Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
PLoS Pathog ; 20(7): e1012345, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38968329

RESUMO

The CRISPR-Cas13 system has been proposed as an alternative treatment of viral infections. However, for this approach to be adopted as an antiviral, it must be optimized until levels of efficacy rival or exceed the performance of conventional approaches. To take steps toward this goal, we evaluated the influenza viral RNA degradation patterns resulting from the binding and enzymatic activity of mRNA-encoded LbuCas13a and two crRNAs from a prior study, targeting PB2 genomic and messenger RNA. We found that the genome targeting guide has the potential for significantly higher potency than originally detected, because degradation of the genomic RNA is not uniform across the PB2 segment, but it is augmented in proximity to the Cas13 binding site. The PB2 genome targeting guide exhibited high levels (>1 log) of RNA degradation when delivered 24 hours post-infection in vitro and maintained that level of degradation over time, with increasing multiplicity of infection (MOI), and across modern influenza H1N1 and H3N2 strains. Chemical modifications to guides with potent LbuCas13a function, resulted in nebulizer delivered efficacy (>1-2 log reduction in viral titer) in a hamster model of influenza (Influenza A/H1N1/California/04/09) infection given prophylactically or as a treatment (post-infection). Maximum efficacy was achieved with two doses, when administered both pre- and post-infection. This work provides evidence that mRNA-encoded Cas13a can effectively mitigate Influenza A infections opening the door to the development of a programmable approach to treating multiple respiratory infections.


Assuntos
Sistemas CRISPR-Cas , Influenza Humana , Estabilidade de RNA , RNA Mensageiro , RNA Viral , Animais , RNA Viral/genética , RNA Viral/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Humanos , Influenza Humana/virologia , Vírus da Influenza A Subtipo H1N1/genética , Vírus da Influenza A Subtipo H3N2/genética , Infecções por Orthomyxoviridae/virologia , Antivirais/farmacologia , Cães , Cricetinae , Proteínas Virais/genética , Proteínas Virais/metabolismo , Mesocricetus , Células Madin Darby de Rim Canino
2.
Nat Microbiol ; 9(8): 2160-2172, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38839984

RESUMO

Dengue is a major global health threat, and there are no approved antiviral agents. Prior research using Cas13 only demonstrated dengue mitigation in vitro. Here we demonstrate that systemic delivery of mRNA-encoded Cas13a and guide RNAs formulated in lipid nanoparticles can be used to treat dengue virus (DENV) 2 and 3 in mice. First, we identified guides against DENV 2 and 3 that demonstrated in vitro efficacy. Next, we confirmed that Cas13 enzymatic activity is necessary for DENV 2 or DENV 3 mitigation in vitro. Last, we show that a single dose of lipid-nanoparticle-formulated mRNA-encoded Cas13a and guide RNA, administered 1 day post-infection, promotes survival of all infected animals and serum viral titre decreases on days 2 and 3 post-infection after lethal challenge in mice. Off-target analysis in mice using RNA sequencing showed no collateral cleavage. Overall, these data demonstrate the potential of mRNA-encoded Cas13 as a pan-DENV drug.


Assuntos
Antivirais , Vírus da Dengue , Dengue , Modelos Animais de Doenças , Nanopartículas , RNA Mensageiro , Animais , Dengue/tratamento farmacológico , Camundongos , Vírus da Dengue/genética , Vírus da Dengue/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Nanopartículas/química , Antivirais/farmacologia , Antivirais/administração & dosagem , RNA Guia de Sistemas CRISPR-Cas/genética , Humanos , Proteínas Associadas a CRISPR/metabolismo , Proteínas Associadas a CRISPR/genética , Lipídeos/química , Carga Viral/efeitos dos fármacos , Feminino , Lipossomos
3.
Nat Chem ; 15(4): 508-515, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36864143

RESUMO

Stereochemistry can alter small-molecule pharmacokinetics, safety and efficacy. However, it is unclear whether the stereochemistry of a single compound within a multicomponent colloid such as a lipid nanoparticle (LNP) can influence its activity in vivo. Here we report that LNPs containing stereopure 20α-hydroxycholesterol (20α) delivered mRNA to liver cells up to 3-fold more potently than LNPs containing a mixture of both 20α- and 20ß-hydroxycholesterols (20mix). This effect was not driven by LNP physiochemical traits. Instead, in vivo single-cell RNA sequencing and imaging revealed that 20mix LNPs were sorted into phagocytic pathways more than 20α LNPs, resulting in key differences between LNP biodistribution and subsequent LNP functional delivery. These data are consistent with the fact that nanoparticle biodistribution is necessary, but not sufficient, for mRNA delivery, and that stereochemistry-dependent interactions between LNPs and target cells can improve mRNA delivery.


Assuntos
Lipídeos , Nanopartículas , Lipídeos/química , RNA Mensageiro/genética , Distribuição Tecidual , Nanopartículas/química
4.
Nat Mater ; 22(3): 369-379, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36443576

RESUMO

Messenger RNA has now been used to vaccinate millions of people. However, the diversity of pulmonary pathologies, including infections, genetic disorders, asthma and others, reveals the lung as an important organ to directly target for future RNA therapeutics and preventatives. Here we report the screening of 166 polymeric nanoparticle formulations for functional delivery to the lungs, obtained from a combinatorial synthesis approach combined with a low-dead-volume nose-only inhalation system for mice. We identify P76, a poly-ß-amino-thio-ester polymer, that exhibits increased expression over formulations lacking the thiol component, delivery to different animal species with varying RNA cargos and low toxicity. P76 allows for dose sparing when delivering an mRNA-expressed Cas13a-mediated treatment in a SARS-CoV-2 challenge model, resulting in similar efficacy to a 20-fold higher dose of a neutralizing antibody. Overall, the combinatorial synthesis approach allowed for the discovery of promising polymeric formulations for future RNA pharmaceutical development for the lungs.


Assuntos
COVID-19 , Animais , Camundongos , RNA Mensageiro/genética , SARS-CoV-2/genética , Polímeros/metabolismo , Pulmão , RNA/metabolismo
5.
Adv Sci (Weinh) ; 9(34): e2202771, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36316224

RESUMO

Despite the success of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) vaccines, there remains a clear need for new classes of preventatives for respiratory viral infections due to vaccine hesitancy, lack of sterilizing immunity, and for at-risk patient populations, including the immunocompromised. While many neutralizing antibodies have been identified, and several approved, to treat COVID-19, systemic delivery, large doses, and high costs have the potential to limit their widespread use, especially in low- and middle-income countries. To use these antibodies more efficiently, an inhalable formulation is developed that allows for the expression of mRNA-encoded, membrane-anchored neutralizing antibodies in the lung to mitigate SARS-CoV-2 infections. First, the ability of mRNA-encoded, membrane-anchored, anti-SARS-CoV-2 antibodies to prevent infections in vitro is demonstrated. Next, it is demonstrated that nebulizer-based delivery of these mRNA-expressed neutralizing antibodies potently abrogates disease in the hamster model. Overall, these results support the use of nebulizer-based mRNA expression of neutralizing antibodies as a new paradigm for mitigating respiratory virus infections.


Assuntos
COVID-19 , Humanos , SARS-CoV-2 , RNA Mensageiro/genética , Anticorpos Neutralizantes/uso terapêutico
6.
ACS Nano ; 16(4): 5660-5671, 2022 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-35357116

RESUMO

Programmable control of gene expression via nuclease-null Cas9 fusion proteins has enabled the engineering of cellular behaviors. Here, both transcriptional and epigenetic gene activation via synthetic mRNA and lipid nanoparticle delivery was demonstrated in vivo. These highly efficient delivery strategies resulted in high levels of activation in multiple tissues. Finally, we demonstrate durable gene activation in vivo via transient delivery of a single dose of a gene activator that combines VP64, p65, and HSF1 with a SWI/SNF chromatin remodeling complex component SS18, representing an important step toward gene-activation-based therapeutics. This induced sustained gene activation could be inhibited via mRNA-encoded AcrIIA4, further improving the safety profile of this approach.


Assuntos
Sistemas CRISPR-Cas , Lipossomos , Ativação Transcricional , RNA Mensageiro/genética , Proteína 9 Associada à CRISPR/genética
7.
Nat Biotechnol ; 39(6): 717-726, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33536629

RESUMO

Cas13a has been used to target RNA viruses in cell culture, but efficacy has not been demonstrated in animal models. In this study, we used messenger RNA (mRNA)-encoded Cas13a for mitigating influenza virus A and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in mice and hamsters, respectively. We designed CRISPR RNAs (crRNAs) specific for PB1 and highly conserved regions of PB2 of influenza virus, and against the replicase and nucleocapsid genes of SARS-CoV-2, and selected the crRNAs that reduced viral RNA levels most efficiently in cell culture. We delivered polymer-formulated Cas13a mRNA and the validated guides to the respiratory tract using a nebulizer. In mice, Cas13a degraded influenza RNA in lung tissue efficiently when delivered after infection, whereas in hamsters, Cas13a delivery reduced SARS-CoV-2 replication and reduced symptoms. Our findings suggest that Cas13a-mediated targeting of pathogenic viruses can mitigate respiratory infections.


Assuntos
COVID-19/terapia , Influenza Humana/terapia , RNA Mensageiro/farmacologia , SARS-CoV-2/genética , Animais , COVID-19/genética , COVID-19/virologia , Sistemas CRISPR-Cas/genética , Cricetinae , Modelos Animais de Doenças , Humanos , Influenza Humana/genética , Influenza Humana/virologia , Camundongos , Orthomyxoviridae/efeitos dos fármacos , Orthomyxoviridae/genética , Orthomyxoviridae/patogenicidade , RNA Mensageiro/genética , RNA Viral/genética , Sistema Respiratório/efeitos dos fármacos , Sistema Respiratório/metabolismo , SARS-CoV-2/patogenicidade
8.
PLoS Pathog ; 16(10): e1008987, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33031461

RESUMO

The ribonucleocapsid complex of respiratory syncytial virus (RSV) is responsible for both viral mRNA transcription and viral replication during infection, though little is known about how this dual function is achieved. Here, we report the use of a recombinant RSV virus with a FLAG-tagged large polymerase protein, L, to characterize and localize RSV ribonucleocapsid structures during the early and late stages of viral infection. Through proximity ligation assays and super-resolution microscopy, viral RNA and proteins in the ribonucleocapsid complex were revealed to dynamically rearrange over time, particularly between 6 and 8 hours post infection, suggesting a connection between the ribonucleocapsid structure and its function. The timing of ribonucleocapsid rearrangement corresponded with an increase in RSV genome RNA accumulation, indicating that this rearrangement is likely involved with the onset of RNA replication and secondary transcription. Additionally, early overexpression of RSV M2-2 from in vitro transcribed mRNA was shown to inhibit virus infection by rearranging the ribonucleocapsid complex. Collectively, these results detail a critical understanding into the localization and activity of RSV L and the ribonucleocapsid complex during RSV infection.


Assuntos
Proteínas do Nucleocapsídeo/metabolismo , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sincicial Respiratório Humano/fisiologia , Ribonucleoproteínas/metabolismo , Proteínas Virais/metabolismo , Replicação Viral , Células A549 , Animais , Chlorocebus aethiops , Humanos , Proteínas do Nucleocapsídeo/genética , RNA Viral/genética , RNA Viral/metabolismo , Infecções por Vírus Respiratório Sincicial/genética , Infecções por Vírus Respiratório Sincicial/metabolismo , Ribonucleoproteínas/genética , Transcrição Gênica , Células Vero , Proteínas Virais/genética
9.
Mol Ther ; 28(3): 805-819, 2020 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-31995741

RESUMO

There is a clear need for low-cost, self-applied, long-lasting approaches to prevent human immunodeficiency virus (HIV) infection in both men and women, even with the advent of pre-exposure prophylaxis (PrEP). Broadly neutralizing antibodies represent an option to improve HIV prophylaxis, but intravenous delivery, cold-chain stability requirements, low cervicovaginal concentrations, and cost may preclude their use. Here, we present an approach to express the anti-GP120 broadly neutralizing antibody PGT121 in the primary site of inoculation, the female reproductive tract, using synthetic mRNA. Expression is achieved through aerosol delivery of unformulated mRNA in water. We demonstrated high levels of antibody expression for over 28 days with a single mRNA administration in the reproductive tract of sheep. In rhesus macaques, neutralizing antibody titers in secretions developed within 4 h and simian-HIV (SHIV) infection of ex vivo explants was prevented. Persistence of PGT121 in vaginal secretions and epithelium was achieved through the incorporation of a glycosylphosphatidylinositol (GPI) anchor into the heavy chain of the antibody. Overall, we present a new paradigm to deliver neutralizing antibodies to the female reproductive tract for the prevention of HIV infections.


Assuntos
Anticorpos Amplamente Neutralizantes/imunologia , Expressão Gênica , Anticorpos Anti-HIV/imunologia , Mucosa/imunologia , Mucosa/metabolismo , RNA Mensageiro/administração & dosagem , Vagina , Vacinas contra a AIDS/administração & dosagem , Vacinas contra a AIDS/genética , Vacinas contra a AIDS/imunologia , Aerossóis , Animais , Chlorocebus aethiops , Feminino , Imunofluorescência , Infecções por HIV/imunologia , HIV-1/imunologia , Camundongos , Testes de Neutralização , RNA Mensageiro/síntese química , Ovinos , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia/imunologia , Vagina/imunologia , Vagina/metabolismo , Células Vero
10.
Adv Mater ; 32(1): e1904905, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31743531

RESUMO

Clinical mRNA delivery remains challenging, in large part because how physiology alters delivery in vivo remains underexplored. For example, mRNA delivered by lipid nanoparticles (LNPs) is being considered to treat inflammation, but whether inflammation itself changes delivery remains understudied. Relationships between immunity, endocytosis, and mRNA translation lead to hypothesize that toll-like receptor 4 (TLR4) activation reduced LNP-mediated mRNA delivery. Therefore, LNP uptake, endosomal escape, and mRNA translation with and without TLR4 activation are quantified. In vivo DNA barcoding is used to discover a novel LNP that delivers mRNA to Kupffer cells at clinical doses; unlike most LNPs, this LNP does not preferentially target hepatocytes. TLR4 activation blocks mRNA translation in all tested cell types, without reducing LNP uptake; inhibiting TLR4 or its downstream effector protein kinase R improved delivery. The discrepant effects of TLR4 on i) LNP uptake and ii) translation suggests TLR4 activation can "override" LNP targeting, even after mRNA is delivered into target cells. Given near-future clinical trials using mRNA to modulate inflammation, this highlights the need to understand inflammatory signaling in on- and off-target cells. More generally, this suggests an LNP which delivers mRNA to one inflammatory disease may not deliver mRNA to another.


Assuntos
Imunidade Inata , Nanopartículas/química , RNA Mensageiro/metabolismo , Animais , Endocitose , Endossomos/metabolismo , Imunidade Inata/efeitos dos fármacos , Lipídeos/química , Lipopolissacarídeos/farmacologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Camundongos Transgênicos , Células RAW 264.7 , RNA Mensageiro/química , Receptor 4 Toll-Like/metabolismo
11.
Cancer Res ; 79(20): 5418-5431, 2019 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-31481502

RESUMO

Abnormal post-transcriptional regulation induced by alterations of mRNA-protein interactions is critical during tumorigenesis and cancer progression and is a hallmark of cancer cells. A more thorough understanding is needed to develop treatments and foresee outcomes. Cellular and mouse tumor models are insufficient for vigorous investigation as they lack consistency and translatability to humans. Moreover, to date, studies in human tumor tissue are predominately limited to expression analysis of proteins and mRNA, which do not necessarily provide information about the frequency of mRNA-protein interactions. Here, we demonstrate novel optimization of a method that is based on FISH and proximity ligation techniques to quantify mRNA interactions with RNA-binding proteins relevant for tumorigenesis and cancer progression in archival patient-derived tumor tissue. This method was validated for multiple mRNA-protein pairs in several cellular models and in multiple types of archival human tumor samples. Furthermore, this approach allowed high-throughput analysis of mRNA-protein interactions across a wide range of tumor types and stages through tumor microarrays. This method is quantitative, specific, and sensitive for detecting interactions and their localization at both the individual cell and whole-tissue scales with single interaction sensitivity. This work presents an important tool in investigating post-transcriptional regulation in cancer on a high-throughput scale, with great potential for translatability into any applications where mRNA-protein interactions are of interest. SIGNIFICANCE: This work presents an approach to sensitively, specifically, and quantitatively detect and localize native mRNA and protein interactions for analysis of abnormal post-transcriptional regulation in patient-derived archival tumor samples.


Assuntos
Neoplasias do Colo/química , Ensaios de Triagem em Larga Escala/métodos , Neoplasias Pulmonares/química , Proteínas de Neoplasias/análise , Processamento Pós-Transcricional do RNA , RNA Mensageiro/análise , RNA Neoplásico/análise , Proteínas de Ligação a RNA/análise , Análise Serial de Tecidos/métodos , Animais , Bancos de Espécimes Biológicos , Linhagem Celular Tumoral , Chlorocebus aethiops , Neoplasias do Colo/patologia , Corantes Fluorescentes , Humanos , Hibridização in Situ Fluorescente , Neoplasias Pulmonares/patologia , Camundongos , Microscopia de Fluorescência , Análise de Célula Única , Organismos Livres de Patógenos Específicos , Células Vero
12.
Nat Biomed Eng ; 3(5): 371-380, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30936432

RESUMO

Visualization of the spatio-temporal trafficking of vaccines after their delivery would help evaluate the efficacy of candidate formulations and aid their rational design for preclinical and translational studies. Here, we show that a dual radionuclide-near-infrared probe allows for quantitative, longitudinal and non-invasive monitoring, via positron emission tomography-computed tomography and near-infrared imaging of cynomolgus macaques, of the trafficking dynamics to draining lymph nodes of a model messenger RNA vaccine labelled with the probe. After intramuscular administration of the vaccine to the monkeys, we observed the dynamics of the mRNA vaccine at the injection site and in the draining lymph nodes, performed cellular analyses of the involved tissues using flow cytometry and identified through immunofluorescence that professional antigen-presenting cells are the primary cells containing the injected mRNA and encoding the antigen. This approach may reveal spatio-temporal determinants of vaccine efficacy in preclinical and translational studies employing large mammals.


Assuntos
Técnicas de Transferência de Genes , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , RNA Mensageiro/administração & dosagem , Espectroscopia de Luz Próxima ao Infravermelho , Vacinas/administração & dosagem , Animais , Células Apresentadoras de Antígenos/metabolismo , Radioisótopos de Cobre/química , Células HeLa , Humanos , Linfonodos/diagnóstico por imagem , Macaca fascicularis , Masculino , Músculos/metabolismo
13.
Mol Ther Nucleic Acids ; 14: 52-66, 2019 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-30579042

RESUMO

The characterization of innate immune activation is crucial for vaccine and therapeutic development, including RNA-based vaccines, a promising approach. Current measurement methods quantify type I interferon and inflammatory cytokine production, but they do not allow for the isolation of individual pathways, do not provide kinetic activation or spatial information within tissues, and cannot be translated into clinical studies. Here we demonstrated the use of proximity ligation assays (PLAs) to detect pattern recognition receptor (PRR) activation in cells and in tissue samples. First, we validated PLA's sensitivity and specificity using well-characterized soluble agonists. Next, we characterized PRR activation from in vitro-transcribed (IVT) mRNAs, as well as the effect of sequence and base modifications in vitro. Finally, we established the measurement of PRR activation in tissue sections via PLA upon IVT mRNA intramuscular (i.m.) injection in mice. Overall, our results indicate that PLA is a valuable, versatile, and sensitive tool to monitor PRR activation for vaccine, adjuvant, and therapeutic screening.

14.
Nat Commun ; 9(1): 3999, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30275522

RESUMO

The lung is a critical prophylaxis target for clinically important infectious agents, including human respiratory syncytial virus (RSV) and influenza. Here, we develop a modular, synthetic mRNA-based approach to express neutralizing antibodies directly in the lung via aerosol, to prevent RSV infections. First, we express palivizumab, which reduces RSV F copies by 90.8%. Second, we express engineered, membrane-anchored palivizumab, which prevents detectable infection in transfected cells, reducing in vitro titer and in vivo RSV F copies by 99.7% and 89.6%, respectively. Finally, we express an anchored or secreted high-affinity, anti-RSV F, camelid antibody (RSV aVHH and sVHH). We demonstrate that RSV aVHH, but not RSV sVHH, significantly inhibits RSV 7 days post transfection, and we show that RSV aVHH is present in the lung for at least 28 days. Overall, our data suggests that expressing membrane-anchored broadly neutralizing antibodies in the lungs could potentially be a promising pulmonary prophylaxis approach.


Assuntos
Anticorpos Neutralizantes/imunologia , Antivirais/administração & dosagem , Palivizumab/imunologia , RNA Mensageiro/administração & dosagem , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vírus Sincicial Respiratório Humano/imunologia , Animais , Anticorpos Neutralizantes/genética , Anticorpos Neutralizantes/metabolismo , Antivirais/imunologia , Linhagem Celular , Membrana Celular/metabolismo , Feminino , Humanos , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Palivizumab/genética , Palivizumab/metabolismo , Profilaxia Pré-Exposição , RNA Mensageiro/genética , RNA Mensageiro/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/virologia , Proteínas Virais de Fusão/imunologia
15.
Bioconjug Chem ; 29(9): 3072-3083, 2018 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-30067354

RESUMO

In vitro transcribed (IVT) mRNA is an appealing platform for next generation vaccines, as it can be manufactured rapidly at large scale to meet emerging pathogens. However, its performance as a robust vaccine is strengthened by supplemental immune stimulation, which is typically provided by adjuvant formulations that facilitate delivery and stimulate immune responses. Here, we present a strategy for increasing translation of a model IVT mRNA vaccine while simultaneously modulating its immune-stimulatory properties in a programmable fashion, without relying on delivery vehicle formulations. Substitution of uridine with the modified base N1-methylpseudouridine reduces the intrinsic immune stimulation of the IVT mRNA and enhances antigen translation. Tethering adjuvants to naked IVT mRNA through antisense nucleotides boosts the immunostimulatory properties of adjuvants in vitro, without impairing transgene production or adjuvant activity. In vivo, intramuscular injection of tethered IVT mRNA-TLR7 agonists leads to enhanced local immune responses, and to antigen-specific cell-mediated and humoral responses. We believe this system represents a potential platform compatible with any adjuvant of interest to enable specific programmable stimulation of immune responses.


Assuntos
Imunidade Inata/efeitos dos fármacos , RNA Mensageiro/genética , Vacinas Sintéticas/farmacologia , Animais , Formação de Anticorpos , Imunidade Celular , Injeções Intramusculares , Camundongos , Células RAW 264.7 , Transcrição Gênica , Vacinas Sintéticas/administração & dosagem
16.
Biomaterials ; 159: 189-203, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29331806

RESUMO

The translational efficiency of an in vitro transcribed (IVT) mRNA was measured upon delivery to primary skeletal muscle cells and to a mouse model system, towards the development of a predictive in vitro assay for the screening and validation of intramuscular mRNA-based vaccines. When IVT mRNA was delivered either naked or complexed with novel aminoglycoside-based delivery vehicles, significant differences in protein expression in vitro and in vivo were observed. We hypothesized that this previously anticipated discrepancy was due to differences in the mechanism of IVT mRNA endosomal entry and release following delivery. To address this, IVT mRNA was fluorescently labeled prior to delivery, to visualize its distribution. Colocalization with endosomal markers indicated that different entry pathways were utilized in vivo and in vitro, depending on the delivery vehicle, resulting in variations in protein expression levels. Since extracellular matrix stiffness (ECM) influences mRNA entry, trafficking and release, the effect of mechanotransduction on mRNA expression was investigated in vitro upon delivery of IVT mRNA alone, and complexed with delivery vehicles to skeletal muscle cells grown on ∼10 kPa hydrogels. This in vitro hydrogel model more accurately recapitulated the results obtained in vivo upon IM injection, indicating that this approach may assist in the characterization of mRNA based vaccines.


Assuntos
Mecanotransdução Celular/fisiologia , Músculo Esquelético/metabolismo , RNA Mensageiro/metabolismo , Animais , Linhagem Celular , Endossomos/química , Matriz Extracelular/química , Feminino , Citometria de Fluxo , Células HeLa , Humanos , Hidrogéis/química , Camundongos , Camundongos Endogâmicos BALB C , Nanopartículas/química
17.
Nat Commun ; 8(1): 667, 2017 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-28939853

RESUMO

The human respiratory syncytial virus G protein plays an important role in the entry and assembly of filamentous virions. Here, we report the use of fluorescently labeled soybean agglutinin to selectively label the respiratory syncytial virus G protein in living cells without disrupting respiratory syncytial virus infectivity or filament formation and allowing for interrogations of respiratory syncytial virus virion assembly. Using this approach, we discovered that plasma membrane-bound respiratory syncytial virus G rapidly recycles from the membrane via clathrin-mediated endocytosis. This event is then followed by the dynamic formation of filamentous and branched respiratory syncytial virus particles, and assembly with genomic ribonucleoproteins and caveolae-associated vesicles prior to re-insertion into the plasma membrane. We demonstrate that these processes are halted by the disruption of microtubules and inhibition of molecular motors. Collectively, our results show that for respiratory syncytial virus assembly, viral filaments are produced and loaded with genomic RNA prior to insertion into the plasma membrane.Assembly of filamentous RSV particles is incompletely understood due to a lack of techniques suitable for live-cell imaging. Here Vanover et al. use labeled soybean agglutinin to selectively label RSV G protein and show how filamentous RSV assembly, initiated in the cytoplasm, uses G protein recycled from the plasma membrane.


Assuntos
Membrana Celular/metabolismo , RNA Viral/metabolismo , Vírus Sincicial Respiratório Humano/genética , Proteínas Virais de Fusão/metabolismo , Animais , Caveolinas/metabolismo , Chlorocebus aethiops , Clatrina/metabolismo , Endocitose/fisiologia , Humanos , Microtúbulos/metabolismo , Imagem Molecular/métodos , Lectinas de Plantas/química , Lectinas de Plantas/metabolismo , Vírus Sincicial Respiratório Humano/patogenicidade , Vírus Sincicial Respiratório Humano/fisiologia , Ribonucleoproteínas/metabolismo , Proteínas de Soja/química , Proteínas de Soja/metabolismo , Células Vero , Replicação Viral
18.
Nucleic Acids Res ; 45(12): e113, 2017 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-28449134

RESUMO

The use of synthetic messenger ribonucleic acid (mRNA) to express specific proteins is a highly promising therapeutic and vaccine approach that avoids many safety issues associated with viral or DNA-based systems. However, in order to optimize mRNA designs and delivery, technology advancements are required to study fundamental mechanisms of mRNA uptake and localization at the single-cell and tissue level. Here, we present a single RNA sensitive fluorescent labeling method which allows us to label and visualize synthetic mRNA without significantly affecting function. This approach enabled single cell characterization of mRNA uptake and release kinetics from endocytic compartments, the measurement of mRNA/protein correlations, and motivated the investigation of mRNA induced cellular stress, all important mechanisms influencing protein production. In addition, we demonstrated this approach can facilitate near-infrared imaging of mRNA localization in vivo and in ex-vivo tissue sections, which will facilitate mRNA trafficking studies in pre-clinical models. Overall, we demonstrate the ability to study fundamental mechanisms necessary to optimize delivery and therapeutic strategies, in order to design the next generation of novel mRNA therapeutics and vaccines.


Assuntos
Endossomos/metabolismo , Imagem Óptica/métodos , RNA Mensageiro/farmacocinética , Análise de Célula Única/métodos , Coloração e Rotulagem/métodos , Animais , Transporte Biológico , Carbocianinas/química , Linhagem Celular , Grânulos Citoplasmáticos/metabolismo , Grânulos Citoplasmáticos/ultraestrutura , Fibroblastos/citologia , Fibroblastos/metabolismo , Corantes Fluorescentes/química , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Humanos , Raios Infravermelhos , Injeções Intramusculares , Camundongos , Sondas Moleculares/química , Hibridização de Ácido Nucleico , RNA Mensageiro/química , RNA Mensageiro/genética
19.
Science ; 354(6309): 197-202, 2016 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-27738167

RESUMO

Antiretroviral drug therapy (ART) effectively suppresses replication of both the immunodeficiency viruses, human (HIV) and simian (SIV); however, virus rebounds soon after ART is withdrawn. SIV-infected monkeys were treated with a 90-day course of ART initiated at 5 weeks post infection followed at 9 weeks post infection by infusions of a primatized monoclonal antibody against the α4ß7 integrin administered every 3 weeks until week 32. These animals subsequently maintained low to undetectable viral loads and normal CD4+ T cell counts in plasma and gastrointestinal tissues for more than 9 months, even after all treatment was withdrawn. This combination therapy allows macaques to effectively control viremia and reconstitute their immune systems without a need for further therapy.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Imunização Passiva/métodos , Integrina alfa4/imunologia , Cadeias beta de Integrinas/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/terapia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/imunologia , Viremia/terapia , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais Humanizados/administração & dosagem , Contagem de Linfócito CD4 , Linfócitos T CD4-Positivos/imunologia , Terapia Combinada , Citocinas/sangue , Modelos Animais de Doenças , Feminino , Trato Gastrointestinal/imunologia , Infusões Intravenosas , Células Matadoras Naturais/imunologia , Macaca mulatta , Masculino , Glicoproteínas de Membrana/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/sangue , Síndrome de Imunodeficiência Adquirida dos Símios/tratamento farmacológico , Vírus da Imunodeficiência Símia/isolamento & purificação , Subpopulações de Linfócitos T/imunologia , Tretinoína/sangue , Proteínas do Envelope Viral/imunologia , Carga Viral/imunologia , Viremia/sangue , Viremia/tratamento farmacológico , Viremia/virologia
20.
Methods ; 98: 91-98, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-26875782

RESUMO

Viruses represent an important class of pathogens that have had an enormous impact on the health of the human race. They are extraordinarily diverse; viral particles can range in size from ∼80nm to ∼10µm in length, and contain genomes with RNA or DNA strands. Regardless of their genome type, RNA species are frequently generated as a part of their replication process, and for viruses with RNA genomes, their loading into the virion represents a critical step in the creation of infectious particles. RNA imaging tools represent a powerful approach to gain insight into fundamental viral processes, including virus entry, replication, and virion assembly. Imaging viral processes in live cells is critical due to both the heterogeneity of these processes on a per cell basis, and the inherent dynamics of these processes. There are a number of methods for labeling RNA in live cells; we'll introduce the myriad of methods and then focus on one approach for labeling viral RNA, using multiply-labeled tetravalent RNA imaging probes (MTRIPs), which do not require engineering of the target RNAs. We feel this approach is advantageous given many viral genomes may not tolerate large nucleotide insertions into their sequences.


Assuntos
Regulação Viral da Expressão Gênica , HIV-1/química , Imagem Molecular/métodos , RNA Mensageiro/química , RNA Viral/química , Vírus Sinciciais Respiratórios/química , Coloração e Rotulagem/métodos , Animais , Chlorocebus aethiops , Corantes Fluorescentes/química , HIV-1/genética , HIV-1/metabolismo , Células Hep G2 , Humanos , Oligonucleotídeos/química , Sondas RNA/química , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Viral/genética , RNA Viral/metabolismo , Vírus Sinciciais Respiratórios/genética , Vírus Sinciciais Respiratórios/metabolismo , Células Vero , Vírion/química , Vírion/genética , Vírion/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA