Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Emerg Microbes Infect ; 13(1): 2284286, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37982370

RESUMEN

The persistence of HBV covalently closed circular DNA (cccDNA) and HBV integration into the host genome in infected hepatocytes pose significant challenges to the cure of chronic HBV infection. Although CRISPR/Cas9-mediated genome editing shows promise for targeted clearance of viral genomes, a safe and efficient delivery method is currently lacking. Here, we developed a novel approach by combining light-induced heterodimerization and protein acylation to enhance the loading efficiency of Cas9 protein into extracellular vesicles (EVs). Moreover, vesicular stomatitis virus-glycoprotein (VSV-G) was incorporated onto the EVs membrane, significantly facilitating the endosomal escape of Cas9 protein and increasing its gene editing activity in recipient cells. Our results demonstrated that engineered EVs containing Cas9/gRNA and VSV-G can effectively reduce viral antigens and cccDNA levels in the HBV-replicating and infected cell models. Notably, we also confirmed the antiviral activity and high safety of the engineered EVs in the HBV-replicating mouse model generated by hydrodynamic injection and the HBV transgenic mouse model. In conclusion, engineered EVs could successfully mediate functional CRISPR/Cas9 delivery both in vitro and in vivo, leading to the clearance of episomal cccDNA and integrated viral DNA fragments, and providing a novel therapeutic approach for curing chronic HBV infection.


Asunto(s)
Virus de la Hepatitis B , Hepatitis B , Animales , Ratones , Virus de la Hepatitis B/metabolismo , Proteína 9 Asociada a CRISPR/genética , Proteína 9 Asociada a CRISPR/metabolismo , Proteína 9 Asociada a CRISPR/farmacología , ADN Circular/genética , ADN Circular/metabolismo , Sistemas CRISPR-Cas , ARN Guía de Sistemas CRISPR-Cas , ADN Viral/genética , ADN Viral/metabolismo , Hepatitis B/genética , Replicación Viral
2.
Rev Soc Bras Med Trop ; 55: e0131, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35976333

RESUMEN

Schistosomiasis is a neglected acute and chronic tropical disease caused by intestinal (Schistosoma mansoni and Schistosoma japonicum) and urogenital (Schistosoma haematobium) helminth parasites (blood flukes or digenetic trematodes). It afflicts over 250 million people worldwide, the majority of whom reside in impoverished tropical and subtropical regions in sub-Saharan Africa. Schistosomiasis is the second most common devastating parasitic disease in the world after malaria and causes over 200,000 deaths annually. Currently, there is no effective and approved vaccine available for human use, and treatment strongly relies on praziquantel drug therapy, which is ineffective in killing immature larval schistosomula stages and eggs already lodged in the tissues. The Clustered Regularly Interspaced Short Palindromic Repeats/CRISPR associated protein 9 (CRISPR/Cas9)-mediated gene editing tool is used to deactivate a gene of interest to scrutinize its role in health and disease, and to identify genes for vaccine and drug targeting. The present review aims to summarize the major findings from the current literature reporting the usage of CRISPR/Cas9-mediated gene editing to inactivate genes in S. mansoni (acetylcholinesterase (AChE), T2 ribonuclease omega-1 (ω1), sulfotransferase oxamniquine resistance protein (SULT-OR), and α-N-acetylgalactosaminidase (SmNAGAL)), and freshwater gastropod snails, Biomphalaria glabrata (allograft inflammatory factor (BgAIF)), an obligatory component of the life cycle of S. mansoni, to identify their roles in the pathogenesis of schistosomiasis, and to highlight the importance of such studies in identifying and developing drugs and vaccines with high therapeutic efficacy.


Asunto(s)
Esquistosomiasis mansoni , Esquistosomiasis , Acetilcolinesterasa/farmacología , Animales , Proteína 9 Asociada a CRISPR/farmacología , Humanos , Schistosoma mansoni , Esquistosomiasis mansoni/prevención & control , Desarrollo de Vacunas
3.
Autophagy ; 18(10): 2350-2367, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35130104

RESUMEN

Zaire ebolavirus (EBOV) causes a severe hemorrhagic fever in humans and non-human primates with high morbidity and mortality. EBOV infection is dependent on its structural glycoprotein (GP), but high levels of GP expression also trigger cell rounding, detachment, and downregulation of many surface molecules that is thought to contribute to its high pathogenicity. Thus, EBOV has evolved an RNA editing mechanism to reduce its GP expression and increase its fitness. We now report that the GP expression is also suppressed at the protein level in cells by protein disulfide isomerases (PDIs). Although PDIs promote oxidative protein folding by catalyzing correct disulfide formation in the endoplasmic reticulum (ER), PDIA3/ERp57 adversely triggered the GP misfolding by targeting GP cysteine residues and activated the unfolded protein response (UPR). Abnormally folded GP was targeted by ER-associated protein degradation (ERAD) machinery and, unexpectedly, was degraded via the macroautophagy/autophagy-lysosomal pathway, but not the proteasomal pathway. PDIA3 also decreased the GP expression from other ebolavirus species but increased the GP expression from Marburg virus (MARV), which is consistent with the observation that MARV-GP does not cause cell rounding and detachment, and MARV does not regulate its GP expression via RNA editing during infection. Furthermore, five other PDIs also had a similar inhibitory activity to EBOV-GP. Thus, PDIs negatively regulate ebolavirus glycoprotein expression, which balances the viral life cycle by maximizing their infection but minimizing their cellular effect. We suggest that ebolaviruses hijack the host protein folding and ERAD machinery to increase their fitness via reticulophagy during infection.Abbreviations: 3-MA: 3-methyladenine; 4-PBA: 4-phenylbutyrate; ACTB: ß-actin; ATF: activating transcription factor; ATG: autophagy-related; BafA1: bafilomycin A1; BDBV: Bundibugyo ebolavirus; CALR: calreticulin; CANX: calnexin; CHX: cycloheximide; CMA: chaperone-mediated autophagy; ConA: concanamycin A; CRISPR: clusters of regularly interspaced short palindromic repeats; Cas9: CRISPR-associated protein 9; dsRNA: double-stranded RNA; EBOV: Zaire ebolavirus; EDEM: ER degradation enhancing alpha-mannosidase like protein; EIF2AK3/PERK: eukaryotic translation initiation factor 2 alpha kinase 3; Env: envelope glycoprotein; ER: endoplasmic reticulum; ERAD: ER-associated protein degradation; ERN1/IRE1: endoplasmic reticulum to nucleus signaling 1; GP: glycoprotein; HA: hemagglutinin; HDAC6: histone deacetylase 6; HMM: high-molecular-mass; HIV-1: human immunodeficiency virus type 1; HSPA5/BiP: heat shock protein family A (Hsp70) member 5; IAV: influenza A virus; IP: immunoprecipitation; KIF: kifenesine; Lac: lactacystin; LAMP: lysosomal associated membrane protein; MAN1B1/ERManI: mannosidase alpha class 1B member 1; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MARV: Marburg virus; MLD: mucin-like domain; NHK/SERPINA1: alpha1-antitrypsin variant null (Hong Kong); NTZ: nitazoxanide; PDI: protein disulfide isomerase; RAVV: Ravn virus; RESTV: Reston ebolavirus; SARS-CoV: severe acute respiratory syndrome coronavirus; SBOV: Sudan ebolavirus; sGP: soluble GP; SQSTM1/p62: sequestosome 1; ssGP: small soluble GP; TAFV: Taï Forest ebolavirus; TIZ: tizoxanide; TGN: thapsigargin; TLD: TXN (thioredoxin)-like domain; Ub: ubiquitin; UPR: unfolded protein response; VLP: virus-like particle; VSV: vesicular stomatitis virus; WB: Western blotting; WT: wild-type; XBP1: X-box binding protein 1.


Asunto(s)
Autofagia , Ebolavirus , Actinas/metabolismo , Animales , Proteína 9 Asociada a CRISPR/genética , Proteína 9 Asociada a CRISPR/metabolismo , Proteína 9 Asociada a CRISPR/farmacología , Calnexina/metabolismo , Calreticulina/genética , Calreticulina/metabolismo , Calreticulina/farmacología , Cicloheximida , Cisteína/metabolismo , Disulfuros , Retículo Endoplásmico/metabolismo , Glicoproteínas/metabolismo , Proteínas de Choque Térmico/metabolismo , Hemaglutininas/metabolismo , Hemaglutininas/farmacología , Histona Desacetilasa 6/genética , Péptidos y Proteínas de Señalización Intercelular , Proteínas de Membrana de los Lisosomas/metabolismo , Lisosomas/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Mucinas/genética , Mucinas/metabolismo , Mucinas/farmacología , Factor 2 Procariótico de Iniciación/genética , Factor 2 Procariótico de Iniciación/metabolismo , Factor 2 Procariótico de Iniciación/farmacología , Proteína Disulfuro Isomerasas/genética , Proteína Disulfuro Isomerasas/metabolismo , ARN Bicatenario/metabolismo , ARN Bicatenario/farmacología , Proteína Sequestosoma-1/metabolismo , Tapsigargina/metabolismo , Tapsigargina/farmacología , Tiorredoxinas/genética , Tiorredoxinas/metabolismo , Tiorredoxinas/farmacología , Ubiquitinas/metabolismo , Proteína 1 de Unión a la X-Box/metabolismo , alfa-Manosidasa/genética , alfa-Manosidasa/metabolismo , alfa-Manosidasa/farmacología
4.
DNA Repair (Amst) ; 109: 103257, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34847381

RESUMEN

Cas9 targets DNA during genome editing by forming an RNA:DNA heteroduplex (R-loop) between the Cas9-bound guide RNA and the targeted DNA strand. We have recently demonstrated that R-loop formation by catalytically inactive Cas9 (dCas9) is inherently mutagenic, in part, by promoting spontaneous cytosine deamination within the non-targeted single-stranded DNA of the dCas9-induced R-loop. However, the extent to which dCas9 binding and R-loop formation affect the subsequent repair of uracil lesions or other damaged DNA bases is unclear. Here, we show that DNA binding by dCas9 inhibits initiation of base excision repair (BER) for uracil lesions in vitro. Our data indicate that cleavage of uracil lesions by Uracil-DNA glycosylase (UDG) is generally inhibited at dCas9-bound DNA, in both the dCas9:sgRNA-bound target strand (TS) or the single-stranded non-target strand (NT). However, cleavage of a uracil lesion within the base editor window of the NT strand was less inhibited than at other locations, indicating that this site is more permissive to UDG activity. Furthermore, our data suggest that dCas9 binding to PAM sites can inhibit UDG activity. However, this non-specific inhibition can be relieved with the addition of an sgRNA lacking sequence complementarity to the DNA substrate. Moreover, we show that dCas9 binding also inhibits human single-strand selective monofunctional uracil-DNA glycosylase (SMUG1). Structural analysis of a Cas9-bound target site subsequently suggests a molecular mechanism for BER inhibition. Taken together, our results imply that dCas9 (or Cas9) binding may promote background mutagenesis by inhibiting the removal of DNA base lesions by BER.


Asunto(s)
Proteína 9 Asociada a CRISPR/metabolismo , Daño del ADN , Reparación del ADN , Uracil-ADN Glicosidasa/antagonistas & inhibidores , Uracilo/metabolismo , Proteína 9 Asociada a CRISPR/farmacología , ADN/metabolismo , Edición Génica , Humanos , Mutagénesis
5.
Bioorg Med Chem Lett ; 40: 127925, 2021 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-33705909

RESUMEN

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.


Asunto(s)
Portadores de Fármacos/química , Endosomas/metabolismo , Homoarginina/química , Membranas Intracelulares/metabolismo , Péptidos/química , Secuencia de Aminoácidos , Proteína 9 Asociada a CRISPR/farmacología , Dextranos/química , Portadores de Fármacos/toxicidad , Liberación de Fármacos , Fluoresceínas/química , Colorantes Fluorescentes/química , Células HeLa , Humanos , Inmunoglobulina G/farmacología , Integrasas/farmacología , Liposomas/química , Péptidos/toxicidad , ARN Guía de Kinetoplastida/farmacología , Ácidos Sulfónicos/química
6.
Anim Sci J ; 91(1): e13386, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32512638

RESUMEN

This study was conducted to investigate the effect of seven concentrations of Cas9 protein (0, 25, 50, 100, 200, 500, and 1,000 ng/µl) on the development and gene editing of porcine embryos. This included the target editing and off-target effect of embryos developed from zygotes that were edited via electroporation of the Cas9 protein with guide RNA targeting Myostatin genes. We found that the development to blastocysts of electroporated zygotes was not affected by the concentration of Cas9 protein. Although the editing rate, which was defined as the ratio of edited blastocysts to total examined blastocysts, did not differ with Cas9 protein concentration, the editing efficiency, which was defined as the frequency of indel mutations in each edited blastocyst, was significantly decreased in the edited blastocysts from zygotes electroporated with 25 ng/µl of Cas9 protein compared with that of blastocysts from zygotes electroporated with higher Cas9 protein concentrations. Moreover the frequency of indel events at the two possible off-target sites was not significantly different with different concentrations of Cas9 protein. These results indicate that the concentration of Cas9 protein affects gene editing efficiency in embryos but not the embryonic development, gene editing rate, and non-specific cleavage of off-target sites.


Asunto(s)
Proteína 9 Asociada a CRISPR , Electroporación/métodos , Electroporación/veterinaria , Desarrollo Embrionario/genética , Edición Génica , Marcación de Gen/veterinaria , Miostatina/genética , ARN Guía de Kinetoplastida , Porcinos/embriología , Porcinos/genética , Cigoto , Animales , Blastocisto , Proteína 9 Asociada a CRISPR/farmacología , Relación Dosis-Respuesta a Droga
7.
ACS Appl Mater Interfaces ; 12(19): 21386-21397, 2020 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-32315156

RESUMEN

A preassembled Cas9/single-guide RNA complex (Cas9 ribonucleoprotein; Cas9 RNP) induces genome editing efficiently, with small off-target effects compared with the conventional techniques, such as plasmid DNA and mRNA systems. However, penetration of Cas9 RNP through the cell membrane is low. In particular, the incorporation of Cas9 RNP into neurons and the brain is challenging. In the present study, we have reported the use of a dendrimer (generation 3; G3)/glucuronylglucosyl-ß-cyclodextrin conjugate (GUG-ß-CDE (G3)) as a carrier of Cas9 RNP and evaluated genome editing activity in the neuron and the brain. A Cas9 RNP ternary complex with GUG-ß-CDE (G3) was prepared by only mixing the components. The resulting complex exhibited higher genome editing activity than the complex with the dendrimer (G3), Lipofectamine 3000 or Lipofectamine CRISPRMAX in SH-SY5Y cells, a human neuroblastoma cell line. In addition, GUG-ß-CDE (G3) enhanced the genome editing activity of Cas9 RNP in the whole mouse brain after a single intraventricular administration. Thus, GUG-ß-CDE (G3) is a useful Cas9 RNP carrier that can induce genome editing in the neuron and brain.


Asunto(s)
Encéfalo/metabolismo , Proteína 9 Asociada a CRISPR/farmacología , Dendrímeros/química , Portadores de Fármacos/química , Edición Génica/métodos , Ribonucleoproteínas/farmacología , Animales , Sistemas CRISPR-Cas , Línea Celular Tumoral , Técnicas de Transferencia de Gen , Humanos , Ratones Endogámicos BALB C
8.
ACS Appl Mater Interfaces ; 11(38): 34717-34724, 2019 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-31469541

RESUMEN

The CRISPR/Cas gene editing system has been successfully applied to combating bacteria, cancer, virus, and genetic disorders. While viral vectors have been used for the delivery of the CRISPR/Cas9 system, the time required for insert cloning, and virus packaging and standardization, hinders its efficient use. Additionally, the high molecular weight of the Cas9 endonuclease makes it not easy for packing into the vehicles. Herein we report the self-assembly of gold nanoclusters (AuNCs) with SpCas9 protein (SpCas9-AuNCs) under physiological conditions and the efficient delivery of SpCas9 into the cell nucleus. This assembly process is highly dependent on pH. SpCas9-AuNCs are stable at a higher pH but are disassembled at a lower pH. Significantly, this assembly-disassembly process facilitates the delivery of SpCas9 into cells and the cell nucleus, where the SpCas9 exerts its cleavage function. As a proof-of-concept, the assembled SpCas9-AuNCs nanoparticles are successfully used for efficient knockout of the E6 oncogene, restoring the function of tumor-suppressive protein p53 and inducing apoptosis in cervical cancer cells with little effect on normal human cells. The SpCas9-AuNCs are useful for sgRNA functional validation, sgRNA library screening, and genomic manipulation.


Asunto(s)
Proteína 9 Asociada a CRISPR , Genes Virales , Oro , Nanopartículas del Metal , Oncogenes , Proteína p53 Supresora de Tumor/metabolismo , Neoplasias del Cuello Uterino , Proteína 9 Asociada a CRISPR/química , Proteína 9 Asociada a CRISPR/farmacología , Femenino , Oro/química , Oro/farmacología , Células HeLa , Humanos , Concentración de Iones de Hidrógeno , Nanopartículas del Metal/química , Nanopartículas del Metal/uso terapéutico , Neoplasias del Cuello Uterino/tratamiento farmacológico , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/virología
9.
Acc Chem Res ; 52(6): 1555-1564, 2019 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-31099553

RESUMEN

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.


Asunto(s)
Proteína 9 Asociada a CRISPR/farmacología , Sistemas CRISPR-Cas/genética , Portadores de Fármacos/química , Edición Génica/métodos , Nanopartículas del Metal/química , Animales , Proteína 9 Asociada a CRISPR/genética , Técnicas de Transferencia de Gen , Humanos , Ratones , ARN Guía de Kinetoplastida/genética , ARN Guía de Kinetoplastida/farmacología , Ribonucleoproteínas/genética , Ribonucleoproteínas/farmacología
10.
Nucleic Acid Ther ; 29(3): 136-147, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30990769

RESUMEN

Clustered regularly interspaced short palindromic repeat (CRISPR) RNAs and their associated effector (Cas) enzymes are being developed into promising therapeutics to treat disease. However, CRISPR-Cas enzymes might produce unwanted gene editing or dangerous side effects. Drug-like molecules that can inactivate CRISPR-Cas enzymes could help facilitate safer therapeutic development. Based on the requirement of guide RNA and target DNA interaction by Cas enzymes, we rationally designed small nucleic acid-based inhibitors (SNuBs) of Streptococcus pyogenes (Sp) Cas9. Inhibitors were initially designed as 2'-O-methyl-modified oligonucleotides that bound the CRISPR RNA guide sequence (anti-guide) or repeat sequence (anti-tracr), or DNA oligonucleotides that bound the protospacer adjacent motif (PAM)-interaction domain (anti-PAM) of SpCas9. Coupling anti-PAM and anti-tracr modules together was synergistic and resulted in high binding affinity and efficient inhibition of Cas9 DNA cleavage activity. Incorporating 2'F-RNA and locked nucleic acid nucleotides into the anti-tracr module resulted in greater inhibition as well as dose-dependent suppression of gene editing in human cells. CRISPR SNuBs provide a platform for rational design of CRISPR-Cas enzyme inhibitors that should translate to other CRISPR effector enzymes and enable better control over CRISPR-based applications.


Asunto(s)
Proteína 9 Asociada a CRISPR/genética , Sistemas CRISPR-Cas/genética , Proteínas de Unión al ADN/genética , Edición Génica , Proteína 9 Asociada a CRISPR/antagonistas & inhibidores , Proteína 9 Asociada a CRISPR/farmacología , Sistemas CRISPR-Cas/efectos de los fármacos , ADN/efectos de los fármacos , ADN/genética , Proteínas de Unión al ADN/efectos de los fármacos , Humanos , Motivos de Nucleótidos/efectos de los fármacos , Motivos de Nucleótidos/genética , Oligonucleótidos/genética , Oligonucleótidos/farmacología , ARN Guía de Kinetoplastida/efectos adversos , ARN Guía de Kinetoplastida/genética , ARN Guía de Kinetoplastida/farmacología , Streptococcus pyogenes/enzimología , Secuencias Repetidas en Tándem/efectos de los fármacos , Secuencias Repetidas en Tándem/genética
11.
Biochem Biophys Res Commun ; 506(1): 278-283, 2018 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-30348526

RESUMEN

Cas9 (CRISPR associated protein 9) is an RNA-guided DNA endonuclease enzyme derived from Streptococcus that has been widely used for genome editing in a variety of organisms, including humans. Here, we report that exogenous Cas9 protein can elicit an inflammatory immune response through the release of MIP3α, CD40L, and MPO in primary human peripheral blood mononuclear cells and human monocytic cell lines (THP1). Inhibition of the STING-STAT6 pathway blocks Cas9-induced proinflammatory mediator release. These results suggest that targeting the STING-STAT6 axis may prevent host immune responses in human gene therapy with the CRISPR-Cas9 system.


Asunto(s)
Proteína 9 Asociada a CRISPR/farmacología , Inflamación/inmunología , Proteínas de la Membrana/metabolismo , Monocitos/inmunología , Factor de Transcripción STAT6/metabolismo , Proteínas Bacterianas/farmacología , Interacciones Microbiota-Huesped/inmunología , Humanos , Inflamación/inducido químicamente , Células THP-1
12.
Antiviral Res ; 152: 58-67, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29458131

RESUMEN

Chronic hepatitis B virus (HBV) infection is difficult to cure due to the presence of covalently closed circular DNA (cccDNA). Accumulating evidence indicates that the CRISPR/Cas9 system effectively disrupts HBV genome, including cccDNA, in vitro and in vivo. However, efficient delivery of CRISPR/Cas9 system to the liver or hepatocytes using an adeno-associated virus (AAV) vector remains challenging due to the large size of Cas9 from Streptococcus pyogenes (Sp). The recently identified Cas9 protein from Staphylococcus aureus (Sa) is smaller than SpCas9 and thus is able to be packaged into the AAV vector. To examine the efficacy of SaCas9 system on HBV genome destruction, we designed 5 guide RNAs (gRNAs) that targeted different HBV genotypes, 3 of which were shown to be effective. The SaCas9 system significantly reduced HBV antigen expression, as well as pgRNA and cccDNA levels, in Huh7, HepG2.2.15 and HepAD38 cells. The dual expression of gRNAs/SaCas9 in these cell lines resulted in more efficient HBV genome cleavage. In the mouse model, hydrodynamic injection of gRNA/SaCas9 plasmids resulted in significantly lower levels of HBV protein expression. We also delivered the SaCas9 system into mice with persistent HBV replication using an AAV vector. Both the AAV vector and the mRNA of Cas9 could be detected in the C3H mouse liver cells. Decreased hepatitis B surface antigen (HBsAg), HBV DNA and pgRNA levels were observed when a higher titer of AAV was injected, although this decrease was not significantly different from the control. In summary, the SaCas9 system accurately and efficiently targeted the HBV genome and inhibited HBV replication both in vitro and in vivo. The system was delivered by an AAV vector and maybe used as a novel therapeutic strategy against chronic HBV infection.


Asunto(s)
Antivirales/farmacología , Proteínas Bacterianas/farmacología , Proteína 9 Asociada a CRISPR/farmacología , Virus de la Hepatitis B/efectos de los fármacos , Virus de la Hepatitis B/genética , Hepatitis B Crónica/virología , Staphylococcus aureus/enzimología , Replicación Viral/efectos de los fármacos , Animales , Antivirales/economía , Antivirales/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Proteína 9 Asociada a CRISPR/genética , Proteína 9 Asociada a CRISPR/metabolismo , División del ADN/efectos de los fármacos , ADN Viral/genética , ADN Viral/metabolismo , Virus de la Hepatitis B/fisiología , Humanos , Ratones , Ratones Endogámicos C3H , Staphylococcus aureus/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...