on a provided repair template. In this Account, we focus on the delivery aspects of CRISPR/Cas for therapeutic applications in vivo. Safe and effective delivery of the CRISPR/Cas components into the nucleus of affected cells is essential for therapeutic gene editing. These components can be delivered in several formats, such as pDNA, viral vectors, or ribonuclear complexes. In the ideal case, the delivery system should address the current limitations of CRISPR gene editing, which are (1) lack of targeting specific tissues or cells, (2) the inability to enter cells, (3) activation of the immune system, and (4) off-target events. To circumvent most of these problems, initial therapeutic applications of CRISPR/Cas were performed on cells ex vivo via classical methods (e.g., microinjection or electroporation) and novel methods (e.g., TRIAMF and iTOP). Ideal candidates for such methods are, for example, hematopoietic cells, but not all tissue types are suited for ex vivo manipulation. For direct in vivo application, however, delivery systems are needed that can target the CRISPR/Cas components to specific tissues or cells in the human body, without causing immune activation or causing high frequencies of off-target effects. Viral systems have been used as a first resort to transduce cells in vivo. These systems suffer from problems related to packaging constraints, immunogenicity, and longevity of Cas expression, which favors off-target events. Viral vectors are as such not the best choice for direct in vivo delivery of CRISPR/Cas. Synthetic vectors can deliver nucleic acids as well, without the innate disadvantages of viral vectors. They can be classed into lipid, polymeric, and inorganic particles, all of which have been reported in the literature. The advantage of synthetic systems is that they can deliver the CRISPR/Cas system also as a preformed ribonucleoprotein complex. The transient nature of this approach favors low frequencies of off-target events and minimizes the window of immune activation. Moreover, from a pharmaceutical perspective, synthetic delivery systems are much easier to scale up for clinical use compared to viral vectors and can be chemically functionalized with ligands to obtain target cell specificity. The first preclinical results with lipid nanoparticles delivering CRISPR/Cas either as mRNA or ribonucleoproteins are very promising. The goal is translating these CRISPR/Cas therapeutics to a clinical setting as well. Taken together, these current trends seem to favor the use of sgRNA/Cas ribonucleoprotein complexes delivered in vivo by synthetic particles.


Assuntos
Proteína 9 Associada à CRISPR/farmacologia , Sistemas CRISPR-Cas/genética , Portadores de Fármacos/química , Edição de Genes/métodos , Nanopartículas Metálicas/química , Animais , Proteína 9 Associada à CRISPR/genética , Técnicas de Transferência de Genes , Humanos , Camundongos , RNA Guia de Cinetoplastídeos/genética , RNA Guia de Cinetoplastídeos/farmacologia , Ribonucleoproteínas/genética , Ribonucleoproteínas/farmacologia
14.
Mol Cell Biol ; 39(13)2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31010806

RESUMO

The nuclear factor (erythroid 2)-like 2 (NRF2 or NFE2L2) transcription factor regulates the expression of many genes that are critical in maintaining cellular homeostasis. Its deregulation has been implicated in many diseases, including cancer and metabolic and neurodegenerative diseases. While several mechanisms by which NRF2 can be activated have gradually been identified over time, a more complete regulatory network of NRF2 is still lacking. Here we show through a genome-wide clustered regularly interspaced short palindromic repeat (CRISPR) screen that a total of 273 genes, when knocked out, will lead to sustained NRF2 activation. Pathway analysis revealed a significant overrepresentation of genes (18 of the 273 genes) involved in autophagy. Molecular validation of a subset of the enriched genes identified 8 high-confidence genes that negatively regulate NRF2 activity irrespective of cell type: ATG12, ATG7, GOSR1, IFT172, NRXN2, RAB6A, VPS37A, and the well-known negative regulator of NRF2, KEAP1 Of these, ATG12, ATG7, KEAP1, and VPS37A are known to be involved in autophagic processes. Our results present a comprehensive list of NRF2 negative regulators and reveal an intimate link between autophagy and NRF2 regulation.


Assuntos
Redes Reguladoras de Genes , Fator 2 Relacionado a NF-E2/metabolismo , RNA Guia de Cinetoplastídeos/farmacologia , Autofagia , Sistemas CRISPR-Cas , Linhagem Celular Tumoral , Regulação da Expressão Gênica , Redes Reguladoras de Genes/efeitos dos fármacos , Genoma Humano , Homeostase , Humanos
15.
J Am Chem Soc ; 140(49): 17095-17105, 2018 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-30394729

RESUMO

Bone marrow endothelial cells (BMECs) regulate their microenvironment, which includes hematopoietic stem cells. This makes BMECs an important target cell type for siRNA or gene editing (e.g., CRISPR) therapies. However, siRNA and sgRNA have not been delivered to BMECs using systemically administered nanoparticles. Given that in vitro nanoparticle screens have not identified nanoparticles with BMEC tropism, we developed a system to quantify how >100 different nanoparticles deliver siRNA in a single mouse. This is the first barcoding system capable of quantifying functional cytosolic siRNA delivery (where the siRNA drug is active), distinguishing it from in vivo screens that quantify biodistribution (where the siRNA drug went). Combining this approach with bioinformatics, we performed in vivo directed evolution, and identified BM1, a lipid nanoparticle (LNP) that delivers siRNA and sgRNA to BMECs. Interestingly, chemical analysis revealed BMEC tropism was not related to LNP size; tropism changed with the structure of poly(ethylene glycol), as well as the presence of cholesterol. These results suggest that significant changes to vascular targeting can be imparted to a LNP by making simple changes to its chemical composition, rather than using active targeting ligands. BM1 is the first nanoparticle to efficiently deliver siRNA and sgRNA to BMECs in vivo, demonstrating that this functional in vivo screen can identify nanoparticles with novel tropism in vivo. More generally, in vivo screening may help reveal the complex relationship between nanoparticle structure and tropism, thereby helping scientists understand how simple chemical changes control nanoparticle targeting.


Assuntos
Medula Óssea/metabolismo , Portadores de Fármacos/química , Nanopartículas/química , RNA Guia de Cinetoplastídeos/farmacologia , RNA Interferente Pequeno/farmacologia , Animais , Antígenos CD/genética , Moléculas de Adesão Celular/genética , Biologia Computacional , Evolução Molecular Direcionada , Portadores de Fármacos/metabolismo , Células Endoteliais/metabolismo , Inativação Gênica , Camundongos , Nanopartículas/metabolismo , Fosfatidilcolinas/química , Fosfatidilcolinas/metabolismo , Polietilenoglicóis/química , Polietilenoglicóis/metabolismo , RNA Guia de Cinetoplastídeos/genética , RNA Interferente Pequeno/genética
16.
Sci Rep ; 6: 36921, 2016 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-27841295

RESUMO

Progressive multifocal leukoencephalopathy (PML) is a debilitating disease resulting from infection of oligodendrocytes by the JC polyomavirus (JCPyV). Currently, there is no anti-viral therapeutic available against JCPyV infection. The clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR-associated protein 9 (Cas9) system (CRISPR/Cas9) is a genome editing tool capable of introducing sequence specific breaks in double stranded DNA. Here we show that the CRISPR/Cas9 system can restrict the JCPyV life cycle in cultured cells. We utilized CRISPR/Cas9 to target the noncoding control region and the late gene open reading frame of the JCPyV genome. We found significant inhibition of virus replication and viral protein expression in cells recipient of Cas9 together with JCPyV-specific single-guide RNA delivered prior to or after JCPyV infection.


Assuntos
Edição de Genes/métodos , Vírus JC/fisiologia , Leucoencefalopatia Multifocal Progressiva/virologia , Infecções por Polyomavirus/virologia , Sistemas CRISPR-Cas , Genoma Viral/efeitos dos fármacos , Células HEK293 , Humanos , Vírus JC/efeitos dos fármacos , Vírus JC/genética , Fases de Leitura Aberta/efeitos dos fármacos , RNA Guia de Cinetoplastídeos/farmacologia , Proteínas Virais/genética , Replicação Viral/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA