Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 121(11): e2307801120, 2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38437539

RESUMEN

Adding a cationic helper lipid to a lipid nanoparticle (LNP) can increase lung delivery and decrease liver delivery. However, it remains unclear whether charge-dependent tropism is universal or, alternatively, whether it depends on the component that is charged. Here, we report evidence that cationic cholesterol-dependent tropism can differ from cationic helper lipid-dependent tropism. By testing how 196 LNPs delivered mRNA to 22 cell types, we found that charged cholesterols led to a different lung:liver delivery ratio than charged helper lipids. We also found that combining cationic cholesterol with a cationic helper lipid led to mRNA delivery in the heart as well as several lung cell types, including stem cell-like populations. These data highlight the utility of exploring charge-dependent LNP tropism.


Asunto(s)
Hígado , Células Madre , Corazón , Cationes , Colesterol , ARN Mensajero
2.
PLoS Pathog ; 20(7): e1012345, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38968329

RESUMEN

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.

3.
Nat Mater ; 22(3): 369-379, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36443576

RESUMEN

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.


Asunto(s)
COVID-19 , Animales , Ratones , ARN Mensajero/genética , SARS-CoV-2/genética , Polímeros/metabolismo , Pulmón , ARN/metabolismo
4.
Nano Lett ; 23(3): 993-1002, 2023 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-36701517

RESUMEN

Lipid nanoparticles (LNPs) have delivered RNA to hepatocytes in patients, underscoring the potential impact of nonliver delivery. Scientists can shift LNP tropism to the lung by adding cationic helper lipids; however, the biological response to these LNPs remains understudied. To evaluate the hypothesis that charged LNPs lead to differential cellular responses, we quantified how 137 LNPs delivered mRNA to 19 cell types in vivo. Consistent with previous studies, we observed helper lipid-dependent tropism. After identifying and individually characterizing three LNPs that targeted different tissues, we studied the in vivo transcriptomic response to these using single-cell RNA sequencing. Out of 835 potential pathways, 27 were upregulated in the lung, and of these 27, 19 were related to either RNA or protein metabolism. These data suggest that endogenous cellular RNA and protein machinery affects mRNA delivery to the lung in vivo.


Asunto(s)
Lípidos , Nanopartículas , Humanos , Liposomas/metabolismo , Hepatocitos/metabolismo , ARN Mensajero/genética , ARN Interferente Pequeño
5.
PLoS Pathog ; 17(5): e1009589, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-34003848

RESUMEN

Respiratory syncytial virus (RSV) is a major cause of respiratory disease in infants and the elderly. RSV is a non-segmented negative strand RNA virus. The viral M2-1 protein plays a key role in viral transcription, serving as an elongation factor to enable synthesis of full-length mRNAs. M2-1 contains an unusual CCCH zinc-finger motif that is conserved in the related human metapneumovirus M2-1 protein and filovirus VP30 proteins. Previous biochemical studies have suggested that RSV M2-1 might bind to specific virus RNA sequences, such as the transcription gene end signals or poly A tails, but there was no clear consensus on what RSV sequences it binds. To determine if M2-1 binds to specific RSV RNA sequences during infection, we mapped points of M2-1:RNA interactions in RSV-infected cells at 8 and 18 hours post infection using crosslinking immunoprecipitation with RNA sequencing (CLIP-Seq). This analysis revealed that M2-1 interacts specifically with positive sense RSV RNA, but not negative sense genome RNA. It also showed that M2-1 makes contacts along the length of each viral mRNA, indicating that M2-1 functions as a component of the transcriptase complex, transiently associating with nascent mRNA being extruded from the polymerase. In addition, we found that M2-1 binds specific cellular mRNAs. In contrast to the situation with RSV mRNA, M2-1 binds discrete sites within cellular mRNAs, with a preference for A/U rich sequences. These results suggest that in addition to its previously described role in transcription elongation, M2-1 might have an additional role involving cellular RNA interactions.


Asunto(s)
ARN Mensajero/metabolismo , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitial Respiratorio Humano/genética , Proteínas Virales/metabolismo , ARN Polimerasas Dirigidas por ADN/genética , ARN Polimerasas Dirigidas por ADN/metabolismo , Humanos , ARN Mensajero/genética , Proteínas de Unión al ARN , Factores de Elongación Transcripcional/genética , Factores de Elongación Transcripcional/metabolismo , Proteínas Virales/genética , Replicación Viral
6.
Nano Lett ; 22(24): 10025-10033, 2022 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-36521071

RESUMEN

Lipid nanoparticles (LNPs) have delivered therapeutic RNA to hepatocytes in humans. Adsorption of apolipoprotein E (ApoE) onto these clinical LNP-mRNA drugs has been shown to facilitate hepatocyte entry via the low-density lipoprotein receptor (LDLR). Since ApoE-LDLR trafficking is conserved in mice, non-human primates, and humans, characterizing this mechanism eased clinical transition. Recently, LNPs have delivered mRNA to non-hepatocytes in mice and non-human primates, suggesting they can target new cell types via ApoE- and LDLR-independent pathways. To test this hypothesis, we quantified how 60 LNPs delivered mRNA with cell type resolution in wild-type mice and three knockout mouse strains related to lipid trafficking: ApoE-/-, LDLR-/-, and PCSK9-/-. These data suggest that the hydrophobic tail length of diketopiperazine-based lipids can be changed to drive ApoE- and LDLR-independent delivery in vivo. More broadly, the results support the hypothesis that endogenous LNP trafficking can be tuned by modifying lipid chemistry.


Asunto(s)
Apolipoproteínas E , Lipoproteínas LDL , Nanopartículas , Animales , Ratones , Apolipoproteínas E/genética , Lipoproteínas LDL/genética , Ratones Noqueados , Nanopartículas/química , ARN Mensajero/química
7.
PLoS Pathog ; 16(10): e1008987, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33031461

RESUMEN

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.


Asunto(s)
Proteínas de la Nucleocápside/metabolismo , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitial Respiratorio Humano/fisiología , Ribonucleoproteínas/metabolismo , Proteínas Virales/metabolismo , Replicación Viral , Células A549 , Animales , Chlorocebus aethiops , Humanos , Proteínas de la Nucleocápside/genética , ARN Viral/genética , ARN Viral/metabolismo , Infecciones por Virus Sincitial Respiratorio/genética , Infecciones por Virus Sincitial Respiratorio/metabolismo , Ribonucleoproteínas/genética , Transcripción Genética , Células Vero , Proteínas Virales/genética
8.
J Immunol ; 202(2): 608-617, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30541883

RESUMEN

Therapeutic strategies based on in vitro-transcribed mRNA (IVT) are attractive because they avoid the permanent signature of genomic integration that is associated with DNA-based therapy and result in the transient production of proteins of interest. To date, IVT has mainly been used in vaccination protocols to generate immune responses to foreign Ags. In this "proof-of-principle" study, we explore a strategy of combinatorial IVT to recruit and reprogram immune effector cells to acquire divergent biological functions in mice in vivo. First, we demonstrate that synthetic mRNA encoding CCL3 is able to recruit murine monocytes in a nonprogrammed state, exhibiting neither bactericidal nor tissue-repairing properties. However, upon addition of either Ifn-γ mRNA or Il-4 mRNA, we successfully polarized these cells to adopt either M1 or M2 macrophage activation phenotypes. This cellular reprogramming was demonstrated through increased expression of known surface markers and through the differential modulation of NADPH oxidase activity, or the superoxide burst. Our study demonstrates how IVT strategies can be combined to recruit and reprogram immune effector cells that have the capacity to fulfill complex biological tasks in vivo.


Asunto(s)
Reprogramación Celular , Macrófagos/inmunología , Monocitos/inmunología , ARN Mensajero/inmunología , Animales , Células Cultivadas , Quimiocina CCL2/genética , Quimiocina CCL2/inmunología , Quimiocina CCL3/genética , Células HeLa , Humanos , Interferón gamma/genética , Interleucina-4/genética , Linfocitos/inmunología , Activación de Macrófagos , Ratones , Ratones Endogámicos C57BL , Prueba de Estudio Conceptual , ARN Mensajero/síntesis química , Transcripción Genética
9.
Mol Ther ; 28(3): 805-819, 2020 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-31995741

RESUMEN

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.


Asunto(s)
Anticuerpos ampliamente neutralizantes/inmunología , Expresión Génica , Anticuerpos Anti-VIH/inmunología , Membrana Mucosa/inmunología , Membrana Mucosa/metabolismo , ARN Mensajero/administración & dosificación , Vagina , Vacunas contra el SIDA/administración & dosificación , Vacunas contra el SIDA/genética , Vacunas contra el SIDA/inmunología , Aerosoles , Animales , Chlorocebus aethiops , Femenino , Técnica del Anticuerpo Fluorescente , Infecciones por VIH/inmunología , VIH-1/inmunología , Ratones , Pruebas de Neutralización , ARN Mensajero/síntesis química , Ovinos , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Vagina/inmunología , Vagina/metabolismo , Células Vero
10.
Proc Natl Acad Sci U S A ; 115(42): E9944-E9952, 2018 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-30275336

RESUMEN

Dysfunctional endothelium causes more disease than any other cell type. Systemically administered RNA delivery to nonliver tissues remains challenging, in large part because there is no high-throughput method to identify nanoparticles that deliver functional mRNA to cells in vivo. Here we report a system capable of simultaneously quantifying how >100 lipid nanoparticles (LNPs) deliver mRNA that is translated into functional protein. Using this system (named FIND), we measured how >250 LNPs delivered mRNA to multiple cell types in vivo and identified 7C2 and 7C3, two LNPs that efficiently deliver siRNA, single-guide RNA (sgRNA), and mRNA to endothelial cells. The 7C3 delivered Cas9 mRNA and sgRNA to splenic endothelial cells as efficiently as hepatocytes, distinguishing it from LNPs that deliver Cas9 mRNA and sgRNA to hepatocytes more than other cell types. These data demonstrate that FIND can identify nanoparticles with novel tropisms in vivo.


Asunto(s)
Sistemas CRISPR-Cas , Células Endoteliales/metabolismo , Edición Génica , Técnicas de Transferencia de Gen , Lípidos/química , Nanopartículas/administración & dosificación , ARN Guía de Kinetoplastida/genética , ARN Mensajero/genética , Animales , Células Cultivadas , Células Endoteliales/citología , Células HEK293 , Hepatocitos/citología , Hepatocitos/metabolismo , Ensayos Analíticos de Alto Rendimiento , Humanos , Ratones , Ratones Endogámicos C57BL , Nanopartículas/química , ARN Guía de Kinetoplastida/química , ARN Mensajero/química
11.
J Neuroinflammation ; 17(1): 197, 2020 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-32563258

RESUMEN

BACKGROUND: Appropriately modulating inflammation after traumatic brain injury (TBI) may prevent disabilities for the millions of those inflicted annually. In TBI, cellular mediators of inflammation, including macrophages and microglia, possess a range of phenotypes relevant for an immunomodulatory therapeutic approach. It is thought that early phenotypic modulation of these cells will have a cascading healing effect. In fact, an anti-inflammatory, "M2-like" macrophage phenotype after TBI has been associated with neurogenesis, axonal regeneration, and improved white matter integrity (WMI). There already exist clinical trials seeking an M2-like bias through mesenchymal stem/stromal cells (MSCs). However, MSCs do not endogenously synthesize key signals that induce robust M2-like phenotypes such as interleukin-4 (IL-4). METHODS: To enrich M2-like macrophages in a clinically relevant manner, we augmented MSCs with synthetic IL-4 mRNA to transiently express IL-4. These IL-4 expressing MSCs (IL-4 MSCs) were characterized for expression and functionality and then delivered in a modified mouse TBI model of closed head injury. Groups were assessed for functional deficits and MR imaging. Brain tissue was analyzed through flow cytometry, multi-plex ELISA, qPCR, histology, and RNA sequencing. RESULTS: We observed that IL-4 MSCs indeed induce a robust M2-like macrophage phenotype and promote anti-inflammatory gene expression after TBI. However, here we demonstrate that acute enrichment of M2-like macrophages did not translate to improved functional or histological outcomes, or improvements in WMI on MR imaging. To further understand whether dysfunctional pathways underlie the lack of therapeutic effect, we report transcriptomic analysis of injured and treated brains. Through this, we discovered that inflammation persists despite acute enrichment of M2-like macrophages in the brain. CONCLUSION: The results demonstrate that MSCs can be engineered to induce a stronger M2-like macrophage response in vivo. However, they also suggest that acute enrichment of only M2-like macrophages after diffuse TBI cannot orchestrate neurogenesis, axonal regeneration, or improve WMI. Here, we also discuss our modified TBI model and methods to assess severity, behavioral studies, and propose that IL-4 expressing MSCs may also have relevance in other cavitary diseases or in improving biomaterial integration into tissues.


Asunto(s)
Lesiones Traumáticas del Encéfalo/metabolismo , Interleucina-4/metabolismo , Macrófagos/metabolismo , Células Madre Mesenquimatosas/metabolismo , Animales , Modelos Animales de Enfermedad , Inflamación/metabolismo , Masculino , Ratones , Microglía/metabolismo
12.
Biol Reprod ; 103(2): 275-285, 2020 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-32607584

RESUMEN

Sexually transmitted infections are highly prevalent, and over 40% of pregnancies are unplanned. We are producing new antibody-based multipurpose prevention technology products to address these problems and fill an unmet need in female reproductive health. We used a Nicotiana platform to manufacture monoclonal antibodies against two prevalent sexually transmitted pathogens, HIV-1 and HSV-2, and incorporated them into a vaginal film (MB66) for preclinical and Phase 1 clinical testing. These tests are now complete and indicate that MB66 is effective and safe in women. We are now developing an antisperm monoclonal antibody to add contraceptive efficacy to this product. The antisperm antibody, H6-3C4, originally isolated by Shinzo Isojima from the blood of an infertile woman, recognizes a carbohydrate epitope on CD52g, a glycosylphosphatidylinositol-anchored glycoprotein found in abundance on the surface of human sperm. We engineered the antibody for production in Nicotiana; the new antibody which we call "human contraception antibody," effectively agglutinates sperm at concentrations >10 µg/ml and maintains activity under a variety of physiological conditions. We are currently seeking regulatory approval for a Phase 1 clinical trial, which will include safety and "proof of principle" efficacy endpoints. Concurrently, we are working with new antibody production platforms to bring the costs down, innovative antibody designs that may produce more effective second-generation antibodies, and delivery systems to provide extended protection.


Asunto(s)
Anticuerpos Monoclonales , Anticoncepción/métodos , Salud Reproductiva , Femenino , Humanos , Masculino
13.
PLoS Pathog ; 14(8): e1007278, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30153309

RESUMEN

The GI tract is preferentially targeted during acute/early HIV-1 infection. Consequent damage to the gut plays a central role in HIV pathogenesis. The basis for preferential targeting of gut tissues is not well defined. Recombinant proteins and synthetic peptides derived from HIV and SIV gp120 bind directly to integrin α4ß7, a gut-homing receptor. Using both cell-surface expressed α4ß7 and a soluble α4ß7 heterodimer we demonstrate that its specific affinity for gp120 is similar to its affinity for MAdCAM (its natural ligand). The gp120 V2 domain preferentially engages extended forms of α4ß7 in a cation -sensitive manner and is inhibited by soluble MAdCAM. Thus, V2 mimics MAdCAM in the way that it binds to α4ß7, providing HIV a potential mechanism to discriminate between functionally distinct subsets of lymphocytes, including those with gut-homing potential. Furthermore, α4ß7 antagonists developed for the treatment of inflammatory bowel diseases, block V2 binding to α4ß7. A 15-amino acid V2 -derived peptide is sufficient to mediate binding to α4ß7. It includes the canonical LDV/I α4ß7 binding site, a cryptic epitope that lies 7-9 amino acids amino terminal to the LDV/I, and residues K169 and I181. These two residues were identified in a sieve analysis of the RV144 vaccine trial as sites of vaccine -mediated immune pressure. HIV and SIV V2 mAbs elicited by both vaccination and infection that recognize this peptide block V2-α4ß7 interactions. These mAbs recognize conformations absent from the ß- barrel presented in a stabilized HIV SOSIP gp120/41 trimer. The mimicry of MAdCAM-α4ß7 interactions by V2 may influence early events in HIV infection, particularly the rapid seeding of gut tissues, and supports the view that HIV replication in gut tissue is a central feature of HIV pathogenesis.


Asunto(s)
Proteína gp120 de Envoltorio del VIH/química , Proteína gp120 de Envoltorio del VIH/inmunología , Proteína gp120 de Envoltorio del VIH/metabolismo , Infecciones por VIH/prevención & control , Integrinas/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Simio/prevención & control , Vacunas contra el SIDA/química , Vacunas contra el SIDA/inmunología , Vacunas contra el SIDA/metabolismo , Animales , Anticuerpos Monoclonales , Sitios de Unión/inmunología , Línea Celular Tumoral , Epítopos/química , Epítopos/inmunología , Anticuerpos Anti-VIH/química , Anticuerpos Anti-VIH/inmunología , Anticuerpos Anti-VIH/metabolismo , Infecciones por VIH/inmunología , VIH-1/inmunología , Macaca , Unión Proteica , Dominios y Motivos de Interacción de Proteínas/inmunología , Vacunas contra el SIDAS/química , Vacunas contra el SIDAS/inmunología , Vacunas contra el SIDAS/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Vacunación/métodos
14.
J Biol Chem ; 293(41): 15867-15886, 2018 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-30108174

RESUMEN

Transforming growth factor-ß (TGFß) signaling through SMAD2/3 is an important driver of pathological fibrosis in multiple organ systems. TGFß signaling and extracellular matrix (ECM) stiffness form an unvirtuous pathological circuit in which matrix stiffness drives activation of latent TGFß, and TGFß signaling then drives cellular stress and ECM synthesis. Moreover, ECM stiffness also appears to sensitize cells to exogenously activated TGFß through unknown mechanisms. Here, using human fibroblasts, we explored the effect of ECM stiffness on a putative inner nuclear membrane protein, LEM domain-containing protein 3 (LEMD3), which is physically connected to the cell's actin cytoskeleton and inhibits TGFß signaling. We showed that LEMD3-SMAD2/3 interactions are inversely correlated with ECM stiffness and TGFß-driven luciferase activity and that LEMD3 expression is correlated with the mechanical response of the TGFß-driven luciferase reporter. We found that actin polymerization but not cellular stress or LEMD3-nuclear-cytoplasmic couplings were necessary for LEMD3-SMAD2/3 interactions. Intriguingly, LEMD3 and SMAD2/3 frequently interacted in the cytosol, and we discovered LEMD3 was proteolytically cleaved into protein fragments. We confirmed that a consensus C-terminal LEMD3 fragment binds SMAD2/3 in a stiffness-dependent manner throughout the cell and is sufficient for antagonizing SMAD2/3 signaling. Using human lung biopsies, we observed that these nuclear and cytosolic interactions are also present in tissue and found that fibrotic tissues exhibit locally diminished and cytoplasmically shifted LEMD3-SMAD2/3 interactions, as noted in vitro Our work reveals novel LEMD3 biology and stiffness-dependent regulation of TGFß by LEMD3, providing a novel target to antagonize pathological TGFß signaling.


Asunto(s)
Mecanotransducción Celular/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Proteínas Nucleares/metabolismo , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Actinas/metabolismo , Citosol/metabolismo , Proteínas de Unión al ADN , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Humanos , Fibrosis Pulmonar Idiopática/metabolismo , Pulmón/metabolismo , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/química , Lámina Nuclear/metabolismo , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/química , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Fosforilación , Proteína Fosfatasa 2C/metabolismo , Proteína Smad2/antagonistas & inhibidores , Proteína Smad2/química , Proteína smad3/antagonistas & inhibidores , Proteína smad3/química , Factor de Crecimiento Transformador beta/antagonistas & inhibidores
15.
Nucleic Acids Res ; 45(12): e113, 2017 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-28449134

RESUMEN

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.


Asunto(s)
Endosomas/metabolismo , Imagen Óptica/métodos , ARN Mensajero/farmacocinética , Análisis de la Célula Individual/métodos , Coloración y Etiquetado/métodos , Animales , Transporte Biológico , Carbocianinas/química , Línea Celular , Gránulos Citoplasmáticos/metabolismo , Gránulos Citoplasmáticos/ultraestructura , Fibroblastos/citología , Fibroblastos/metabolismo , Colorantes Fluorescentes/química , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Rayos Infrarrojos , Inyecciones Intramusculares , Ratones , Sondas Moleculares/química , Hibridación de Ácido Nucleico , ARN Mensajero/química , ARN Mensajero/genética
16.
Nano Lett ; 18(3): 2148-2157, 2018 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-29489381

RESUMEN

Endothelial cells and macrophages play active roles in disease and as a result are important targets for nucleic acid therapies. While thousands of chemically distinct lipid nanoparticles (LNPs) can be synthesized to deliver nucleic acids, studying more than a few LNPs in vivo is challenging. As a result, it is difficult to understand how nanoparticles target these cells in vivo. Using high throughput LNP barcoding, we quantified how well LNPs delivered DNA barcodes to endothelial cells and macrophages in vitro, as well as endothelial cells and macrophages isolated from the lung, heart, and bone marrow in vivo. We focused on two fundamental questions in drug delivery. First, does in vitro LNP delivery predict in vivo LNP delivery? By comparing how 281 LNPs delivered barcodes to endothelial cells and macrophages in vitro and in vivo, we found in vitro delivery did not predict in vivo delivery. Second, does LNP delivery change within the microenvironment of a tissue? We quantified how 85 LNPs delivered barcodes to eight splenic cell populations, and found that cell types derived from myeloid progenitors tended to be targeted by similar LNPs, relative to cell types derived from lymphoid progenitors. These data demonstrate that barcoded LNPs can elucidate fundamental questions about in vivo nanoparticle delivery.


Asunto(s)
Sistemas de Liberación de Medicamentos , Lípidos/química , Nanopartículas/química , Ácidos Nucleicos/administración & dosificación , Animales , Línea Celular , Células Cultivadas , Células Endoteliales/metabolismo , Femenino , Humanos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Nanotecnología , Ácidos Nucleicos/farmacocinética
17.
Nat Methods ; 12(5): 427-32, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25751144

RESUMEN

The detection of viral dynamics and localization in the context of controlled HIV infection remains a challenge and is limited to blood and biopsies. We developed a method to capture total-body simian immunodeficiency virus (SIV) replication using immunoPET (antibody-targeted positron emission tomography). The administration of a poly(ethylene glycol)-modified, (64)Cu-labeled SIV Gp120-specific antibody led to readily detectable signals in the gastrointestinal and respiratory tract, lymphoid tissues and reproductive organs of viremic monkeys. Viral signals were reduced in aviremic antiretroviral-treated monkeys but detectable in colon, select lymph nodes, small bowel, nasal turbinates, the genital tract and lung. In elite controllers, virus was detected primarily in foci in the small bowel, select lymphoid areas and the male reproductive tract, as confirmed by quantitative reverse-transcription PCR (qRT-PCR) and immunohistochemistry. This real-time, in vivo viral imaging method has broad applications to the study of immunodeficiency virus pathogenesis, drug and vaccine development, and the potential for clinical translation.


Asunto(s)
Antirretrovirales/uso terapéutico , Tomografía de Emisión de Positrones/métodos , Virus de la Inmunodeficiencia de los Simios , Imagen de Cuerpo Entero/métodos , Adenina/análogos & derivados , Adenina/uso terapéutico , Animales , Radioisótopos de Cobre , Desoxicitidina/análogos & derivados , Desoxicitidina/uso terapéutico , Emtricitabina , Inmunohistoquímica , Masculino , Glicoproteínas de Membrana/metabolismo , Naftiridinas/uso terapéutico , Organofosfonatos/uso terapéutico , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sensibilidad y Especificidad , Tenofovir , Proteínas del Envoltorio Viral/metabolismo , Viremia , Replicación Viral
18.
Bioconjug Chem ; 29(9): 3072-3083, 2018 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-30067354

RESUMEN

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.


Asunto(s)
Inmunidad Innata/efectos de los fármacos , ARN Mensajero/genética , Vacunas Sintéticas/farmacología , Animales , Formación de Anticuerpos , Inmunidad Celular , Inyecciones Intramusculares , Ratones , Células RAW 264.7 , Transcripción Genética , Vacunas Sintéticas/administración & dosificación
19.
Curr HIV/AIDS Rep ; 15(2): 127-135, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29478152

RESUMEN

PURPOSE OF REVIEW: Acute HIV infection is characterized by high-level viral replication throughout the body's lymphoid system, particularly in gut-associated lymphoid tissues resulting in damage to structural components of gut tissue. This damage is irreversible and believed to contribute to the development of immune deficiencies. Antiretroviral therapy (ART) does not restore gut structure and function. Studies in macaques point to an alternative treatment strategy that may ameliorate gut damage. Integrin α4ß7 mediates the homing of lymphocytes to gut tissues. Vedolizumab, a monoclonal antibody (mAb) antagonist of α4ß7, has demonstrated efficacy and has been approved for the treatment of inflammatory bowel disease in humans. Here, we describe our current knowledge, and the gaps in our understanding, of the role of α4ß7 in HIV pathogenesis and treatment. RECENT FINDINGS: When administered to macaques prior to infection, a nonhuman primate analogue of vedolizumab prevents transmission of SIV. In combination with ART, this mAb facilitates durable virologic control following treatment interruption. Targeting α4ß7 represents a novel therapeutic approach to prevent and treat HIV infection.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Fármacos Gastrointestinales/uso terapéutico , Infecciones por VIH/inmunología , Infecciones por VIH/terapia , Integrinas/antagonistas & inhibidores , Animales , Enfermedades Gastrointestinales/etiología , Enfermedades Gastrointestinales/terapia , Infecciones por VIH/complicaciones , Humanos , Integrinas/metabolismo
20.
Methods ; 98: 91-98, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26875782

RESUMEN

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.


Asunto(s)
Regulación Viral de la Expresión Génica , VIH-1/química , Imagen Molecular/métodos , ARN Mensajero/química , ARN Viral/química , Virus Sincitiales Respiratorios/química , Coloración y Etiquetado/métodos , Animales , Chlorocebus aethiops , Colorantes Fluorescentes/química , VIH-1/genética , VIH-1/metabolismo , Células Hep G2 , Humanos , Oligonucleótidos/química , Sondas ARN/química , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , Virus Sincitiales Respiratorios/genética , Virus Sincitiales Respiratorios/metabolismo , Células Vero , Virión/química , Virión/genética , Virión/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA