Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
ACS Chem Biol ; 17(2): 292-298, 2022 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-35020348

RESUMO

Extrahepatic targeted delivery of oligonucleotides, such as small interfering RNA (siRNA) and antisense oligonucleotides (ASOs), is an attractive technology for the development of nucleic acid-based medicines. To target CD22-expressing B cells, several drug platforms have shown promise, including antibodies, antibody-drug conjugates, and nanoparticles, but to date CD22-targeted delivery of oligonucleotide therapeutics has not been reported. Here we report the uptake and enhancement of siRNA gene expression knockdown in CD22-expressing B cells using a chemically stabilized and modified CD22 glycan ligand-conjugated siRNA. This finding has the potential to broaden the use of siRNA technology, opening up novel therapeutic opportunities, and presents an innovative approach for targeted delivery of siRNAs to B cell lymphomas.


Assuntos
Oligonucleotídeos Antissenso , Polissacarídeos , Técnicas de Silenciamento de Genes , Ligantes , Polissacarídeos/metabolismo , RNA Interferente Pequeno/genética
2.
J Biol Chem ; 294(6): 1831-1845, 2019 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-30545939

RESUMO

The interaction between the receptor 4-1BB and its ligand 4-1BBL provides co-stimulatory signals for T-cell activation and proliferation. However, differences in the mouse and human molecules might result in differential engagement of this pathway. Here, we report the crystal structure of mouse 4-1BBL and of the mouse 4-1BB/4-1BBL complex, which together provided insights into the molecular mechanism by which m4-1BBL and its cognate receptor recognize each other. Unlike all human or mouse tumor necrosis factor ligands that form noncovalent and mostly trimeric assemblies, the m4-1BBL structure formed a disulfide-linked dimeric assembly. The structure disclosed that certain differences in the amino acid composition along the intramolecular interface, together with two specific residues (Cys-246 and Ser-256) present exclusively in m4-1BBL, are responsible for this unique dimerization. Unexpectedly, upon m4-1BB binding, m4-1BBL undergoes structural changes within each protomer; moreover, the individual m4-1BBL protomers rotate relative to each other, yielding a dimerization interface with more inter-subunit interactions. We also observed that in the m4-1BB/4-1BBL complex, each receptor monomer binds exclusively to a single ligand subunit with contributions of cysteine-rich domain 1 (CRD1), CRD2, and CRD3. Furthermore, structure-guided mutagenesis of the binding interface revealed that novel binding interactions with the GH loop, rather than the DE loop, are energetically critical and define the m4-1BB receptor selectivity for m4-1BBL. A comparison with the human 4-1BB/4-1BBL complex highlighted several differences between the ligand- and receptor-binding interfaces, providing an explanation for the absence of inter-species cross-reactivity between human and mouse 4-1BB and 4-1BBL molecules.


Assuntos
Ligante 4-1BB/química , Complexos Multiproteicos/química , Multimerização Proteica , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/química , Animais , Células HEK293 , Humanos , Camundongos , Domínios Proteicos , Estrutura Quaternária de Proteína , Células Sf9 , Spodoptera
4.
PLoS One ; 11(5): e0154616, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27171010

RESUMO

A proof-of-concept study evaluating the potential of Streptococcus pneumoniae Pneumococcal Surface Protein A (PspA) as a passive immunization target was conducted. We describe the generation and isolation of several broadly reactive mouse anti-PspA monoclonal antibodies (mAbs). MAb 140H1 displayed (i) 98% strain coverage, (ii) activity in complement deposition and opsonophagocytic killing (OPK) assays, which are thought to predict the in vivo efficacy of anti-pneumococcal mAbs, (iii) efficacy in mouse sepsis models both alone and in combination with standard-of-care antibiotics, and (iv) therapeutic activity in a mouse pneumonia model. Moreover, we demonstrate that antibody engineering can significantly enhance anti-PspA mAb effector function. We believe that PspA has promising potential as a target for the therapy of invasive pneumococcal disease by mAbs, which could be used alone or in conjunction with standard-of-care antibiotics.


