Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Mol Pharm ; 12(2): 342-50, 2015 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-25521817

RESUMO

To improve the efficiency of gene delivery for effective gene therapy, it is essential that the vector carries functional components that can promote overcoming barriers in various steps leading to the transport of DNA from extracellular to ultimately nuclear compartment. In this study, we designed genetically engineered fusion proteins as a platform to incorporate multiple functionalities in one chimeric protein. Prototypes of such a chimera tested here contain two domains: one that binds to DNA; the other that can facilitate endosomal escape of DNA. The fusion proteins are composed of listeriolysin O (LLO), the endosomolytic pore-forming protein from Listeria monocytogenes, and a 22 amino acid sequence of the DNA-condensing polypeptide protamine (PN), singly or as a pair: LLO-PN and LLO-PNPN. We demonstrate dramatic enhancement of the gene delivery efficiency of protamine-condensed DNA upon incorporation of a small amount of LLO-PN fusion protein and further improvement with LLO-PNPN in vitro using cultured cells. Additionally, the association of anionic liposomes with cationic LLO-PNPN/protamine/DNA complexes, yielding a net negative surface charge, resulted in better in vitro transfection efficiency in the presence of serum. An initial, small set of data in mice indicated that the observed enhancement in gene expression could also be applicable to in vivo gene delivery. This study suggests that incorporation of a recombinant fusion protein with multiple functional components, such as LLO-protamine fusion protein, in a nonviral vector is a promising strategy for various nonviral gene delivery systems.


Assuntos
Toxinas Bacterianas/química , Técnicas de Transferência de Genes , Proteínas de Choque Térmico/química , Proteínas Hemolisinas/química , Protaminas/química , Proteínas Recombinantes de Fusão/química
2.
Anal Biochem ; 411(2): 194-9, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21237129

RESUMO

A simple homogeneous assay for the detection of membrane permeabilization by antimicrobial peptides and synthetic copolymers is described. Liposomes encapsulating pyrroloquinoline quinone (PQQ), the prosthetic group of the apoenzyme glucose dehydrogenase (GDH), are used to detect membrane permeabilization by the antimicrobial peptides MSI-594 and MSI-78 as well as various synthetic antimicrobial copolymers in an optical microwell assay. PQQ-loaded liposomes and the peptide or copolymer are added to wells of a 96-well microtiter plate. If the integrity of the liposome is compromised, the PQQ encapsulated in the liposomes is released and available for activating the apoenzyme. The release of PQQ catalyzes a color change in the presence of apo-GDH, glucose, and the redox dye 1,6-dichlorophenol indophenol (DCPIP) that can be evaluated through a visual color change. For more quantitative measurements, the absorbance change over a 30min period was measured. The absorbance change is related to the activity and concentration for a given antimicrobial agent. Furthermore, by varying liposome compositions to include cholesterol, the potential toxicity of the peptide or polymer toward mammalian cells can be readily evaluated. The assay is simple and sensitive and will be useful for analyzing the membrane permeation/disruption properties of a host of antimicrobial peptides and synthetic polymers.


Assuntos
Peptídeos Catiônicos Antimicrobianos/química , Lipossomos/química , Cofator PQQ/química , Polímeros/química , Espectrofotometria/métodos , 2,6-Dicloroindofenol/química , Apoenzimas/metabolismo , Glucose 1-Desidrogenase/metabolismo , Cinética , Bicamadas Lipídicas/química , Oxirredução , Peptídeos/química , Permeabilidade/efeitos dos fármacos
3.
Eur J Pharm Biopharm ; 67(3): 752-8, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17560100

RESUMO

The therapeutic efficacy of prophalan-L, the L-proline prodrug of melphalan that demonstrated prolidase-dependent bioactivation to melphalan, was examined in vivo in a mouse melanoma model. Prophalan-L exhibited 2- to 2.5-fold higher hydrolytic and cytotoxic activity than prophalan-D, the D-analog, in B16-F10 murine melanoma cells in vitro. Prophalan-L cytotoxicity in B16-F10 cells was lower (GI50=221 microM) than that of melphalan (GI50=173 microM). The tumor growth profiles in C57BL/6J mice injected with B16-F10 cells and treated with melphalan (5.5 microg/g i.p.) and equimolar concentrations of the prodrugs demonstrated significant difference between the control (buffered saline) and melphalan or prophalan-L but no significant difference between control and prophalan-D or between melphalan and prophalan-L. Prophalan-L was significantly less toxic than melphalan, while no significant difference was observed in toxicity, measured as percent weight loss, between the prodrugs and saline control. Tumor reduction efficacy at high doses (12 microg/g i.p.) was similar for melphalan and prophalan-L; however, fatal toxicity was associated with melphalan while prophalan-L exhibited significantly lower systemic toxicity. An excellent correlation between GI50 and tumor reduction efficacy was observed for the tested drugs (r2=0.95). Prophalan-L thus demonstrates higher therapeutic index than melphalan in the murine melanoma model.


Assuntos
Antineoplásicos Alquilantes/uso terapêutico , Melanoma Experimental/tratamento farmacológico , Melfalan/análogos & derivados , Melfalan/uso terapêutico , Pró-Fármacos/uso terapêutico , Prolina/análogos & derivados , Animais , Antineoplásicos Alquilantes/metabolismo , Peso Corporal/efeitos dos fármacos , Calorimetria , Proliferação de Células/efeitos dos fármacos , Cromatografia Líquida de Alta Pressão , Feminino , Hidrólise , Indicadores e Reagentes , Melanoma Experimental/patologia , Melfalan/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Pró-Fármacos/metabolismo , Prolina/metabolismo , Prolina/uso terapêutico , Espectrofotometria Ultravioleta , Estereoisomerismo , Análise de Sobrevida
4.
Cell Microbiol ; 9(7): 1695-704, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17346313

RESUMO

Three proteins secreted by Listeria monocytogenes facilitate escape from macrophage vacuoles: the cholesterol-dependent cytolysin listeriolysin O (LLO), a phosphoinositide-specific phospholipase C (PI-PLC) and a broad-range phospholipase C (PC-PLC). LLO and PI-PLC can activate several members of the protein kinase C (PKC) family during infection. PKCepsilon is a novel PKC that contributes to macrophage activation, defence against bacterial infection, and phagocytosis; however, a role for PKCepsilon in Lm infections has not been described. To study PKCepsilon dynamics, PKCepsilon-YFP chimeras were visualized in macrophages during Lm infection. PKCepsilon-YFP was recruited to forming vacuoles during macrophage phagocytosis of Lm and again later to fully formed Lm vacuoles. The PKCepsilon-YFP localization to the fully formed Lm vacuole was LLO-dependent but independent of PI-PLC or PC-PLC. PKCepsilon-YFP recruitment often followed LLO perforation of the membrane, as indicated by localization of PKCepsilon-YFP to Lm vacuoles after they released small fluorescent dyes into the cytoplasm. PKCepsilon-YFP recruitment to vesicles also followed phagocytosis of LLO-containing liposomes or osmotic lysis of endocytic vesicles, indicating that vacuole perforation by LLO was the chief cause of the PKCepsilon response. These studies implicate PKCepsilon in a cellular mechanism for recognizing damaged membranous organelles, including the disrupted vacuoles created when Lm escapes into cytoplasm.


Assuntos
Listeria monocytogenes/patogenicidade , Macrófagos/enzimologia , Macrófagos/microbiologia , Proteína Quinase C-épsilon/metabolismo , Vacúolos/enzimologia , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/toxicidade , Linhagem Celular , Proteínas de Choque Térmico/metabolismo , Proteínas de Choque Térmico/toxicidade , Proteínas Hemolisinas/metabolismo , Proteínas Hemolisinas/toxicidade , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Macrófagos/imunologia , Macrófagos/ultraestrutura , Camundongos , Microscopia de Fluorescência , Proteína Quinase C-épsilon/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
5.
J Immunol ; 176(4): 2095-104, 2006 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-16455965

RESUMO

Increased monocyte/macrophage (Mphi) apoptosis occurs in patients with systemic lupus erythematosus (SLE) and is mediated, at least in part, by an autoreactive CD4(+) T cell subset. Furthermore, autoreactive murine CD4(+) T cells that kill syngeneic Mphi in vitro induce a lupus-like disease in vivo. However, it is unclear whether increased Mphi apoptosis in SLE per se is sufficient to accelerate/promote autoimmunity. We have investigated whether increased Mphi apoptosis in vivo, induced by the administration of clodronate liposomes, can exacerbate the autoimmune phenotype in NZB x SWR (SNF(1)) lupus-prone mice, and induce autoantibody production in haplotype-matched BALB/c x DBA1 (DBF(1)) non-lupus-prone mice. Lupus-prone mice SNF(1) mice that were treated with clodronate liposomes, but not mice treated with vehicle, developed significant increases in autoantibodies to dsDNA, nucleosomes, and the idiotypically related family of nephritic Abs Id(LN)F(1), when compared with untreated SNF(1) mice. Furthermore, clodronate treatment hastened the onset of proteinuria and worsened SNF(1) lupus nephritis. When compared with vehicle-treated controls, clodronate-treated non-lupus-prone DBF(1) mice developed significantly higher levels of anti-nucleosome and Id(LN)F(1) Abs but did not develop lupus nephritis. We propose that Mphi apoptosis contributes to the pathogenesis of autoantibody formation and organ damage through both an increase in the apoptotic load and impairment in the clearance of apoptotic material. This study suggests that mechanisms that induce scavenger cell apoptosis, such as death induced by autoreactive cytotoxic T cells observed in SLE, could play a pathogenic role and contribute to the severity of the disease.


Assuntos
Formação de Anticorpos/imunologia , Apoptose , Autoanticorpos/biossíntese , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/patologia , Macrófagos/citologia , Animais , Autoanticorpos/imunologia , Feminino , Rim/patologia , Lipossomos , Macrófagos/imunologia , Camundongos , Baço/patologia
6.
Mol Pharm ; 2(1): 74-82, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15804180

RESUMO

Delivery of antigenic protein to the cytosol of antigen-presenting cells (APCs), such as macrophages (MPhi) and dendritic cells (DCs), is required for an efficient CD8 T-cell-mediated immune response. We have previously shown that co-encapsulation of antigenic protein inside pH-sensitive liposomes with listeriolysin O (LLO), a pore-forming protein of Listeria monocytogenes, generates efficient major histocompatibility complex class I (MHC I)-restricted immune responses both in vitro and in vivo. In this study, we sought to analyze the relative efficiency of LLO-mediated cytosolic delivery of liposomal antigen in two important APCs, macrophages and dendritic cells, by examining the sequential steps involved in antigen presentation to T-cells in cultured mouse bone marrow-derived MPhis (BMMPhis) and DCs (BMDCs). BMMPhis overall presented liposomal antigen better than BMDCs at a given concentration of liposomal antigen incubated with cells, and the trend was also observed after the presentation was normalized by the uptake of antigen. When soluble antigen was directly introduced into the cytosol, however, BMDCs presented the antigen more efficiently than BMMPhis. In addition, when the APCs were externally loaded with the antigenic peptide of the protein, BMDCs displayed a higher level of cell surface MHC I-peptide complexes and presented the peptide more efficiently than BMMPhis. These results combined together suggest that LLO-mediated release of liposomal antigen from the endosomal/lysosomal compartment may be more pronounced in BMMPhis than in BMDCs, and further implicates differential activity of LLO and varying efficiency of LLO-mediated endosomal escape in different antigen-presenting cell types.


Assuntos
Toxinas Bacterianas , Citosol/imunologia , Células Dendríticas/imunologia , Proteínas de Choque Térmico , Lipossomos , Macrófagos/imunologia , Animais , Apresentação de Antígeno , Células Apresentadoras de Antígenos/imunologia , Células Cultivadas , Sistemas de Liberação de Medicamentos , Feminino , Citometria de Fluxo , Proteínas Hemolisinas , Hibridomas , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Recombinantes/administração & dosagem , Linfócitos T/imunologia
7.
Proc Natl Acad Sci U S A ; 101(31): 11386-91, 2004 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-15269347

RESUMO

Innate and adaptive immunity depends critically on host recognition of pathogen-associated molecules. Toll-like receptors (TLRs) are key mediators of pathogen surveillance at the cell or phagocytic vacuole surface. However, mechanisms underlying recognition of pathogens in other cellular compartments remain unclear, and responses elicited by cytosolic challenge are poorly characterized. We therefore used mouse cDNA microarrays to investigate gene expression triggered by infection of bone marrow-derived macrophages with cytosol- and vacuole-localized Listeria monocytogenes (Lm), a model cytosolic pathogen. The resulting gene expression program included two basic categories of induced genes: an "early/persistent" cluster consistent with NF-kappaB-dependent responses downstream of TLRs, and a subsequent "late response" cluster largely composed of IFN-responsive genes (IRGs). The early/persistent cluster was observed upon infection with WT, heat-killed, or mutant Lm lacking listeriolysin O, the pore-forming hemolysin that promotes escape from phagocytic vacuoles. However, the IRG cluster depended on entry of WT Lm into the cytosol. Infection with listeriolysin O-expressing, cytosolic Bacillus subtilis (Bs) strikingly recapitulated the expression profile associated with WT Lm, including IRG induction. IRG up-regulation was associated with MyD88-independent induction of IFN-beta transcription and activity. Whereas Staphylococcus aureus (Sa) lipoteichoic acid treatment confirmed that many late-response genes could also be stimulated through TLRs, our study identified a cytosol-specific transcriptional program independent of TLR signaling through MyD88. Further characterization of cytosolic surveillance pathway(s) and their points of convergence with TLR- and IFN-dependent pathways will enhance our understanding of the means by which mammals detect and respond to pathogens.


Assuntos
Regulação Bacteriana da Expressão Gênica/fisiologia , Listeria monocytogenes/genética , Listeriose/microbiologia , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas Adaptadoras de Transdução de Sinal , Animais , Antígenos de Diferenciação/genética , Bacillus subtilis/genética , Ciclo Celular/fisiologia , Citosol/microbiologia , Citosol/fisiologia , Feminino , Interferons/fisiologia , Macrófagos/microbiologia , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Mutantes , Fator 88 de Diferenciação Mieloide , NF-kappa B/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores Imunológicos/genética , Staphylococcus aureus/genética , Receptores Toll-Like , Transcrição Gênica/fisiologia , Vacúolos/microbiologia , Vacúolos/fisiologia
8.
Mol Pharm ; 1(1): 2-8, 2004 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-15832496

RESUMO

Cytotoxic T lymphocytes (CTLs) are capable of conferring protection against intracellular pathogens and tumor. Protective antiviral immunity, mediated by the activation of antigenic epitope-specific CTL, can be achieved by delivering exogenous antigen into the cytosol of antigen-presenting cells. Cytosolic introduction of vaccine antigen, however, requires a specialized delivery strategy due to the membrane barrier limiting the access of macromolecules to the cytosol. In this study, we have investigated the potential ability of listeriolysin O-containing liposomes (LLO-liposomes) to deliver lymphocytic choriomeningitis virus (LCMV) nucleoprotein (NP), harnessing the intracellular invasion mechanism of Listeria monocytogenes, to stimulate a NP-specific CTL response. We have analyzed the ability of LLO-liposomes to induce an enhanced CTL response and determined the extent of CTL-mediated protection using an in vivo infection model. Mice immunized with LLO-liposomes containing NP generated a higher frequency of NP-specific CD8+ T cells with greater effector activity than the control groups immunized with either non-LLO-liposomal NP or LLO-liposomes containing control protein. Moreover, LLO-liposomal NP-immunized mice were completely protected against a lethal intracerebral challenge with a virulent strain of LCMV and were capable of clearing a chronic LCMV infection. Our study demonstrates that LLO-liposomes can be used as an efficient vaccine delivery system carrying a viral antigenic protein to generate protective antiviral immunity.


Assuntos
Toxinas Bacterianas/farmacologia , Toxinas Bacterianas/farmacocinética , Proteínas de Choque Térmico/farmacologia , Proteínas de Choque Térmico/farmacocinética , Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Nucleoproteínas/farmacocinética , Linfócitos T Citotóxicos/imunologia , Proteínas Virais/farmacocinética , Animais , Portadores de Fármacos , Feminino , Proteínas Hemolisinas , Imunidade , Lipossomos , Ativação Linfocitária , Coriomeningite Linfocítica/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Linfócitos T Citotóxicos/efeitos dos fármacos , Proteínas Virais/imunologia
9.
Pharm Res ; 20(8): 1239-48, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12948022

RESUMO

PURPOSE: CTLA4Ig, a fusion protein of CTLA-4 and Fc of immunoglobulin (Ig) heavy chain, inhibits the essential costimulatory signal for full T cell activation via blocking the interaction between CD28 and B7 molecules and renders T cell nonresponsiveness. CTLA4Ig has been used to control deleterious T cell activation in many experimental systems. We hypothesized that by conjugating CTLA4Ig to liposomes the efficacy of CTLA4Ig could be enhanced through multivalent ligand effect, superior targetability, and modification of the fate of ligated costimulatory molecules. METHODS AND RESULTS: Consistent with this hypothesis, liposome-conjugated CTLA4Ig bound to B7 and blocked their binding sites more efficiently than free CTLA4Ig, lowering the half maximal dose for B7 blocking by an order of the magnitude. These results were similar both in B7-1 expressing p815 cells and in activated macrophages. Moreover, CTLA4Ig-liposomes underwent rapid internalization upon cell surface binding through B7 molecules. In allogenic mixed lymphocyte reaction assays, the CTLA4Ig-liposomes were tested to show effective inhibition of T cell proliferation. In vivo, however, when CTLA4Ig-liposomes were injected into mice, a significant fraction was localized to the reticuloendothelial system (RES), presumably because of its binding to Fc receptors expressed on tissue macrophages. The Fc receptor-mediated uptake could be alleviated by coinjection of anti-FcR monoclonal antibody. In the mouse engrafted with pancreatic islets of Langerhans underneath the capsule of one kidney, despite the increased localization in RES, enhanced accumulation of CTLA4Ig-conjugated liposome was observed in the engrafted kidney compared to the contralateral kidney. CONCLUSION: We show that the conjugation of CTLA4Ig to liposome could increase the efficiency of the targeting by increasing the binding avidity at cellular level and by increasing the concentration at the target site in in vivo system. The biodistribution and circulation time data suggested that the CTLA4Ig-liposomes could be improved upon minimizing the FcR-mediated uptake by Fc receptor-bearing cells. Thus, the strategy of conjugating CTLA4Ig to liposomes could be exploited for immune intervention in transplantation and autoimmune diseases for the efficient blocking of costimulation.


Assuntos
Células Apresentadoras de Antígenos/metabolismo , Antígeno B7-1/metabolismo , Imunoconjugados/farmacocinética , Abatacepte , Animais , Células da Medula Óssea/metabolismo , Células Cultivadas , Regulação para Baixo , Imunoconjugados/administração & dosagem , Técnicas In Vitro , Injeções Intraperitoneais , Transplante das Ilhotas Pancreáticas , Lipossomos , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Fatores de Tempo , Distribuição Tecidual
10.
J Biol Chem ; 278(37): 35102-8, 2003 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-12832408

RESUMO

Gelonin is a type I plant toxin that has potential as an effective anti-tumor agent by virtue of its enzymatic capacity to inactivate ribosomes and arrest protein synthesis, thereby effectively limiting the growth of cancer cells. Being a hydrophilic macromolecule, however, gelonin has limited access to its target subcellular compartment, the cytosol; it is effectively plasma membrane-impermeant and subject to rapid degradation within endosomes and lysosomes upon cellular uptake as it lacks the membrane-translocating capability that is typically provided by a disulfide-linked B polypeptide found in the type II toxins (e.g. ricin). These inherent characteristics generate the need for the development of a specialized cytosolic delivery strategy for gelonin as an effective anti-tumor therapeutic agent. Here we describe an efficient means of delivering gelonin to the cytosol of B16 melanoma cells. Gelonin was co-encapsulated inside pH-sensitive liposomes with listeriolysin O, the pore-forming protein that mediates escape of the intracellular pathogen Listeria monocytogenes from the endosome into the cytosol. In in vitro experiments, co-encapsulated listeriolysin O enabled liposomal gelonin-mediated B16 cell killing with a gelonin IC50 of approximately 0.1 nM with an extreme efficiency requiring an incubation time of only 1 h. By contrast, cells treated with equivalent concentrations of unencapsulated gelonin or gelonin encapsulated alone in pH-sensitive liposomes exhibited no detectable cytotoxicity. Moreover, treatment by direct intratumor injection into subcutaneous solid tumors of B16 melanoma in a mouse model showed that pH-sensitive liposomes containing both listeriolysin O and gelonin were more effective than control formulations in curtailing tumor growth rates.


Assuntos
Sobrevivência Celular/efeitos dos fármacos , Proteínas de Choque Térmico/toxicidade , Lipossomos , Melanoma Experimental/tratamento farmacológico , Proteínas de Plantas/farmacocinética , Proteínas de Plantas/toxicidade , Animais , Antineoplásicos Fitogênicos/farmacocinética , Antineoplásicos Fitogênicos/toxicidade , Toxinas Bacterianas/toxicidade , Transporte Biológico , Portadores de Fármacos , Proteínas Hemolisinas , Concentração de Íons de Hidrogênio , Lipossomos/síntese química , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Inativadoras de Ribossomos Tipo 1 , Células Tumorais Cultivadas
11.
J Biol Chem ; 278(28): 25348-56, 2003 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-12732646

RESUMO

Valacyclovir is the 5'-valyl ester prodrug of acyclovir, an effective anti-herpetic drug. Systemic availability of acyclovir in humans is three to five times higher when administered orally as the prodrug. The increased bioavailability of valacyclovir is attributed to carrier-mediated intestinal absorption, via the hPEPT1 peptide transporter, followed by the rapid and complete conversion to acyclovir. The one or more human enzymes responsible for in vivo activation of the prodrug to the active drug and its conversion sites, however, have not been identified. In this report, we describe the purification, identification, and characterization of a human enzyme that activates valacyclovir to acyclovir. A protein with significant hydrolytic activity toward valacyclovir, the 5'-glycyl ester of acyclovir, and the 5'-valyl ester of zidovudine (AZT), was purified from Caco-2 cells derived from human intestine. Using a non-redundant data base search, the N-terminal 19-amino acid sequence of the purified 27-kDa, basic protein revealed a perfect match within the N terminus of a serine hydrolase, Biphenyl hydrolase-like (BPHL, gi:4757862) protein, previously cloned from human breast carcinoma. Recombinant BPHL exhibited significant hydrolytic activity for both valacyclovir and valganciclovir with specificity constants (kcat/Km), 420 and 53.2 mm-1.s-1, respectively. We conclude that BPHL may be an important enzyme activating valacyclovir and valganciclovir in humans and an important new target for prodrug design.


Assuntos
Aciclovir/análogos & derivados , Aciclovir/química , Hidrolases de Éster Carboxílico/química , Hidrolases de Éster Carboxílico/farmacologia , Hidrolases/química , Valina/análogos & derivados , Valina/química , Sequência de Aminoácidos , Western Blotting , Linhagem Celular , Relação Dose-Resposta a Droga , Eletroforese em Gel de Poliacrilamida , Humanos , Concentração de Íons de Hidrogênio , Hidrólise , Mucosa Intestinal/metabolismo , Cinética , Modelos Químicos , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Homologia de Sequência de Aminoácidos , Valaciclovir
12.
Proc Natl Acad Sci U S A ; 99(21): 13861-6, 2002 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-12359878

RESUMO

Host recognition of bacterial pathogens is a critical component of the immune response. Intracellular bacterial pathogens are able to evade the humoral immune system by residing within the host cell. Here we show the existence of an innate host surveillance mechanism in macrophages that specifically distinguishes bacteria in the cytosol from bacteria in the vacuole. Recognition of Gram-positive and Gram-negative bacterial products by this surveillance system results in transcription of the ifnb gene. The activation of cytosol-specific signaling is associated with translocation of NF-kappaB into the nucleus and phosphorylation of the p38 mitogen-activated protein (MAP) kinase. Activation of the p38 kinase is required for the induction of gene expression by the cytosolic surveillance pathway. Our studies suggest that infection by intracellular bacterial pathogens results in an immune response distinct from that of infection by extracellular bacterial pathogens.


Assuntos
Toxinas Bacterianas , Citosol/imunologia , Citosol/microbiologia , Bactérias Gram-Negativas/imunologia , Bactérias Gram-Positivas/imunologia , Macrófagos/imunologia , Macrófagos/microbiologia , Animais , Bacillus subtilis/imunologia , Células Cultivadas , Citosol/metabolismo , Ativação Enzimática , Escherichia coli/imunologia , Regulação da Expressão Gênica , Células HeLa , Proteínas de Choque Térmico/imunologia , Proteínas Hemolisinas , Humanos , Interferon beta/genética , Listeria monocytogenes/imunologia , Macrófagos/metabolismo , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Fosforilação , Transdução de Sinais , Vacúolos/imunologia , Vacúolos/microbiologia , Proteínas Quinases p38 Ativadas por Mitógeno
13.
Biochim Biophys Acta ; 1563(1-2): 7-17, 2002 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-12007619

RESUMO

Cytotoxic T lymphocytes (CTLs) are primed by peptide antigens that are endogenously processed in the cytosol and presented in the context of major histocompatibility complex I (MHC I) molecules of antigen-presenting cells (APCs). Exogenous soluble protein antigens do not gain efficient entry into the cytosol of APCs, and therefore requires a special cytosolic delivery method. We have developed such a delivery strategy adopting the well-elucidated cytosol-invading listerial endosomal escape mechanism, and report here an efficient delivery of exogenous whole protein antigen into the cytosol in a mouse model. Co-encapsulation of listeriolysin O (LLO) inside liposome (LLO-liposome) was required for delivery of ovalbumin (OVA) into the cytosol of APCs in primary cultures. LLO-liposome-mediated OVA immunization in mice engendered significantly higher OVA-specific CTL activity and increased antigenic peptide-specific CTL precursor (CTLp) frequency as compared to non-LLO-liposome or soluble OVA immunizations. Interferon-gamma (IFN-gamma) production upon specific stimulation by MHC I-restricted peptide was also significantly stronger by the inclusion of LLO in the liposomes. Rerouting of antigen into the cytosol by LLO-liposomes, however, did not reduce the extent of anti-OVA antibody responses. Moreover, LLO-liposome-antigen vaccination was robust in conferring protection to mice from lethal challenges with antigen-expressing tumor cells. Our study demonstrates a novel delivery system for efficient introduction of exogenous protein into the cytosol in vivo, priming cellular immune responses, which are protective in nature.


Assuntos
Toxinas Bacterianas , Proteínas de Choque Térmico/administração & dosagem , Linfócitos T Citotóxicos/imunologia , Animais , Células Apresentadoras de Antígenos/imunologia , Vacinas Anticâncer/administração & dosagem , Células Cultivadas , Citosol/imunologia , Sistemas de Liberação de Medicamentos , Proteínas do Ovo/administração & dosagem , Proteínas do Ovo/imunologia , Proteínas do Ovo/farmacologia , Proteínas Hemolisinas , Imunização , Interferon gama/metabolismo , Interleucina-2/metabolismo , Lipossomos , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Ovalbumina/administração & dosagem , Ovalbumina/imunologia , Ovalbumina/farmacologia , Fragmentos de Peptídeos , Proteínas Recombinantes/administração & dosagem , Linfócitos T Citotóxicos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA