Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
J Biol Chem ; 299(6): 104749, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37100284

RESUMEN

The recent SARS-CoV-2 and mpox outbreaks have highlighted the need to expand our arsenal of broad-spectrum antiviral agents for future pandemic preparedness. Host-directed antivirals are an important tool to accomplish this as they typically offer protection against a broader range of viruses than direct-acting antivirals and have a lower susceptibility to viral mutations that cause drug resistance. In this study, we investigate the exchange protein activated by cAMP (EPAC) as a target for broad-spectrum antiviral therapy. We find that the EPAC-selective inhibitor, ESI-09, provides robust protection against a variety of viruses, including SARS-CoV-2 and Vaccinia (VACV)-an orthopox virus from the same family as mpox. We show, using a series of immunofluorescence experiments, that ESI-09 remodels the actin cytoskeleton through Rac1/Cdc42 GTPases and the Arp2/3 complex, impairing internalization of viruses that use clathrin-mediated endocytosis (e.g. VSV) or micropinocytosis (e.g. VACV). Additionally, we find that ESI-09 disrupts syncytia formation and inhibits cell-to-cell transmission of viruses such as measles and VACV. When administered to immune-deficient mice in an intranasal challenge model, ESI-09 protects mice from lethal doses of VACV and prevents formation of pox lesions. Altogether, our finding shows that EPAC antagonists such as ESI-09 are promising candidates for broad-spectrum antiviral therapy that can aid in the fight against ongoing and future viral outbreaks.


Asunto(s)
Antivirales , COVID-19 , Mpox , Vaccinia , Animales , Ratones , Antivirales/farmacología , Mpox/tratamiento farmacológico , SARS-CoV-2/efectos de los fármacos , Vaccinia/tratamiento farmacológico , Virus Vaccinia/efectos de los fármacos
2.
Mol Ther ; 30(9): 2998-3016, 2022 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-35526097

RESUMEN

We established a split nanoluciferase complementation assay to rapidly screen for inhibitors that interfere with binding of the receptor binding domain (RBD) of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike glycoprotein with its target receptor, angiotensin-converting enzyme 2 (ACE2). After a screen of 1,200 US Food and Drug Administration (FDA)-approved compounds, we identified bifonazole, an imidazole-based antifungal agent, as a competitive inhibitor of RBD-ACE2 binding. Mechanistically, bifonazole binds ACE2 around residue K353, which prevents association with the RBD, affecting entry and replication of spike-pseudotyped viruses as well as native SARS-CoV-2 and its variants of concern (VOCs). Intranasal administration of bifonazole reduces lethality in K18-hACE2 mice challenged with vesicular stomatitis virus (VSV)-spike by 40%, with a similar benefit after live SARS-CoV-2 challenge. Our screen identified an antiviral agent that is effective against SARS-CoV-2 and VOCs such as Omicron that employ the same receptor to infect cells and therefore has high potential to be repurposed to control, treat, or prevent coronavirus disease 2019 (COVID-19).


Asunto(s)
Antivirales , Tratamiento Farmacológico de COVID-19 , Imidazoles , SARS-CoV-2 , Enzima Convertidora de Angiotensina 2/antagonistas & inhibidores , Animales , Antivirales/farmacología , Imidazoles/farmacología , Ratones , Unión Proteica , SARS-CoV-2/efectos de los fármacos , Glicoproteína de la Espiga del Coronavirus/química , Estados Unidos , United States Food and Drug Administration
3.
Mol Ther ; 30(5): 1885-1896, 2022 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-34687845

RESUMEN

The coronavirus disease 2019 (COVID-19) pandemic requires the continued development of safe, long-lasting, and efficacious vaccines for preventive responses to major outbreaks around the world, and especially in isolated and developing countries. To combat severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), we characterize a temperature-stable vaccine candidate (TOH-Vac1) that uses a replication-competent, attenuated vaccinia virus as a vector to express a membrane-tethered spike receptor binding domain (RBD) antigen. We evaluate the effects of dose escalation and administration routes on vaccine safety, efficacy, and immunogenicity in animal models. Our vaccine induces high levels of SARS-CoV-2 neutralizing antibodies and favorable T cell responses, while maintaining an optimal safety profile in mice and cynomolgus macaques. We demonstrate robust immune responses and protective immunity against SARS-CoV-2 variants after only a single dose. Together, these findings support further development of our novel and versatile vaccine platform as an alternative or complementary approach to current vaccines.