Assuntos
Anticorpos Monoclonais/imunologia , Proteínas de Bactérias/imunologia , Streptococcus pneumoniae/imunologia , Animais , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/uso terapêutico , Complemento C3/metabolismo , Modelos Animais de Doenças , Mapeamento de Epitopos , Feminino , Humanos , Imunoglobulina G/sangue , Pneumopatias/imunologia , Pneumopatias/microbiologia , Camundongos Endogâmicos BALB C , Proteínas Opsonizantes/metabolismo , Fagócitos/metabolismo , Fagocitose , Infecções Pneumocócicas/tratamento farmacológico , Infecções Pneumocócicas/imunologia , Infecções Pneumocócicas/microbiologia , Ligação Proteica , Sepse/tratamento farmacológico , Sepse/imunologia , Sepse/microbiologia , Resultado do Tratamento
5.
Nanomedicine (Lond) ; 7(6): 877-88, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22394183

RESUMO

AIMS: Detection of atherosclerosis has generally been limited to the late stages of development, after cardiovascular symptoms present or a clinical event occurs. One possibility for early detection is the use of functionalized nanoparticles. The aim of this study was the early imaging of atherosclerosis using nanoparticles with a natural affinity for inflammatory cells in the lesion. MATERIALS & METHODS: We investigated uptake of cowpea mosaic virus by macrophages and foam cells in vitro and correlated this with vimentin expression. We also examined the ability of cowpea mosaic virus to interact with atherosclerotic lesions in a murine model of atherosclerosis. RESULTS & CONCLUSION: We found that uptake of cowpea mosaic virus is increased in areas of atherosclerotic lesion. This correlated with increased surface vimentin in the lesion compared with nonlesion vasculature. In conclusion, cowpea mosaic virus and its vimentin-binding region holds potential for use as a targeting ligand for early atherosclerotic lesions, and as a probe for detecting upregulation of surface vimentin during inflammation.


Assuntos
Aterosclerose/diagnóstico , Comovirus/imunologia , Nanopartículas , Vimentina/imunologia , Animais , Artérias/imunologia , Artérias/patologia , Aterosclerose/imunologia , Aterosclerose/patologia , Linhagem Celular , Células Cultivadas , Comovirus/química , Células Endoteliais/imunologia , Células Endoteliais/patologia , Células Espumosas/imunologia , Células Espumosas/patologia , Lipoproteínas LDL/imunologia , Macrófagos/imunologia , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nanopartículas/química , Placa Aterosclerótica/diagnóstico , Placa Aterosclerótica/imunologia , Placa Aterosclerótica/patologia
6.
Nat Protoc ; 5(8): 1406-17, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20671724

RESUMO

Viral nanoparticles are a novel class of biomolecular agents that take advantage of the natural circulatory and targeting properties of viruses to allow the development of therapeutics, vaccines and imaging tools. We have developed a multivalent nanoparticle platform based on the cowpea mosaic virus (CPMV) that facilitates particle labeling at high density with fluorescent dyes and other functional groups. Compared with other technologies, CPMV-based viral nanoparticles are particularly suited for long-term intravital vascular imaging because of their biocompatibility and retention in the endothelium with minimal side effects. The stable, long-term labeling of the endothelium allows the identification of vasculature undergoing active remodeling in real time. In this study, we describe the synthesis, purification and fluorescent labeling of CPMV nanoparticles, along with their use for imaging of vascular structure and for intravital vascular mapping in developmental and tumor angiogenesis models. Dye-labeled viral nanoparticles can be synthesized and purified in a single day, and imaging studies can be conducted over hours, days or weeks, depending on the application.


Assuntos
Comovirus/isolamento & purificação , Embrião de Mamíferos/irrigação sanguínea , Embrião não Mamífero/irrigação sanguínea , Endotélio Vascular/anatomia & histologia , Nanopartículas , Nanotecnologia/métodos , Neoplasias/irrigação sanguínea , Animais , Galinhas , Técnicas de Cultura Embrionária , Fabaceae/virologia , Corantes Fluorescentes/análise , Corantes Fluorescentes/química , Camundongos , Microinjeções/métodos , Microscopia Confocal/métodos , Microscopia de Fluorescência/métodos
7.
Nano Lett ; 10(3): 1093-7, 2010 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-20163184

RESUMO

Multivalent nanoparticle platforms are attractive for biomedical applications because of their improved target specificity, sensitivity, and solubility. However, their controlled assembly remains a considerable challenge. An efficient hydrazone ligation chemistry was applied to the assembly of Cowpea mosaic virus (CPMV) nanoparticles with individually tunable levels of a VEGFR-1 ligand and a fluorescent PEGylated peptide. The nanoparticles recognized VEGFR-1 on endothelial cell lines and VEGFR1-expressing tumor xenografts in mice, validating targeted CPMV as a nanoparticle platform in vivo.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Hidrazonas/química , Aumento da Imagem/métodos , Nanoestruturas/química , Vírion/química , Animais , Células HT29 , Humanos , Camundongos
8.
PLoS Pathog ; 5(5): e1000417, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19412526

RESUMO

Cowpea mosaic virus (CPMV) is a plant comovirus in the picornavirus superfamily, and is used for a wide variety of biomedical and material science applications. Although its replication is restricted to plants, CPMV binds to and enters mammalian cells, including endothelial cells and particularly tumor neovascular endothelium in vivo. This natural capacity has lead to the use of CPMV as a sensor for intravital imaging of vascular development. Binding of CPMV to endothelial cells occurs via interaction with a 54 kD cell-surface protein, but this protein has not previously been identified. Here we identify the CPMV binding protein as a cell-surface form of the intermediate filament vimentin. The CPMV-vimentin interaction was established using proteomic screens and confirmed by direct interaction of CPMV with purified vimentin, as well as inhibition in a vimentin-knockout cell line. Vimentin and CPMV were also co-localized in vascular endothelium of mouse and rat in vivo. Together these studies indicate that surface vimentin mediates binding and may lead to internalization of CPMV in vivo, establishing surface vimentin as an important vascular endothelial ligand for nanoparticle targeting to tumors. These results also establish vimentin as a ligand for picornaviruses in both the plant and animal kingdoms of life. Since bacterial pathogens and several other classes of viruses also bind to surface vimentin, these studies suggest a common role for surface vimentin in pathogen transmission.


Assuntos
Comovirus/metabolismo , Endotélio Vascular/metabolismo , Endotélio Vascular/virologia , Vimentina/metabolismo , Animais , Aorta/metabolismo , Linhagem Celular , Membrana Celular/metabolismo , Cromatografia Líquida , Células HeLa , Humanos , Masculino , Camundongos , Ligação Proteica , Proteômica , Ratos , Ratos Sprague-Dawley , Espectrometria de Massas em Tandem , Vírion/metabolismo
9.
PLoS One ; 3(10): e3315, 2008 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-18830402

RESUMO

BACKGROUND: Cowpea Mosaic Virus (CPMV) is increasingly being used as a nanoparticle platform for multivalent display of molecules via chemical bioconjugation to the capsid surface. A growing variety of applications have employed the CPMV multivalent display technology including nanoblock chemistry, in vivo imaging, and materials science. CPMV nanoparticles can be inexpensively produced from experimentally infected cowpea plants at high yields and are extremely stable. Although CPMV has not been shown to replicate in mammalian cells, uptake in mammalian cells does occur in vitro and in vivo. Thus, inactivation of the virus RNA genome is important for biosafety considerations, however the surface characteristics and chemical reactivity of the particles must be maintained in order to preserve chemical and structural functionality. METHODOLOGY/PRINCIPAL FINDINGS: Short wave (254 nm) UV irradiation was used to crosslink the RNA genome within intact particles. Lower doses of UV previously reported to inactivate CPMV infectivity inhibited symptoms on inoculated leaves but did not prohibit systemic virus spread in plants, whereas higher doses caused aggregation of the particles and an increase in chemical reactivity further indicating broken particles. Intermediate doses of 2.0-2.5 J/cm(2) were shown to maintain particle structure and chemical reactivity, and cellular binding properties were similar to CPMV-WT. CONCLUSIONS: These studies demonstrate that it is possible to inactivate CPMV infectivity while maintaining particle structure and function, thus paving the way for further development of CPMV nanoparticles for in vivo applications.


Assuntos
Comovirus/efeitos da radiação , Nanopartículas , Raios Ultravioleta , Inativação de Vírus , Comovirus/patogenicidade
10.
Chem Biol ; 14(10): 1152-62, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17961827

RESUMO

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.


Assuntos
Comovirus/efeitos dos fármacos , Ácido Fólico/farmacologia , Nanopartículas , Nanotecnologia/métodos , Polietilenoglicóis/farmacologia , Alcinos/química , Animais , Azidas/química , Western Blotting , Catálise , Linhagem Celular Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral/patologia , Cromatografia , Comovirus/fisiologia , Cobre/química , Ácido Fólico/química , Humanos , Ligantes , Microscopia , Polietilenoglicóis/química
11.
J Control Release ; 120(1-2): 41-50, 2007 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-17512998

RESUMO

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.


Assuntos
Comovirus , Portadores de Fármacos/farmacocinética , Portadores de Fármacos/toxicidade , Nanopartículas , Vírion , Animais , Portadores de Fármacos/administração & dosagem , Portadores de Fármacos/química , Compostos Heterocíclicos/química , Injeções Intravenosas , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Modelos Moleculares , Conformação Molecular , Compostos Organometálicos/química , Espectrofotometria/métodos , Baço/efeitos dos fármacos , Baço/metabolismo , Baço/patologia , Térbio/química
12.
Nat Med ; 12(3): 354-60, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16501571

RESUMO

A significant impediment to the widespread use of noninvasive in vivo vascular imaging techniques is the current lack of suitable intravital imaging probes. We describe here a new strategy to use viral nanoparticles as a platform for the multivalent display of fluorescent dyes to image tissues deep inside living organisms. The bioavailable cowpea mosaic virus (CPMV) can be fluorescently labeled to high densities with no measurable quenching, resulting in exceptionally bright particles with in vivo dispersion properties that allow high-resolution intravital imaging of vascular endothelium for periods of at least 72 h. We show that CPMV nanoparticles can be used to visualize the vasculature and blood flow in living mouse and chick embryos to a depth of up to 500 microm. Furthermore, we show that the intravital visualization of human fibrosarcoma-mediated tumor angiogenesis using fluorescent CPMV provides a means to identify arterial and venous vessels and to monitor the neovascularization of the tumor microenvironment.


Assuntos
Comovirus/isolamento & purificação , Diagnóstico por Imagem/instrumentação , Diagnóstico por Imagem/métodos , Endotélio Vascular/citologia , Nanoestruturas/análise , Animais , Artérias/citologia , Embrião de Galinha , Membrana Corioalantoide/irrigação sanguínea , Membrana Corioalantoide/citologia , Membrana Corioalantoide/ultraestrutura , Comovirus/química , Endotélio Vascular/virologia , Corantes Fluorescentes/análise , Corantes Fluorescentes/química , Humanos , Camundongos , Microcirculação , Nanoestruturas/química , Neoplasias/irrigação sanguínea , Neoplasias/patologia , Neovascularização Patológica , Polietilenoglicóis , Fatores de Tempo , Veias/citologia
13.
J Nanobiotechnology ; 4: 2, 2006 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-16476163

RESUMO

Specific targeting of tumor cells is an important goal for the design of nanotherapeutics for the treatment of cancer. Recently, viruses have been explored as nano-containers for specific targeting applications, however these systems typically require modification of the virus surface using chemical or genetic means to achieve tumor-specific delivery. Interestingly, there exists a subset of viruses with natural affinity for receptors on tumor cells that could be exploited for nanotechnology applications. For example, the canine parvovirus (CPV) utilizes transferrin receptors (TfRs) for binding and cell entry into canine as well as human cells. TfRs are over-expressed by a variety of tumor cells and are widely being investigated for tumor-targeted drug delivery. We explored whether the natural tropism of CPV to TfRs could be harnessed for targeting tumor cells. Towards this goal, CPV virus-like particles (VLPs) produced by expression of the CPV-VP2 capsid protein in a baculovirus expression system were examined for attachment of small molecules and delivery to tumor cells. Structural modeling suggested that six lysines per VP2 subunit are presumably addressable for bioconjugation on the CPV capsid exterior. Between 45 and 100 of the possible 360 lysines/particle could be routinely derivatized with dye molecules depending on the conjugation conditions. Dye conjugation also demonstrated that the CPV-VLPs could withstand conditions for chemical modification on lysines. Attachment of fluorescent dyes neither impaired binding to the TfRs nor affected internalization of the 26 nm-sized VLPs into several human tumor cell lines. CPV-VLPs therefore exhibit highly favorable characteristics for development as a novel nanomaterial for tumor targeting.

14.
Virology ; 343(2): 224-35, 2005 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-16185741

RESUMO

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.


Assuntos
Comovirus/ultraestrutura , Nanoestruturas/ultraestrutura , Administração Oral , Animais , Ácidos Carboxílicos , Comovirus/patogenicidade , Fabaceae/virologia , Feminino , Corantes Fluorescentes , Suco Gástrico/virologia , Técnicas In Vitro , Cinética , Camundongos , Camundongos Endogâmicos C57BL , Nanotecnologia , Distribuição Tecidual
15.
J Infect Dis ; 192(6): 1047-51, 2005 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-16107958

RESUMO

Successful postexposure treatment for inhalation anthrax is thought to include neutralization of anthrax toxin. The soluble anthrax toxin receptor/tumor endothelial marker 8 and capillary morphogenesis protein 2 (sATR/TEM8 and sCMG2, respectively) receptor decoys bind to anthrax toxin protective antigen (PA) and compete with cellular receptors for binding. Here, we show that, in a tissue-culture model of intoxication, sCMG2 is a 11.4-fold more potent antitoxin than sATR/TEM8 and that this increased activity corresponds to an approximately 1000-fold higher PA-binding affinity. Stoichiometric concentrations of sCMG2 protect rats against lethal toxin challenge, making sCMG2 one of the most effective anthrax antitoxins described to date.


Assuntos
Antígenos de Bactérias/metabolismo , Toxinas Bacterianas/antagonistas & inibidores , Toxinas Bacterianas/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Membrana/farmacologia , Receptores de Peptídeos/metabolismo , Animais , Antígenos de Bactérias/toxicidade , Toxinas Bacterianas/toxicidade , Células CHO , Cricetinae , Masculino , Ligação Proteica , Ratos , Ratos Endogâmicos F344 , Análise de Sobrevida , Fatores de Tempo
16.
Langmuir ; 21(6): 2098-103, 2005 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-15751992

RESUMO

Viruses are exemplary models in nanoassembly for their regular geometries, well characterized surface properties, and nanoscale dimensions. Armed with versatile tools aimed at site-directed mutagenesis to modify the virion's surface, conjugation chemistry for capsid coupling, and manipulation of nanoparticles, we have demonstrated nanoscale assembly of inorganic carbon nanotubes and quantum dots with engineered viruses to produce an intimate array of hybrid structures.


Assuntos
Comovirus/ultraestrutura , Compostos Inorgânicos/química , Nodaviridae/ultraestrutura , Compostos Orgânicos/química , Capsídeo/química , Comovirus/química , Comovirus/genética , Mutagênese Sítio-Dirigida , Nanotecnologia , Nodaviridae/química , Nodaviridae/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...