Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 120(3): e2205049120, 2023 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-36634134

RESUMEN

Stimulator of interferon genes (STING) signaling has been extensively studied in inflammatory diseases and cancer, while its role in T cell responses to infection is unclear. Using Listeria monocytogenes strains engineered to induce different levels of c-di-AMP, we found that high STING signals impaired T cell memory upon infection via increased Bim levels and apoptosis. Unexpectedly, reduction of TCR signal strength or T cell-STING expression decreased Bim expression, T cell apoptosis, and recovered T cell memory. We found that TCR signal intensity coupled STING signal strength to the unfolded protein response (UPR) and T cell survival. Under strong STING signaling, Indoleamine-pyrrole 2,3-dioxygenase (IDO) inhibition also reduced apoptosis and led to a recovery of T cell memory in STING sufficient CD8 T cells. Thus, STING signaling regulates CD8 T cell memory fitness through both cell-intrinsic and extrinsic mechanisms. These studies provide insight into how IDO and STING therapies could improve long-term T cell protective immunity.


Asunto(s)
Receptores de Antígenos de Linfocitos T , Transducción de Señal , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T CD8-positivos , Células T de Memoria , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo
2.
Infect Immun ; 87(8)2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31235641

RESUMEN

Live-attenuated Listeria monocytogenes has shown encouraging potential as an immunotherapy platform in preclinical and clinical settings. However, additional safety measures will enable application across malignant and infectious diseases. Here, we describe a new vaccine platform, termed Lm-RIID (L. monocytogenes recombinase-induced intracellular death), that induces the deletion of genes required for bacterial viability yet maintains potent T cell responses to encoded antigens. Lm-RIID grows normally in broth but commits suicide inside host cells by inducing Cre recombinase and deleting essential genes flanked by loxP sites, resulting in a self-limiting infection even in immunocompromised mice. Lm-RIID vaccination of mice induces potent CD8+ T cells and protects against virulent challenges, similar to live L. monocytogenes vaccines. When combined with α-PD-1, Lm-RIID is as effective as live-attenuated L. monocytogenes in a therapeutic tumor model. This impressive efficacy, together with the increased clearance rate, makes Lm-RIID ideal for prophylactic immunization against diseases that require T cells for protection.


Asunto(s)
Vacunas Bacterianas/inmunología , Listeria monocytogenes/inmunología , Animales , Femenino , Inmunoterapia , Listeria monocytogenes/patogenicidad , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Linfocitos T/inmunología , Vacunas Atenuadas/inmunología , Virulencia
3.
Cell Rep ; 23(5): 1435-1447, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29719256

RESUMEN

There are a limited number of adjuvants that elicit effective cell-based immunity required for protection against intracellular bacterial pathogens. Here, we report that STING-activating cyclic dinucleotides (CDNs) formulated in a protein subunit vaccine elicit long-lasting protective immunity to Mycobacterium tuberculosis in the mouse model. Subcutaneous administration of this vaccine provides equivalent protection to that of the live attenuated vaccine strain Bacille Calmette-Guérin (BCG). Protection is STING dependent but type I IFN independent and correlates with an increased frequency of a recently described subset of CXCR3-expressing T cells that localize to the lung parenchyma. Intranasal delivery results in superior protection compared with BCG, significantly boosts BCG-based immunity, and elicits both Th1 and Th17 immune responses, the latter of which correlates with enhanced protection. Thus, a CDN-adjuvanted protein subunit vaccine has the capability of eliciting a multi-faceted immune response that results in protection from infection by an intracellular pathogen.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Vacuna BCG/farmacología , Proteínas de la Membrana/inmunología , Mycobacterium tuberculosis/inmunología , Células Th17/inmunología , Tuberculosis Pulmonar/prevención & control , Animales , Vacuna BCG/inmunología , Modelos Animales de Enfermedad , Inmunidad Celular/efectos de los fármacos , Ratones , Ratones Noqueados , Células TH1/inmunología , Células TH1/patología , Células Th17/patología , Tuberculosis Pulmonar/inmunología , Tuberculosis Pulmonar/patología , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/farmacocinética
4.
Nature ; 501(7468): 512-6, 2013 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-24005326

RESUMEN

Ubiquitin-mediated targeting of intracellular bacteria to the autophagy pathway is a key innate defence mechanism against invading microbes, including the important human pathogen Mycobacterium tuberculosis. However, the ubiquitin ligases responsible for catalysing ubiquitin chains that surround intracellular bacteria are poorly understood. The parkin protein is a ubiquitin ligase with a well-established role in mitophagy, and mutations in the parkin gene (PARK2) lead to increased susceptibility to Parkinson's disease. Surprisingly, genetic polymorphisms in the PARK2 regulatory region are also associated with increased susceptibility to intracellular bacterial pathogens in humans, including Mycobacterium leprae and Salmonella enterica serovar Typhi, but the function of parkin in immunity has remained unexplored. Here we show that parkin has a role in ubiquitin-mediated autophagy of M. tuberculosis. Both parkin-deficient mice and flies are sensitive to various intracellular bacterial infections, indicating parkin has a conserved role in metazoan innate defence. Moreover, our work reveals an unexpected functional link between mitophagy and infectious disease.


Asunto(s)
Drosophila melanogaster/inmunología , Drosophila melanogaster/microbiología , Inmunidad Innata/inmunología , Mycobacterium marinum/inmunología , Mycobacterium tuberculosis/inmunología , Salmonella typhimurium/inmunología , Ubiquitina-Proteína Ligasas/inmunología , Animales , Autofagia/inmunología , Células de la Médula Ósea/microbiología , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Femenino , Lisina/metabolismo , Macrófagos/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Mitocondrias/patología , Mitofagia , Modelos Inmunológicos , Mycobacterium tuberculosis/crecimiento & desarrollo , Mycobacterium tuberculosis/metabolismo , Poliubiquitina/química , Poliubiquitina/metabolismo , Simbiosis/inmunología , Tuberculosis/enzimología , Tuberculosis/inmunología , Tuberculosis/microbiología , Tuberculosis/patología , Ubiquitina/análisis , Ubiquitina/química , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/deficiencia , Ubiquitina-Proteína Ligasas/metabolismo
5.
Adv Immunol ; 113: 135-56, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22244582

RESUMEN

Acquired cell-mediated immunity to Listeria monocytogenes is induced by infection with live, replicating bacteria that grow in the host cell cytosol, whereas killed bacteria, or those trapped in a phagosome, fail to induce protective immunity. In this chapter, we focus on how L. monocytogenes is sensed by the innate immune system, with the presumption that innate immunity affects the development of acquired immunity. Infection by L. monocytogenes induces three innate immune pathways: an MyD88-dependent pathway emanating from a phagosome leading to expression of inflammatory cytokines; a STING/IRF3-dependent pathway emanating from the cytosol leading to the expression of IFN-ß and coregulated genes; and very low levels of a Caspase-1-dependent, AIM2-dependent inflammasome pathway resulting in proteolytic activation and secretion of IL-1ß and IL-18 and pyroptotic cell death. Using a combination of genetics and biochemistry, we identified the listerial ligand that activates the STING/IRF3 pathway as secreted cyclic diadenosine monophosphate, a newly discovered conserved bacterial signaling molecule. We also identified L. monocytogenes mutants that caused robust inflammasome activation due to bacteriolysis in the cytosol, release of DNA, and activation of the AIM2 inflammasome. A strain was constructed that ectopically expressed and secreted a fusion protein containing Legionella pneumophila flagellin that robustly activated the Nlrc4-dependent inflammasome and was highly attenuated in mice, also in an Nlrc4-dependent manner. Surprisingly, this strain was a poor inducer of adaptive immunity, suggesting that inflammasome activation is not necessary to induce cell-mediated immunity and may even be detrimental under some conditions. To the best of our knowledge, no single innate immune pathway is necessary to mount a robust acquired immune response to L. monocytogenes infection.


Asunto(s)
Citocinas/inmunología , Inmunidad Celular , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Animales , Humanos , Inmunidad Innata , Inflamación , Ratones , Transducción de Señal
6.
Biochim Biophys Acta ; 1818(3): 689-94, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22100748

RESUMEN

Genetically engineered cells with mutations of relevance to electroporation, cell membrane permeabilization by electric pulses, can become a promising new tool for fundamental research on this important biotechnology. Listeria monocytogenes mutants lacking DltA or MprF and assayed for sensitivity to the cathelicidin like anti-microbial cationic peptide (mCRAMP), were developed to study the effect of cell wall charge on electroporation. Working in the irreversible electroporation regime (IRE), we found that application of a sequence of 50 pulses, each 50µs duration, 12.5kV/cm field, delivered at 2Hz led to 2.67±0.29 log reduction in wild-type L. monocytogenes, log 2.60±0.19 in the MprF-minus mutant, and log 1.33±0.13 in the DltA-minus mutant. The experimental observation that the DltA-minus mutant was highly susceptible to cationic mCRAMP and resistant to IRE suggests that the charge on the bacterial cell wall affects electroporation and shows that this approach may be promising for fundamental studies on electroporation.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/química , Pared Celular , Electroporación/métodos , Listeria monocytogenes/enzimología , Aminoaciltransferasas/genética , Aminoaciltransferasas/metabolismo , Ligasas de Carbono-Oxígeno/genética , Ligasas de Carbono-Oxígeno/metabolismo , Eliminación de Gen , Listeria monocytogenes/genética
7.
Infect Immun ; 79(9): 3596-606, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21768286

RESUMEN

Listeria monocytogenes is a Gram-positive intracellular pathogen that is naturally resistant to lysozyme. Recently, it was shown that peptidoglycan modification by N-deacetylation or O-acetylation confers resistance to lysozyme in various Gram-positive bacteria, including L. monocytogenes. L. monocytogenes peptidoglycan is deacetylated by the action of N-acetylglucosamine deacetylase (Pgd) and acetylated by O-acetylmuramic acid transferase (Oat). We characterized Pgd(-), Oat(-), and double mutants to determine the specific role of L. monocytogenes peptidoglycan acetylation in conferring lysozyme sensitivity during infection of macrophages and mice. Pgd(-) and Pgd(-) Oat(-) double mutants were attenuated approximately 2 and 3.5 logs, respectively, in vivo. In bone-marrow derived macrophages, the mutants demonstrated intracellular growth defects and increased induction of cytokine transcriptional responses that emanated from a phagosome and the cytosol. Lysozyme-sensitive mutants underwent bacteriolysis in the macrophage cytosol, resulting in AIM2-dependent pyroptosis. Each of the in vitro phenotypes was rescued upon infection of LysM(-) macrophages. The addition of extracellular lysozyme to LysM(-) macrophages restored cytokine induction, host cell death, and L. monocytogenes growth inhibition. This surprising observation suggests that extracellular lysozyme can access the macrophage cytosol and act on intracellular lysozyme-sensitive bacteria.


Asunto(s)
Amidohidrolasas/metabolismo , Inmunidad Innata , Listeria monocytogenes/enzimología , Listeria monocytogenes/inmunología , Muramidasa/metabolismo , Transferasas/metabolismo , Acetilación , Amidohidrolasas/genética , Animales , Apoptosis , Bacteriólisis , Citocinas/biosíntesis , Femenino , Listeria monocytogenes/genética , Listeria monocytogenes/crecimiento & desarrollo , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Mutación , Peptidoglicano/inmunología , Peptidoglicano/metabolismo , Interferencia de ARN , ARN Interferente Pequeño , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Transferasas/genética
8.
PLoS Pathog ; 7(3): e1001326, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21455492

RESUMEN

Host defense against the intracellular pathogen Listeria monocytogenes (Lm) requires innate and adaptive immunity. Here, we directly imaged immune cell dynamics at Lm foci established by dendritic cells in the subcapsular red pulp (scDC) using intravital microscopy. Blood borne Lm rapidly associated with scDC. Myelomonocytic cells (MMC) swarmed around non-motile scDC forming foci from which blood flow was excluded. The depletion of scDC after foci were established resulted in a 10-fold reduction in viable Lm, while graded depletion of MMC resulted in 30-1000 fold increase in viable Lm in foci with enhanced blood flow. Effector CD8+ T cells at sites of infection displayed a two-tiered reduction in motility with antigen independent and antigen dependent components, including stable interactions with infected and non-infected scDC. Thus, swarming MMC contribute to control of Lm prior to development of T cell immunity by direct killing and sequestration from blood flow, while scDC appear to promote Lm survival while preferentially interacting with CD8+ T cells in effector sites.


Asunto(s)
Inmunidad Adaptativa , Inmunidad Innata , Listeria monocytogenes/patogenicidad , Listeriosis/patología , Linfocitos T Citotóxicos/patología , Animales , Citotoxicidad Inmunológica , Células Dendríticas/inmunología , Células Dendríticas/patología , Femenino , Técnicas de Sustitución del Gen , Interacciones Huésped-Patógeno , Listeriosis/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Monocitos/inmunología , Monocitos/patología , Linfocitos T Citotóxicos/inmunología
9.
Infect Immun ; 79(2): 688-94, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21098106

RESUMEN

Type I interferons (IFNs) are central regulators of the innate and adaptive immune responses to viral and bacterial infections. Type I IFNs are induced upon cytosolic detection of microbial nucleic acids, including DNA, RNA, and the bacterial second messenger cyclic-di-GMP (c-di-GMP). In addition, a recent study demonstrated that the intracellular bacterial pathogen Listeria monocytogenes stimulates a type I IFN response due to cytosolic detection of bacterially secreted c-di-AMP. The transmembrane signaling adaptor Sting (Tmem173, Mita, Mpys, Eris) has recently been implicated in the induction of type I IFNs in response to cytosolic DNA and/or RNA. However, the role of Sting in response to purified cyclic dinucleotides or during in vivo L. monocytogenes infection has not been addressed. In order to identify genes important in the innate immune response, we have been conducting a forward genetic mutagenesis screen in C57BL/6 mice using the mutagen N-ethyl-N-nitrosourea (ENU). Here we describe a novel mutant mouse strain, Goldenticket (Gt), that fails to produce type I IFNs upon L. monocytogenes infection. By genetic mapping and complementation experiments, we found that Gt mice harbor a single nucleotide variant (T596A) of Sting that functions as a null allele and fails to produce detectable protein. Analysis of macrophages isolated from Gt mice revealed that Sting is absolutely required for the type I interferon response to both c-di-GMP and c-di-AMP. Additionally, Sting is required for the response to c-di-GMP and L. monocytogenes in vivo. Our results provide new functions for Sting in the innate interferon response to pathogens.


Asunto(s)
GMP Cíclico/análogos & derivados , Fosfatos de Dinucleósidos/metabolismo , Interferón Tipo I/metabolismo , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Proteínas de la Membrana/fisiología , Alelos , Animales , Línea Celular , GMP Cíclico/metabolismo , Etilnitrosourea/toxicidad , Femenino , Regulación de la Expresión Génica/fisiología , Prueba de Complementación Genética , Humanos , Interferón Tipo I/genética , Listeriosis/metabolismo , Macrófagos/microbiología , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Mutantes , Mutación , Polimorfismo de Nucleótido Simple
10.
PLoS One ; 5(1): e8610, 2010 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-20062534

RESUMEN

BACKGROUND: Recent studies have suggested that autophagy is utilized by cells as a protective mechanism against Listeria monocytogenes infection. METHODOLOGY/PRINCIPAL FINDINGS: However we find autophagy has no measurable role in vacuolar escape and intracellular growth in primary cultured bone marrow derived macrophages (BMDMs) deficient for autophagy (atg5-/-). Nevertheless, we provide evidence that the pore forming activity of the cholesterol-dependent cytolysin listeriolysin O (LLO) can induce autophagy subsequent to infection by L. monocytogenes. Infection of BMDMs with L. monocytogenes induced microtubule-associated protein light chain 3 (LC3) lipidation, consistent with autophagy activation, whereas a mutant lacking LLO did not. Infection of BMDMs that express LC3-GFP demonstrated that wild-type L. monocytogenes was encapsulated by LC3-GFP, consistent with autophagy activation, whereas a mutant lacking LLO was not. Bacillus subtilis expressing either LLO or a related cytolysin, perfringolysin O (PFO), induced LC3 colocalization and LC3 lipidation. Further, LLO-containing liposomes also recruited LC3-GFP, indicating that LLO was sufficient to induce targeted autophagy in the absence of infection. The role of autophagy had variable effects depending on the cell type assayed. In atg5-/- mouse embryonic fibroblasts, L. monocytogenes had a primary vacuole escape defect. However, the bacteria escaped and grew normally in atg5-/- BMDMs. CONCLUSIONS/SIGNIFICANCE: We propose that membrane damage, such as that caused by LLO, triggers bacterial-targeted autophagy, although autophagy does not affect the fate of wild-type intracellular L. monocytogenes in primary BMDMs.


Asunto(s)
Autofagia/fisiología , Proteínas de Choque Térmico/fisiología , Proteínas Hemolisinas/fisiología , Listeriosis/inmunología , Animales , Toxinas Bacterianas , Células Cultivadas , Liposomas , Listeriosis/fisiopatología , Ratones
11.
PLoS One ; 4(11): e7981, 2009 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-19956734

RESUMEN

BACKGROUND: Plant viruses such as Cowpea mosaic virus (CPMV) are increasingly being developed for applications in nanobiotechnology including vaccine development because of their potential for producing large quantities of antigenic material in plant hosts. In order to improve efficacy of viral nanoparticles in these types of roles, an investigation of the individual cell types that interact with the particles is critical. In particular, it is important to understand the interactions of a potential vaccine with antigen presenting cells (APCs) of the immune system. CPMV was previously shown to interact with vimentin displayed on cell surfaces to mediate cell entry, but the expression of surface vimentin on APCs has not been characterized. METHODOLOGY: The binding and internalization of CPMV by several populations of APCs was investigated both in vitro and in vivo by flow cytometry and fluorescence confocal microscopy. The association of the particles with mouse gastrointestinal epithelium and Peyer's patches was also examined by confocal microscopy. The expression of surface vimentin on APCs was also measured. CONCLUSIONS: We found that CPMV is bound and internalized by subsets of several populations of APCs both in vitro and in vivo following intravenous, intraperitoneal, and oral administration, and also by cells isolated from the Peyer's patch following gastrointestinal delivery. Surface vimentin was also expressed on APC populations that could internalize CPMV. These experiments demonstrate that APCs capture CPMV particles in vivo, and that further tuning the interaction with surface vimentin may facilitate increased uptake by APCs and priming of antibody responses. These studies also indicate that CPMV particles likely access the systemic circulation following oral delivery via the Peyer's patch.


Asunto(s)
Células Presentadoras de Antígenos/metabolismo , Células Presentadoras de Antígenos/virología , Biotecnología/métodos , Comovirus/metabolismo , Nanopartículas , Nanotecnología/métodos , Animales , Membrana Celular/metabolismo , Separación Celular , Fibroblastos/metabolismo , Concentración de Iones de Hidrógeno , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal/métodos , Unión Proteica , Vimentina/química , Vimentina/metabolismo
12.
Chem Biol ; 14(10): 1152-62, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17961827

RESUMEN

Cowpea mosaic virus (CPMV) is a well-characterized nanoparticle that has been used for a variety of nanobiotechnology applications. CPMV interacts with several mammalian cell lines and tissues in vivo. To overcome natural CPMV targeting and redirect CPMV particles to cells of interest, we attached a folic acid-PEG conjugate by using the copper-catalyzed azide-alkyne cycloaddition reaction. PEGylation of CPMV completely eliminated background binding of the virus to tumor cells. The PEG-folate moiety allowed CPMV-specific recognition of tumor cells bearing the folate receptor. In addition, by testing CPMV formulations with different amounts of the PEG-FA moiety displayed on the surface, we show that higher-density loading of targeting ligands on CPMV may not be necessary for efficient targeting to tumor cells. These studies help to define the requirements for efficiently targeting nanoparticles and protein cages to tumors.


Asunto(s)
Comovirus/efectos de los fármacos , Ácido Fólico/farmacología , Nanopartículas , Nanotecnología/métodos , Polietilenglicoles/farmacología , Alquinos/química , Animales , Azidas/química , Western Blotting , Catálisis , Línea Celular Tumoral/efectos de los fármacos , Línea Celular Tumoral/patología , Cromatografía , Comovirus/fisiología , Cobre/química , Ácido Fólico/química , Humanos , Ligandos , Microscopía , Polietilenglicoles/química
13.
J Control Release ; 120(1-2): 41-50, 2007 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-17512998

RESUMEN

Virus-based nanoparticles (VNPs) from a variety of sources are being developed for biomedical and nanotechnology applications that include tissue targeting and drug delivery. However, the fate of most of those particles in vivo has not been investigated. Cowpea mosaic virus (CPMV), a plant comovirus, has been found to be amenable to the attachment of a variety of molecules to its coat protein, as well as to modification of the coat protein sequence by genetic means. We report here the results of studies of the bio-distribution, toxicology, and pathology of CPMV in mice. Plasma clearance and tissue biodistribution were measured using CPMV particles derivatized with lanthanide metal complexes. CPMV particles were cleared rapidly from plasma, falling to undetectable levels within 20 min. By 30 min the majority of the injected VNPs were trapped in the liver and to a lesser extent the spleen with undetectable amounts in other tissues. At doses of 1 mg, 10 mg and 100 mg per kg body weight, no toxicity was noted and the mice appeared to be normal. Hematology was essentially normal, although with the highest dose examined, the mice were somewhat leukopenic with relative decreases in both neutrophils and lymphocytes. Histological examination of the spleen showed cellular infiltration, which upon flow cytometry analyses revealed elevated B lymphocytes on the first day following virus administration that subsequently subsided. Microscopic evaluation of various other tissues revealed a lack of apparent tissue degeneration or necrosis. Overall, CPMV appears to be a safe and non-toxic platform for in vivo biomedical applications.


Asunto(s)
Comovirus , Portadores de Fármacos/farmacocinética , Portadores de Fármacos/toxicidad , Nanopartículas , Virión , Animales , Portadores de Fármacos/administración & dosificación , Portadores de Fármacos/química , Compuestos Heterocíclicos/química , Inyecciones Intravenosas , Hígado/metabolismo , Ratones , Ratones Endogámicos BALB C , Modelos Moleculares , Conformación Molecular , Compuestos Organometálicos/química , Espectrofotometría/métodos , Bazo/efectos de los fármacos , Bazo/metabolismo , Bazo/patología , Terbio/química
14.
J Virol ; 81(4): 1632-40, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17121801

RESUMEN

Cowpea mosaic virus (CPMV), a plant virus that is a member of the picornavirus superfamily, is increasingly being used for nanotechnology applications, including material science, vascular imaging, vaccine development, and targeted drug delivery. For these applications, it is critical to understand the in vivo interactions of CPMV within the mammalian system. Although the bioavailability of CPMV in the mouse has been demonstrated, the specific interactions between CPMV and mammalian cells need to be characterized further. Here we demonstrate that although the host range for replication of CPMV is confined to plants, mammalian cells nevertheless bind and internalize CPMV in significant amounts. This binding is mediated by a conserved 54-kDa protein found on the plasma membranes of both human and murine cell lines. Studies using a deficient cell line, deglycosidases, and glycosylation inhibitors showed that the CPMV binding protein (CPMV-BP) is not glycosylated. A possible 47-kDa isoform of the CPMV-BP was also detected in the organelle and nuclear subcellular fraction prepared from murine fibroblasts. Further characterization of CPMV-BP is important to understand how CPMV is trafficked through the mammalian system and may shed light on how picornaviruses may have evolved between plant and animal hosts.


Asunto(s)
Comovirus/metabolismo , Proteínas de la Membrana/metabolismo , Animales , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Humanos , Membranas Intracelulares/metabolismo , Mamíferos , Proteínas de la Membrana/química , Ratones , Ratones Endogámicos BALB C , Peso Molecular , Orgánulos/química , Orgánulos/metabolismo , Unión Proteica , Isoformas de Proteínas/metabolismo , Especificidad de la Especie
15.
Virology ; 343(2): 224-35, 2005 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-16185741

RESUMEN

The plant virus, cowpea mosaic virus (CPMV), is increasingly being used as a nanoparticle platform for multivalent display of peptides. A growing variety of applications have employed the CPMV display technology including vaccines, antiviral therapeutics, nanoblock chemistry, and materials science. CPMV chimeras can be inexpensively produced from experimentally infected cowpea plants and are completely stable at 37 degrees C and low pH, suggesting that they could be used as edible or mucosally-delivered vaccines or therapeutics. However, the fate of CPMV particles in vivo, or following delivery via the oral route, is unknown. To address this question, we examined CPMV in vitro and in vivo. CPMV was shown to be stable under simulated gastric conditions in vitro. The pattern of localization of CPMV particles to mouse tissues following oral or intravenous dosing was then determined. For several days following oral or intravenous inoculation, CPMV was found in a wide variety of tissues throughout the body, including the spleen, kidney, liver, lung, stomach, small intestine, lymph nodes, brain, and bone marrow. CPMV particles were detected after cardiac perfusion, suggesting that the particles entered the tissues. This pattern was confirmed using methods to specifically detect the viral capsid proteins and the internal viral RNA. The stability of CPMV virions in the gastrointestinal tract followed by their systemic dissemination supports their use as orally bioavailable nanoparticles.


Asunto(s)
Comovirus/ultraestructura , Nanoestructuras/ultraestructura , Administración Oral , Animales , Ácidos Carboxílicos , Comovirus/patogenicidad , Fabaceae/virología , Femenino , Colorantes Fluorescentes , Jugo Gástrico/virología , Técnicas In Vitro , Cinética , Ratones , Ratones Endogámicos C57BL , Nanotecnología , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...