Asunto(s)
COVID-19 , Vacunas , Animales , Ratones , Anticuerpos Neutralizantes , Anticuerpos Antivirales , COVID-19/prevención & control , Vacunas contra la COVID-19 , Inmunidad , SARS-CoV-2/genética , Glicoproteína de la Espiga del Coronavirus , Linfocitos T
4.
Mol Ther ; 29(6): 1984-2000, 2021 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-33578036

RESUMEN

The ongoing COVID-19 pandemic has highlighted the immediate need for the development of antiviral therapeutics targeting different stages of the SARS-CoV-2 life cycle. We developed a bioluminescence-based bioreporter to interrogate the interaction between the SARS-CoV-2 viral spike (S) protein and its host entry receptor, angiotensin-converting enzyme 2 (ACE2). The bioreporter assay is based on a nanoluciferase complementation reporter, composed of two subunits, large BiT and small BiT, fused to the S receptor-binding domain (RBD) of the SARS-CoV-2 S protein and ACE2 ectodomain, respectively. Using this bioreporter, we uncovered critical host and viral determinants of the interaction, including a role for glycosylation of asparagine residues within the RBD in mediating successful viral entry. We also demonstrate the importance of N-linked glycosylation to the RBD's antigenicity and immunogenicity. Our study demonstrates the versatility of our bioreporter in mapping key residues mediating viral entry as well as screening inhibitors of the ACE2-RBD interaction. Our findings point toward targeting RBD glycosylation for therapeutic and vaccine strategies against SARS-CoV-2.


Asunto(s)
Enzima Convertidora de Angiotensina 2/química , Anticuerpos Neutralizantes/farmacología , Bioensayo , Lectinas/farmacología , Receptores Virales/química , Glicoproteína de la Espiga del Coronavirus/química , Enzima Convertidora de Angiotensina 2/antagonistas & inhibidores , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/inmunología , Asparagina/química , Asparagina/metabolismo , Sitios de Unión , COVID-19/diagnóstico , COVID-19/inmunología , COVID-19/virología , Genes Reporteros , Glicosilación/efectos de los fármacos , Células HEK293 , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/genética , Humanos , Luciferasas/genética , Luciferasas/metabolismo , Mediciones Luminiscentes , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Estructura Secundaria de Proteína , Receptores Virales/antagonistas & inhibidores , Receptores Virales/genética , Receptores Virales/inmunología , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/crecimiento & desarrollo , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/antagonistas & inhibidores , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/inmunología , Internalización del Virus/efectos de los fármacos , Tratamiento Farmacológico de COVID-19
5.
Int J Mol Sci ; 22(5)2021 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-33668756

RESUMEN

Despite sequence similarity to SARS-CoV-1, SARS-CoV-2 has demonstrated greater widespread virulence and unique challenges to researchers aiming to study its pathogenicity in humans. The interaction of the viral receptor binding domain (RBD) with its main host cell receptor, angiotensin-converting enzyme 2 (ACE2), has emerged as a critical focal point for the development of anti-viral therapeutics and vaccines. In this study, we selectively identify and characterize the impact of mutating certain amino acid residues in the RBD of SARS-CoV-2 and in ACE2, by utilizing our recently developed NanoBiT technology-based biosensor as well as pseudotyped-virus infectivity assays. Specifically, we examine the mutational effects on RBD-ACE2 binding ability, efficacy of competitive inhibitors, as well as neutralizing antibody activity. We also look at the implications the mutations may have on virus transmissibility, host susceptibility, and the virus transmission path to humans. These critical determinants of virus-host interactions may provide more effective targets for ongoing vaccines, drug development, and potentially pave the way for determining the genetic variation underlying disease severity.


Asunto(s)
Enzima Convertidora de Angiotensina 2/química , Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19/metabolismo , COVID-19/virología , SARS-CoV-2/metabolismo , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/metabolismo , Secuencia de Aminoácidos , Enzima Convertidora de Angiotensina 2/genética , Anticuerpos Neutralizantes/inmunología , Antivirales/farmacología , Sitios de Unión , COVID-19/inmunología , Células HEK293 , Interacciones Microbiota-Huesped , Humanos , Modelos Moleculares , Mutación , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Receptores Virales/química , Receptores Virales/metabolismo , SARS-CoV-2/efectos de los fármacos , Alineación de Secuencia , Tratamiento Farmacológico de COVID-19
6.
J Biol Chem ; 294(51): 19785-19794, 2019 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-31694919

RESUMEN

Upon immune recognition of viruses, the mammalian innate immune response activates a complex signal transduction network to combat infection. This activation requires phosphorylation of key transcription factors regulating IFN production and signaling, including IFN regulatory factor 3 (IRF3) and STAT1. The mechanisms regulating these STAT1 and IRF3 phosphorylation events remain unclear. Here, using human and mouse cell lines along with gene microarrays, quantitative RT-PCR, viral infection and plaque assays, and reporter gene assays, we demonstrate that a microRNA cluster conserved among bilaterian animals, encoding miR-96, miR-182, and miR-183, regulates IFN signaling. In particular, we observed that the miR-183 cluster promotes IFN production and signaling, mediated by enhancing IRF3 and STAT1 phosphorylation. We also found that the miR-183 cluster activates the IFN pathway and inhibits vesicular stomatitis virus infection by directly targeting several negative regulators of IRF3 and STAT1 activities, including protein phosphatase 2A (PPP2CA) and tripartite motif-containing 27 (TRIM27). Overall, our work reveals an important role of the evolutionarily conserved miR-183 cluster in the regulation of mammalian innate immunity.


Asunto(s)
Inmunidad Innata , Factor 3 Regulador del Interferón/metabolismo , MicroARNs/metabolismo , Familia de Multigenes , Factor de Transcripción STAT1/metabolismo , Células A549 , Animales , Fibroblastos/inmunología , Fibroblastos/virología , Genes Reporteros , Células HEK293 , Células Hep G2 , Humanos , Interferones/inmunología , Células MCF-7 , Macrófagos/inmunología , Macrófagos/virología , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosforilación , Transducción de Señal , Replicación Viral
7.
Stem Cells ; 37(6): 716-723, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30875126

RESUMEN

Cancer stem cells (CSCs) have the capacity to self-renew and differentiate to give rise to heterogenous cancer cell lineages in solid tumors. These CSC populations are associated with metastasis, tumor relapse, and resistance to conventional anticancer therapies. Here, we focus on the use of oncolytic viruses (OVs) to target CSCs as well as the OV-driven interferon production in the tumor microenvironment (TME) that can repress CSC properties. We explore the ability of OVs to deliver combinations of immune-modulating therapeutic transgenes, such as immune checkpoint inhibitor antibodies. In particular, we highlight the advantages of virally encoded bi-specific T cell engagers (BiTEs) to not only target cell-surface markers on CSCs, but also tumor-associated antigens on contributing components of the surrounding TME and other cancer cells. We also highlight the crucial role of combination anticancer treatments, evidenced by synergy of OV-delivered BiTEs and chimeric-antigen receptor T cell therapy. Stem Cells 2019;37:716-723.


Asunto(s)
Antineoplásicos Inmunológicos/uso terapéutico , Neoplasias/terapia , Células Madre Neoplásicas/virología , Viroterapia Oncolítica/métodos , Virus Oncolíticos/inmunología , Microambiente Tumoral/inmunología , Anticuerpos/uso terapéutico , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Diferenciación Celular , Terapia Combinada/métodos , Humanos , Inmunoterapia Adoptiva/métodos , Terapia Molecular Dirigida/métodos , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/patología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/inmunología , Células Madre Neoplásicas/patología , Virus Oncolíticos/genética , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/inmunología , Linfocitos T/patología , Linfocitos T/virología , Transgenes , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/genética
8.
Arterioscler Thromb Vasc Biol ; 37(6): 1058-1067, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28428217

RESUMEN

OBJECTIVE: Defective autophagy in macrophages leads to pathological processes that contribute to atherosclerosis, including impaired cholesterol metabolism and defective efferocytosis. Autophagy promotes the degradation of cytoplasmic components in lysosomes and plays a key role in the catabolism of stored lipids to maintain cellular homeostasis. microRNA-33 (miR-33) is a post-transcriptional regulator of genes involved in cholesterol homeostasis, yet the complete mechanisms by which miR-33 controls lipid metabolism are unknown. We investigated whether miR-33 targeting of autophagy contributes to its regulation of cholesterol homeostasis and atherogenesis. APPROACH AND RESULTS: Using coherent anti-Stokes Raman scattering microscopy, we show that miR-33 drives lipid droplet accumulation in macrophages, suggesting decreased lipolysis. Inhibition of neutral and lysosomal hydrolysis pathways revealed that miR-33 reduced cholesterol mobilization by a lysosomal-dependent mechanism, implicating repression of autophagy. Indeed, we show that miR-33 targets key autophagy regulators and effectors in macrophages to reduce lipid droplet catabolism, an essential process to generate free cholesterol for efflux. Notably, miR-33 regulation of autophagy lies upstream of its known effects on ABCA1 (ATP-binding cassette transporter A1)-dependent cholesterol efflux, as miR-33 inhibitors fail to increase efflux upon genetic or chemical inhibition of autophagy. Furthermore, we find that miR-33 inhibits apoptotic cell clearance via an autophagy-dependent mechanism. Macrophages treated with anti-miR-33 show increased efferocytosis, lysosomal biogenesis, and degradation of apoptotic material. Finally, we show that treating atherosclerotic Ldlr-/- mice with anti-miR-33 restores defective autophagy in macrophage foam cells and plaques and promotes apoptotic cell clearance to reduce plaque necrosis. CONCLUSIONS: Collectively, these data provide insight into the mechanisms by which miR-33 regulates cellular cholesterol homeostasis and atherosclerosis.


Asunto(s)
Aterosclerosis/metabolismo , Autofagia , Macrófagos Peritoneales/metabolismo , MicroARNs/metabolismo , Animales , Aterosclerosis/genética , Aterosclerosis/patología , Proteína 5 Relacionada con la Autofagia/deficiencia , Proteína 5 Relacionada con la Autofagia/genética , Colesterol/metabolismo , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Humanos , Células Jurkat , Gotas Lipídicas/metabolismo , Lisosomas/metabolismo , Macrófagos Peritoneales/patología , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/genética , Necrosis , Fenotipo , Placa Aterosclerótica , Receptores de LDL/deficiencia , Receptores de LDL/genética , Transducción de Señal , Transfección
9.
Nat Chem Biol ; 11(12): 988-93, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26479438

RESUMEN

Immune regulation of cellular metabolism can be responsible for successful responses to invading pathogens. Viruses alter their hosts' cellular metabolism to facilitate infection. Conversely, the innate antiviral responses of mammalian cells target these metabolic pathways to restrict viral propagation. We identified miR-130b and miR-185 as hepatic microRNAs (miRNAs) whose expression is stimulated by 25-hydroxycholesterol (25-HC), an antiviral oxysterol secreted by interferon-stimulated macrophages and dendritic cells, during hepatitis C virus (HCV) infection. However, 25-HC only directly stimulated miR-185 expression, whereas HCV regulated miR-130b expression. Independently, miR-130b and miR-185 inhibited HCV infection. In particular, miR-185 significantly restricted host metabolic pathways crucial to the HCV life cycle. Interestingly, HCV infection decreased miR-185 and miR-130b levels to promote lipid accumulation and counteract 25-HC's antiviral effect. Furthermore, miR-185 can inhibit other viruses through the regulation of immunometabolic pathways. These data establish these microRNAs as a key link between innate defenses and metabolism in the liver.


Asunto(s)
Hepatitis C/inmunología , Hepatitis C/metabolismo , Hígado/inmunología , Hígado/metabolismo , MicroARNs/metabolismo , Antivirales/metabolismo , Antivirales/farmacología , Línea Celular , Hepacivirus/efectos de los fármacos , Hepatitis C/tratamiento farmacológico , Humanos , Hidroxicolesteroles/farmacología , Hígado/efectos de los fármacos , Hígado/virología , MicroARNs/genética , Conformación Molecular
10.
Circ Res ; 117(3): 266-78, 2015 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-26002865

RESUMEN

RATIONALE: Therapeutically targeting macrophage reverse cholesterol transport is a promising approach to treat atherosclerosis. Macrophage energy metabolism can significantly influence macrophage phenotype, but how this is controlled in foam cells is not known. Bioinformatic pathway analysis predicts that miR-33 represses a cluster of genes controlling cellular energy metabolism that may be important in macrophage cholesterol efflux. OBJECTIVE: We hypothesized that cellular energy status can influence cholesterol efflux from macrophages, and that miR-33 reduces cholesterol efflux via repression of mitochondrial energy metabolism pathways. METHODS AND RESULTS: In this study, we demonstrated that macrophage cholesterol efflux is regulated by mitochondrial ATP production, and that miR-33 controls a network of genes that synchronize mitochondrial function. Inhibition of mitochondrial ATP synthase markedly reduces macrophage cholesterol efflux capacity, and anti-miR33 required fully functional mitochondria to enhance ABCA1-mediated cholesterol efflux. Specifically, anti-miR33 derepressed the novel target genes PGC-1α, PDK4, and SLC25A25 and boosted mitochondrial respiration and production of ATP. Treatment of atherosclerotic Apoe(-/-) mice with anti-miR33 oligonucleotides reduced aortic sinus lesion area compared with controls, despite no changes in high-density lipoprotein cholesterol or other circulating lipids. Expression of miR-33a/b was markedly increased in human carotid atherosclerotic plaques compared with normal arteries, and there was a concomitant decrease in mitochondrial regulatory genes PGC-1α, SLC25A25, NRF1, and TFAM, suggesting these genes are associated with advanced atherosclerosis in humans. CONCLUSIONS: This study demonstrates that anti-miR33 therapy derepresses genes that enhance mitochondrial respiration and ATP production, which in conjunction with increased ABCA1 expression, works to promote macrophage cholesterol efflux and reduce atherosclerosis.


Asunto(s)
Adenosina Trifosfato/biosíntesis , Aterosclerosis/metabolismo , Colesterol/metabolismo , Macrófagos Peritoneales/metabolismo , Macrófagos/metabolismo , MicroARNs/antagonistas & inhibidores , Mitocondrias/metabolismo , Oligonucleótidos Antisentido/uso terapéutico , Sistemas de Transporte de Aminoácidos Acídicos/biosíntesis , Sistemas de Transporte de Aminoácidos Acídicos/genética , Animales , Apolipoproteínas E/deficiencia , Aterosclerosis/genética , Aterosclerosis/terapia , Secuencia de Bases , Proteínas de Unión al Calcio/biosíntesis , Proteínas de Unión al Calcio/genética , Línea Celular , Regulación de la Expresión Génica/efectos de los fármacos , Terapia Genética , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , Proteínas de Transporte de Membrana Mitocondrial , Oligonucleótidos Antisentido/farmacología , Proteínas Serina-Treonina Quinasas/genética , Alineación de Secuencia , Homología de Secuencia de Ácido Nucleico , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética
11.
Hepatology ; 59(1): 98-108, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23897856

RESUMEN

UNLABELLED: MicroRNAs (miRNAs) are small RNAs that posttranscriptionally regulate gene expression. Their aberrant expression is commonly linked with diseased states, including hepatitis C virus (HCV) infection. Herein, we demonstrate that HCV replication induces the expression of miR-27 in cell culture and in vivo HCV infectious models. Overexpression of the HCV proteins core and NS4B independently activates miR-27 expression. Furthermore, we establish that miR-27 overexpression in hepatocytes results in larger and more abundant lipid droplets, as observed by coherent anti-Stokes Raman scattering (CARS) microscopy. This hepatic lipid droplet accumulation coincides with miR-27b's repression of peroxisome proliferator-activated receptor (PPAR)-α and angiopoietin-like protein 3 (ANGPTL3), known regulators of triglyceride homeostasis. We further demonstrate that treatment with a PPAR-α agonist, bezafibrate, is able to reverse the miR-27b-induced lipid accumulation in Huh7 cells. This miR-27b-mediated repression of PPAR-α signaling represents a novel mechanism of HCV-induced hepatic steatosis. This link was further demonstrated in vivo through the correlation between miR-27b expression levels and hepatic lipid accumulation in HCV-infected SCID-beige/Alb-uPa mice. CONCLUSION: Collectively, our results highlight HCV's up-regulation of miR-27 expression as a novel mechanism contributing to the development of hepatic steatosis.


Asunto(s)
Hígado Graso/etiología , Hepacivirus/fisiología , Hepatitis C/complicaciones , MicroARNs/metabolismo , Animales , Bezafibrato , Línea Celular Tumoral , Hepatitis C/metabolismo , Hepatitis C/virología , Homeostasis , Humanos , Metabolismo de los Lípidos , Hígado/metabolismo , Ratones , Ratones SCID , PPAR alfa/agonistas , Regulación hacia Arriba
12.
Can J Microbiol ; 61(8): 521-30, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26182401

RESUMEN

The metabolic interplay between hosts and viruses plays a crucial role in determining the outcome of viral infection. Viruses reorchestrate the host's primary metabolic gene networks, including genes associated with mevalonate and isoprenoid synthesis, to acquire the necessary energy and structural components for their viral life cycles. Recent work has demonstrated that the interferon-mediated antiviral response suppresses the sterol pathway through production of a signalling molecule, 25-hydroxycholesterol (25HC). This oxysterol has been shown to exert multiple effects, both through incorporation into host cellular membranes as well as through transcriptional control. Herein, we summarize our current understanding of the multifunctional roles of 25HC in the mammalian innate antiviral response.


Asunto(s)
Antivirales/farmacología , Hidroxicolesteroles/farmacología , Inmunidad Innata/efectos de los fármacos , Inmunidad Adaptativa , Animales , Membrana Celular/efectos de los fármacos , Homeostasis , Humanos , Esteroide Hidroxilasas/genética
13.
Biochem Biophys Res Commun ; 441(2): 447-52, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24161736

RESUMEN

Human hepatocytes constitutively express the lipid droplet (LD) associated protein cell death-inducing DFFA-like effector B (CIDEB). CIDEB mediates LD fusion, as well as very-low-density lipoprotein (VLDL) maturation. However, there are limited cell culture models readily available to study CIDEB's role in these biological processes, as hepatoma cell lines express negligible levels of CIDEB. Recent work has highlighted the ability of human serum to differentiate hepatoma cells. Herein, we demonstrate that culturing Huh7.5 cells in media supplemented with human serum activates CIDEB expression. This activation occurs through the induced expression of PGC-1α, a positive transcriptional regulator of CIDEB. Coherent anti-Stokes Raman scattering (CARS) microscopy revealed a correlation between CIDEB levels and LD size in human serum treated Huh7.5 cells. Human serum treatment also resulted in a rapid decrease in the levels of adipose differentiation-related protein (ADRP). Furthermore, individual overexpression of CIDEB was sufficient to down-regulate ADRP protein levels. siRNA knockdown of CIDEB revealed that the human serum mediated increase in LD size was CIDEB-dependent. Overall, our work highlights CIDEB's role in LD fusion, and presents a new model system to study the PGC-1α/CIDEB pathway's role in LD dynamics and the VLDL pathway.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/fisiología , Hepatocitos/metabolismo , Lipoproteínas VLDL/metabolismo , Suero/fisiología , Proteínas Reguladoras de la Apoptosis/genética , Diferenciación Celular , Línea Celular Tumoral , Técnicas de Silenciamiento del Gen , Hepatocitos/citología , Humanos , Cuerpos de Inclusión , Modelos Biológicos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , ARN Interferente Pequeño/genética , Factores de Transcripción/metabolismo
14.
Nat Chem Biol ; 7(3): 137-45, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21321552

RESUMEN

Cellular biomolecules contain unique molecular vibrations that can be visualized by coherent anti-Stokes Raman scattering (CARS) microscopy without the need for labels. Here we review the application of CARS microscopy for label-free imaging of cells and tissues using the natural vibrational contrast that arises from biomolecules like lipids as well as for imaging of exogenously added probes or drugs. High-resolution CARS microscopy combined with multimodal imaging has allowed for dynamic monitoring of cellular processes such as lipid metabolism and storage, the movement of organelles, adipogenesis and host-pathogen interactions and can also be used to track molecules within cells and tissues. The CARS imaging modality provides a unique tool for biological chemists to elucidate the state of a cellular environment without perturbing it and to perceive the functional effects of added molecules.


Asunto(s)
Rastreo Celular/métodos , Medios de Contraste , Imagen Molecular/métodos , Espectrometría Raman/métodos , Metabolismo de los Lípidos , Vibración
15.
Mol Ther Methods Clin Dev ; 31: 101110, 2023 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-37822719

RESUMEN

SARS-CoV-2, the etiological agent behind the coronavirus disease 2019 (COVID-19) pandemic, has continued to mutate and create new variants with increased resistance against the WHO-approved spike-based vaccines. With a significant portion of the worldwide population still unvaccinated and with waning immunity against newly emerging variants, there is a pressing need to develop novel vaccines that provide broader and longer-lasting protection. To generate broader protective immunity against COVID-19, we developed our second-generation vaccinia virus-based COVID-19 vaccine, TOH-VAC-2, encoded with modified versions of the spike (S) and nucleocapsid (N) proteins as well as a unique poly-epitope antigen that contains immunodominant T cell epitopes from seven different SARS-CoV-2 proteins. We show that the poly-epitope antigen restimulates T cells from the PBMCs of individuals formerly infected with SARS-CoV-2. In mice, TOH-VAC-2 vaccination produces high titers of S- and N-specific antibodies and generates robust T cell immunity against S, N, and poly-epitope antigens. The immunity generated from TOH-VAC-2 is also capable of protecting mice from heterologous challenge with recombinant VSV viruses that express the same SARS-CoV-2 antigens. Altogether, these findings demonstrate the effectiveness of our versatile vaccine platform as an alternative or complementary approach to current vaccines.

16.
Nat Commun ; 14(1): 3035, 2023 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-37236967

RESUMEN

The large coding potential of vaccinia virus (VV) vectors is a defining feature. However, limited regulatory switches are available to control viral replication as well as timing and dosing of transgene expression in order to facilitate safe and efficacious payload delivery. Herein, we adapt drug-controlled gene switches to enable control of virally encoded transgene expression, including systems controlled by the FDA-approved rapamycin and doxycycline. Using ribosome profiling to characterize viral promoter strength, we rationally design fusions of the operator element of different drug-inducible systems with VV promoters to produce synthetic promoters yielding robust inducible expression with undetectable baseline levels. We also generate chimeric synthetic promoters facilitating additional regulatory layers for VV-encoded synthetic transgene networks. The switches are applied to enable inducible expression of fusogenic proteins, dose-controlled delivery of toxic cytokines, and chemical regulation of VV replication. This toolbox enables the precise modulation of transgene circuitry in VV-vectored oncolytic virus design.


Asunto(s)
Viroterapia Oncolítica , Virus Oncolíticos , Vectores Genéticos/genética , Virus Vaccinia/genética , Virus Oncolíticos/genética , Regiones Promotoras Genéticas/genética
17.
Front Immunol ; 13: 1050250, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36713447

RESUMEN

Poxvirus vectors represent versatile modalities for engineering novel vaccines and cancer immunotherapies. In addition to their oncolytic capacity and immunogenic influence, they can be readily engineered to express multiple large transgenes. However, the integration of multiple payloads into poxvirus genomes by traditional recombination-based approaches can be highly inefficient, time-consuming and cumbersome. Herein, we describe a simple, cost-effective approach to rapidly generate and purify a poxvirus vector with multiple transgenes. By utilizing a simple, modular CRISPR/Cas9 assisted-recombinant vaccinia virus engineering (CARVE) system, we demonstrate generation of a recombinant vaccinia virus expressing three distinct transgenes at three different loci in less than 1 week. We apply CARVE to rapidly generate a novel immunogenic vaccinia virus vector, which expresses a bacterial diadenylate cyclase. This novel vector, STINGPOX, produces cyclic di-AMP, a STING agonist, which drives IFN signaling critical to the anti-tumor immune response. We demonstrate that STINGPOX can drive IFN signaling in primary human cancer tissue explants. Using an immunocompetent murine colon cancer model, we demonstrate that intratumoral administration of STINGPOX in combination with checkpoint inhibitor, anti-PD1, promotes survival post-tumour challenge. These data demonstrate the utility of CRISPR/Cas9 in the rapid arming of poxvirus vectors with therapeutic payloads to create novel immunotherapies.


Asunto(s)
Neoplasias , Poxviridae , Humanos , Animales , Ratones , Vectores Genéticos/genética , Virus Vaccinia , Poxviridae/genética , Inmunoterapia
18.
Nat Commun ; 13(1): 1898, 2022 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-35393414

RESUMEN

Recent advances in cancer therapeutics clearly demonstrate the need for innovative multiplex therapies that attack the tumour on multiple fronts. Oncolytic or "cancer-killing" viruses (OVs) represent up-and-coming multi-mechanistic immunotherapeutic drugs for the treatment of cancer. In this study, we perform an in-vitro screen based on virus-encoded artificial microRNAs (amiRNAs) and find that a unique amiRNA, herein termed amiR-4, confers a replicative advantage to the VSVΔ51 OV platform. Target validation of amiR-4 reveals ARID1A, a protein involved in chromatin remodelling, as an important player in resistance to OV replication. Virus-directed targeting of ARID1A coupled with small-molecule inhibition of the methyltransferase EZH2 leads to the synthetic lethal killing of both infected and uninfected tumour cells. The bystander killing of uninfected cells is mediated by intercellular transfer of extracellular vesicles carrying amiR-4 cargo. Altogether, our findings establish that OVs can serve as replicating vehicles for amiRNA therapeutics with the potential for combination with small molecule and immune checkpoint inhibitor therapy.


Asunto(s)
Vesículas Extracelulares , MicroARNs , Neoplasias , Viroterapia Oncolítica , Virus Oncolíticos , Humanos , MicroARNs/genética , Neoplasias/terapia , Virus Oncolíticos/genética
19.
Biochemistry ; 50(36): 7745-55, 2011 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-21819044

RESUMEN

Tombusviruses express a 19 kDa protein (p19) that, as a dimeric protein, suppresses the RNAs silencing pathway during infection by binding short-interfering RNA (siRNA) and preventing their association with the RNA-induced silencing complex (RISC). The p19 protein can bind to both endogenous and synthetic siRNAs with a high degree of size selectivity but with little sequence dependence. It also binds to other endogenous small RNAs such as microRNAs (miRNAs) but with lower affinity than to canonical siRNAs. It has become apparent, however, that miRNAs play a large role in gene regulation; their influence extends to expression and processing that affects virtually all eukaryotic processes. In order to develop new tools to study endogenous small RNAs, proteins that suppress specific miRNAs are required. Herein we describe mutational analysis of the p19 binding surface with the aim of creating p19 mutants with increased affinity for miR-122. By site-directed mutagenesis of a single residue, we describe p19 mutants with a nearly 50-fold increased affinity for miR-122 without altering the affinity for siRNA. Upon further mutational analysis of this site, we postulate that the higher affinity relies on hydrogen-bonding interactions but can be sterically hindered by residues with bulky side chains. Finally, we demonstrate the effectiveness of a mutant p19, p19-T111S, at sequestering miR-122 in human hepatoma cell lines, as compared to wild-type p19. Overall, our results suggest that p19 can be engineered to enhance its affinity toward specific small RNA molecules, particularly noncanonical miRNAs that are distinguishable based on locations of base-pair mismatches. The p19-T111S mutant also represents a new tool for the study of the function of miR-122 in post-transcriptional silencing in the human liver.


Asunto(s)
MicroARNs/química , Interferencia de ARN , Proteínas Virales/química , Sitios de Unión , Línea Celular Tumoral , Dicroismo Circular , Genoma Viral , Humanos , Enlace de Hidrógeno , MicroARNs/genética , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Nicotiana/metabolismo , Tombusvirus/genética , Tombusvirus/metabolismo , Transfección , Proteínas Virales/genética
20.
Anal Biochem ; 412(2): 165-72, 2011 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-21284927

RESUMEN

MicroRNAs (miRNAs) are endogenous posttranscriptional regulators found in all metazoa and play crucial roles in virtually all cellular processes. Their aberrant expression has been linked to several diseased states; therefore, techniques capable of sensitive and specific profiling of the miRNA milieu will have significant application in prognostics, diagnostics, and therapeutics. Here we present a method for rapid quantification of miRNA levels using p19, a tombusvirus-encoded suppressor of RNA interference with sequence-independent and size-selective affinity toward 19-bp RNA duplexes. We present a surface plasmon resonance (SPR)-based miRNA sensing method where RNA probes are immobilized on gold surfaces demonstrating p19's utility in recognition of miRNA-bound probes. This allows detection of miRNAs in the low nanomolar range. To increase the sensitivity, a bead-based enzyme immunoassay was performed, and this technique displays a lower detection limit of 1fmol and a linear dynamic range from 1pmol to 1fmol.


Asunto(s)
Pruebas de Enzimas/métodos , Genes Supresores , MicroARNs/análisis , Interferencia de ARN , Tombusvirus/genética , Proteínas Virales/genética , Adsorción , Disparidad de Par Base , Línea Celular Tumoral , Ensayo de Cambio de Movilidad Electroforética , Humanos , Inmunoensayo , Límite de Detección , Magnetismo , MicroARNs/genética , Sondas ARN/metabolismo , Resonancia por Plasmón de Superficie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA