Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Vaccine ; 35(21): 2840-2847, 2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28413132

RESUMO

Respiratory syncytial virus (RSV) is a massive medical burden in infants, children and the elderly worldwide, and an effective, safe RSV vaccine remains an unmet need. Here we assess a novel vaccination strategy based on the intradermal delivery of a SynCon® DNA-based vaccine encoding engineered RSV-F antigen using a surface electroporation device (SEP) to target epidermal cells, in clinically relevant experimental models. We demonstrate the ability of this strategy to elicit robust immune responses. Importantly we demonstrate complete resistance to pulmonary infection at a single low dose of vaccine in the cotton rat RSV/A challenge model. In contrast to the formalin-inactivated RSV (FI-RSV) vaccine, there was no enhanced lung inflammation upon virus challenge after DNA vaccination. In summary the data presented outline the pre-clinical development of a highly efficacious, tolerable and safe non-replicating vaccine delivery strategy.


Assuntos
Eletroporação/instrumentação , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Vírus Sincicial Respiratório/administração & dosagem , Vacinas contra Vírus Sincicial Respiratório/imunologia , Vacinas de DNA/administração & dosagem , Vacinas de DNA/imunologia , Animais , Modelos Animais de Doenças , Feminino , Pulmão/patologia , Sigmodontinae , Resultado do Tratamento
2.
Vaccine ; 35(1): 61-70, 2017 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-27894716

RESUMO

The skin is an ideal target tissue for vaccine delivery for a number of reasons. It is highly accessible, and most importantly, enriched in professional antigen presenting cells. Possessing strong similarities to human skin physiology and displaying a defined epidermis, the guinea pig is an appropriate model to study epidermal delivery of vaccine. However, whilst we have characterized the humoral responses in the guinea pig associated with skin vaccine protocols we have yet to investigate the T cell responses. In response to this inadequacy, we developed an IFN-γ ELISpot assay to characterize the cellular immune response in the peripheral blood of guinea pigs. Using a nucleoprotein (NP) influenza pDNA vaccination regimen, we characterized host T cell responses. After delivery of the DNA vaccine to the guinea pig epidermis we detected robust and rapid T cell responses. The levels of IFN-γ spot-forming units averaged approximately 5000 per million cells after two immunizations. These responses were broad in that multiple regions across the NP antigen elicited a T cell response. Interestingly, we identified a number of NP immunodominant T cell epitopes to be conserved across an outbred guinea pig population, a phenomenon which was also observed after immunization with a RSV DNA vaccine. We believe this data enhances our understanding of the cellular immune response elicited to a vaccine in guinea pigs, and globally, will advance the use of this model for vaccine development, especially those targeting skin as a delivery site.


Assuntos
Eletroporação , Vacinas contra Influenza/imunologia , Proteínas de Ligação a RNA/imunologia , Pele/imunologia , Linfócitos T/imunologia , Vacinas de DNA/imunologia , Proteínas do Core Viral/imunologia , Animais , ELISPOT , Feminino , Cobaias , Vacinas contra Influenza/administração & dosagem , Interferon gama/metabolismo , Proteínas do Nucleocapsídeo , Vacinas de DNA/administração & dosagem
3.
Clin Pharmacol Drug Dev ; 5(2): 91-101, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27138022

RESUMO

VGX-1027, a novel oral immune modulator, is under development for the treatment of rheumatoid arthritis. The safety, tolerability, and pharmacokinetics of single (1-800 mg) and multiple (40-400 mg) oral doses were evaluated in 2 clinical studies. The doses were well tolerated up to 800 mg in a single dose and 200 mg twice daily in multiple doses. Adverse events were mild to moderate in severity with no identifiable dose-related pattern. There were no clinically significant physical or laboratory findings. The pharmacokinetic data indicated that increases in Cmax and AUC0-inf were dose-proportional, and AUC0- τ was approximately dose-proportional. For the single-dose study, median Tmax ranged from 0.5 to 2 hours and mean t1/2 ranged from 4.9 to 8.7 hours. For the multiple-dose study, median Tmax ranged from 0.5 to 2.0 hours and mean t1/2 ranged from 7.05 to 10.05 hours. No accumulation of the drug was observed after day 1, indicating that steady-state concentrations were attained with single and multiple dosing for 5 days. Approximately 90% of the administered dose was excreted in urine as unchanged drug.


Assuntos
Acetatos/administração & dosagem , Anti-Inflamatórios/administração & dosagem , Fatores Imunológicos/administração & dosagem , Oxazóis/administração & dosagem , Acetatos/efeitos adversos , Acetatos/farmacocinética , Administração Oral , Adulto , Anti-Inflamatórios/efeitos adversos , Anti-Inflamatórios/farmacocinética , Área Sob a Curva , Disponibilidade Biológica , Relação Dose-Resposta a Droga , Método Duplo-Cego , Feminino , Meia-Vida , Humanos , Fatores Imunológicos/efeitos adversos , Fatores Imunológicos/farmacocinética , Masculino , Oxazóis/efeitos adversos , Oxazóis/farmacocinética
4.
J Infect Dis ; 214(3): 369-78, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27001960

RESUMO

BACKGROUND: Vaccination and passive antibody therapies are critical for controlling infectious diseases. Passive antibody administration has limitations, including the necessity for purification and multiple injections for efficacy. Vaccination is associated with a lag phase before generation of immunity. Novel approaches reported here utilize the benefits of both methods for the rapid generation of effective immunity. METHODS: A novel antibody-based prophylaxis/therapy entailing the electroporation-mediated delivery of synthetic DNA plasmids encoding biologically active anti-chikungunya virus (CHIKV) envelope monoclonal antibody (dMAb) was designed and evaluated for antiviral efficacy, as well as for the ability to overcome shortcomings inherent with conventional active vaccination and passive immunotherapy. RESULTS: One intramuscular injection of dMAb produced antibodies in vivo more rapidly than active vaccination with an anti-CHIKV DNA vaccine. This dMAb neutralized diverse CHIKV clinical isolates and protected mice from viral challenge. Combination of dMAb and the CHIKV DNA vaccine afforded rapid and long-lived protection. CONCLUSIONS: A DNA-based dMAb strategy induced rapid protection against an emerging viral infection. This method can be combined with DNA vaccination as a novel strategy to provide both short- and long-term protection against this emerging infectious disease. These studies have implications for pathogen treatment and control strategies.


Assuntos
Anticorpos Antivirais/imunologia , Quimioprevenção/métodos , Febre de Chikungunya/prevenção & controle , Vacinas de DNA/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Antivirais/administração & dosagem , Modelos Animais de Doenças , Eletroporação , Injeções Intramusculares , Camundongos Endogâmicos BALB C , Fatores de Tempo , Resultado do Tratamento , Vacinas de DNA/administração & dosagem , Vacinas Virais/administração & dosagem
5.
NPJ Vaccines ; 1: 16021, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-29263859

RESUMO

Significant concerns have been raised owing to the rapid global spread of infection and disease caused by the mosquito-borne Zika virus (ZIKV). Recent studies suggest that ZIKV can also be transmitted sexually, further increasing the exposure risk for this virus. Associated with this spread is a dramatic increase in cases of microcephaly and additional congenital abnormalities in infants of ZIKV-infected mothers, as well as a rise in the occurrence of Guillain Barre' syndrome in infected adults. Importantly, there are no licensed therapies or vaccines against ZIKV infection. In this study, we generate and evaluate the in vivo efficacy of a novel, synthetic, DNA vaccine targeting the pre-membrane+envelope proteins (prME) of ZIKV. Following initial in vitro development and evaluation studies of the plasmid construct, mice and non-human primates were immunised with this prME DNA-based immunogen through electroporation-mediated enhanced DNA delivery. Vaccinated animals were found to generate antigen-specific cellular and humoral immunity and neutralisation activity. In mice lacking receptors for interferon (IFN)-α/ß (designated IFNAR-/-) immunisation with this DNA vaccine induced, following in vivo viral challenge, 100% protection against infection-associated weight loss or death in addition to preventing viral pathology in brain tissue. In addition, passive transfer of non-human primate anti-ZIKV immune serum protected IFNAR-/- mice against subsequent viral challenge. This study in NHP and in a pathogenic mouse model supports the importance of immune responses targeting prME in ZIKV infection and suggests that additional research on this vaccine approach may have relevance for ZIKV control and disease prevention in humans.

6.
Sci Transl Med ; 7(301): 301ra132, 2015 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-26290414

RESUMO

First identified in 2012, Middle East respiratory syndrome (MERS) is caused by an emerging human coronavirus, which is distinct from the severe acute respiratory syndrome coronavirus (SARS-CoV), and represents a novel member of the lineage C betacoronoviruses. Since its identification, MERS coronavirus (MERS-CoV) has been linked to more than 1372 infections manifesting with severe morbidity and, often, mortality (about 495 deaths) in the Arabian Peninsula, Europe, and, most recently, the United States. Human-to-human transmission has been documented, with nosocomial transmission appearing to be an important route of infection. The recent increase in cases of MERS in the Middle East coupled with the lack of approved antiviral therapies or vaccines to treat or prevent this infection are causes for concern. We report on the development of a synthetic DNA vaccine against MERS-CoV. An optimized DNA vaccine encoding the MERS spike protein induced potent cellular immunity and antigen-specific neutralizing antibodies in mice, macaques, and camels. Vaccinated rhesus macaques seroconverted rapidly and exhibited high levels of virus-neutralizing activity. Upon MERS viral challenge, all of the monkeys in the control-vaccinated group developed characteristic disease, including pneumonia. Vaccinated macaques were protected and failed to demonstrate any clinical or radiographic signs of pneumonia. These studies demonstrate that a consensus MERS spike protein synthetic DNA vaccine can induce protective responses against viral challenge, indicating that this strategy may have value as a possible vaccine modality against this emerging pathogen.


Assuntos
Coronavírus da Síndrome Respiratória do Oriente Médio/imunologia , Vacinas de DNA/uso terapêutico , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Camelus , Macaca mulatta , Camundongos
7.
PLoS One ; 10(8): e0136323, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26301899

RESUMO

REDD1 is a highly conserved stress response protein that is upregulated following many types of cellular stress, including hypoxia, DNA damage, energy stress, ER stress, and nutrient deprivation. Recently, REDD1 was shown to be involved in dexamethasone induced autophagy in murine thymocytes. However, we know little of REDD1's function in mature T cells. Here we show for the first time that REDD1 is upregulated following T cell stimulation with PHA or CD3/CD28 beads. REDD1 knockout T cells exhibit a defect in proliferation and cell survival, although markers of activation appear normal. These findings demonstrate a previously unappreciated role for REDD1 in T cell function.


Assuntos
Autofagia/genética , Proliferação de Células/genética , Sobrevivência Celular/genética , Fatores de Transcrição/genética , Animais , Autofagia/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Dano ao DNA/genética , Dexametasona/administração & dosagem , Camundongos , Camundongos Knockout , Linfócitos T/metabolismo , Timócitos/metabolismo , Timócitos/patologia , Fatores de Transcrição/metabolismo , Ativação Transcricional/efeitos dos fármacos
8.
Immunology ; 142(4): 594-602, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24527796

RESUMO

VGX-1027 [(S,R)-3-phenyl-4,5-dihydro-5-isoxasole acetic acid] is a small molecule compound with immunomodulatory properties, which favourably influences the development of immuno-inflammatory and autoimmune diseases in different animal models such as type 1 diabetes mellitus, pleurisy, rheumatoid arthritis and inflammatory bowel disease. However, the precise mechanism of action of VGX-1027 remains to be ascertained. With this aim, we have studied the immunomodulatory effects of VGX-1027 in vitro, using a genome-wide oligonucleotide microarray approach, and in vivo, using the NZB/NZW F1 model of systemic lupus erythematosus. Microarray data revealed that the administration of VGX-1027 profoundly affected the immune response to exogenous antigens, by modulating the expression of genes that are primarily involved in antigen processing and presentation as well as genes that regulate immune activation. When administered in vivo VGX-1027 ameliorated the course of the disease in the NZB/NZW F1 mice, which correlated with higher per cent survival and improved clinical and histopathological signs. The data presented herein support the theory that VGX-1027 modulates immunity, probably by inhibiting inflammatory antigen presentation and so limiting immune cell expansion.


Assuntos
Acetatos/farmacologia , Fatores Imunológicos/farmacologia , Lipopolissacarídeos/toxicidade , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Oxazóis/farmacologia , Receptor 4 Toll-Like/imunologia , Animais , Modelos Animais de Doenças , Feminino , Humanos , Lúpus Eritematoso Sistêmico/induzido quimicamente , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/patologia , Masculino , Camundongos
9.
J Vasc Surg ; 59(3): 786-93, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23850058

RESUMO

OBJECTIVE: Hypoxia-inducible factor-1 alpha (HIF-1α) is a transcription factor that stimulates angiogenesis during tissue ischemia. In vivo electroporation (EP) enhances tissue DNA transfection. We hypothesized that in vivo EP of plasmid DNA encoding a constitutively expressed HIF-1α gene enhances neovascularization compared with intramuscular (IM) injection alone. METHODS: Left femoral artery ligation was performed in mice assigned to three groups: (1) HIF-EP (n = 13); (2) HIF-IM (n = 14); and (3) empty plasmid (pVAX)-EP (n = 12). A single dose of HIF-1α or pVAX DNA (20 µL of 5 µg/µL each) was injected into the ischemic adductor muscle followed by EP (groups one and three). Mice in group two received IM injection of HIF-1α plasmid DNA alone. From preligation to days 0, 3, 7, 14, and 21 postligation, limb perfusion recovery quantified by laser Doppler perfusion imager, limb function, and limb necrosis were measured. On day 21, the surviving mice (4-5 per group) were sacrificed and adductor muscle tissues stained for necrosis using hematoxylin and eosin, capillary density (anti-CD31 antibodies), and collateral vessels via anti-α-smooth muscle actin antibodies. RESULTS: In vivo EP of HIF-1α DNA significantly improved limb perfusion (HIF-EP: 1.03 ± 0.15 vs HIF-IM: 0.78 ± 0.064; P < .05, vs pVAX-EP: 0.41 ± 0.019; P < .001), limb functional recovery (HIF-EP: 3.5 ± 0.58 vs HIF-IM, 2.4 ± 1.14; P < .05, vs pVAX-EP: 2.4 ± 1.14; P < .001), and limb autoamputation on day 21 (HIF-EP: 77% ± 12% vs HIF-IM: 43% ± 14%; P < .05 vs pVAX-EP: 17% ± 11%; P < .01). Adductor muscle tissue necrosis decreased (HIF-EP: 20.7% ± 1.75% vs HIF-IM: 44% ± 3.73; P < .001, vs pVAX-EP: 60.05% ± 2.17%; P < .0001), capillary density increased (HIF-EP: 96.83 ± 5.72 vessels/high-powered field [hpf] vs HIF-IM: 62.87 ± 2.0 vessels/hpf; P < .001, vs pVAX-EP: 39.37 ± 2.76 vessels/hpf; P < .0001), collateral vessel formation increased (HI-EP: 76.33 ± 1.94 vessels/hpf vs HIF-IM: 37.5 ± 1.56 vessels/hpf; P < .0001, vs pVAX-EP: 18.5 ± 1.34 vessels/hpf; P < .00001), and the vessels were larger (HIF-EP: 15,521.67 ± 1298.16 µm(2) vs HIF-IM: 7788.87 ± 392.04 µm(2); P < .001 vs pVAX-EP: 4640.25 ± 614.01 µm(2); P < .0001). CONCLUSIONS: In vivo EP-mediated delivery of HIF-1α plasmid DNA improves neovascularization in a mouse model of limb ischemia and is a potentially suitable nonviral, noninvasive intervention to facilitate therapeutic angiogenesis in critical limb ischemia.


Assuntos
Eletroporação , Técnicas de Transferência de Genes , Terapia Genética/métodos , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , Isquemia/terapia , Músculo Esquelético/irrigação sanguínea , Neovascularização Fisiológica , Animais , Velocidade do Fluxo Sanguíneo , Circulação Colateral , Modelos Animais de Doenças , Membro Posterior , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Injeções Intramusculares , Isquemia/genética , Isquemia/metabolismo , Isquemia/patologia , Isquemia/fisiopatologia , Fluxometria por Laser-Doppler , Camundongos , Camundongos Endogâmicos BALB C , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Necrose , Recuperação de Função Fisiológica , Fluxo Sanguíneo Regional , Fatores de Tempo
10.
Immunology ; 140(2): 202-10, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23710940

RESUMO

A sudden upsurge of fever cases with joint pain was observed in the outpatient department, Community Health Centre, Rangat during July-August 2010 in Rangat Middle Andaman, India. The aetiological agent responsible for the outbreak was identified as chikungunya virus (CHIKV), by using RT-PCR and IgM ELISA. The study investigated the association of polymorphisms in the human leucocyte antigen class II genes with susceptibility or protection against CHIKV. One hundred and one patients with clinical features suggestive of CHIKV infection and 104 healthy subjects were included in the study. DNA was extracted and typed for HLA-DRB1 and DQB1 alleles. Based on the amino acid sequences of HLA-DQB1 retrieved from the IMGT/HLA database, critical amino acid differences in the specific peptide-binding pockets of HLA-DQB1 molecules were investigated. The frequencies of HLA-DRB1 alleles were not significantly different, whereas lower frequency of HLA-DQB1*03:03 was observed in CHIKV patients compared with the control population [P = 0·001, corrected P = 0·024; odds ratio (OR)  = 0, 95% confidence interval (95% CI) 0·0-0·331; Peto's OR = 0·1317, 95% CI 0·0428-0·405). Significantly lower frequency of glutamic acid at position 86 of peptide-binding pocket 1 coding HLA-DQB1 genotypes was observed in CHIKV patients compared with healthy controls (P = 0·004, OR = 0·307, 95% CI 0·125-0·707). Computational binding predictions of CD4 epitopes of CHIKV by NetMHCII revealed that HLA-DQ molecules are known to bind more CHIKV peptides than HLA-DRB1 molecules. The results suggest that HLA-DQB1 alleles and critical amino acid differences in the peptide-binding pockets of HLA-DQB1 alleles might have role in influencing infection and pathogenesis of CHIKV.


Assuntos
Infecções por Alphavirus/genética , Predisposição Genética para Doença/genética , Antígenos de Histocompatibilidade Classe II/genética , Polimorfismo Genético/genética , Adulto , Alelos , Infecções por Alphavirus/epidemiologia , Sítios de Ligação , Febre de Chikungunya , Surtos de Doenças , Ensaio de Imunoadsorção Enzimática , Feminino , Genótipo , Humanos , Índia , Masculino , Reação em Cadeia da Polimerase Via Transcriptase Reversa
11.
Viral Immunol ; 26(1): 75-83, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23409931

RESUMO

Chikungunya virus (CHIKV) is an important emerging mosquito-borne alphavirus, indigenous to tropical Africa and Asia. It can cause epidemic fever and acute illness characterized by fever and arthralgias. The epidemic cycle of this infection is similar to dengue and urban yellow fever viral infections. The generation of an efficient vaccine against CHIKV is necessary to prevent and/or control the disease manifestations of the infection. In this report, we studied immune response against a CHIKV-envelope DNA vaccine (pEnv) and the role of the CHIKV nonstructural gene 2 (nsP2) as an adjuvant for the induction of protective immune responses in a relevant mouse challenge model. When injected with the CHIKV pEnv alone, 70% of the immunized mice survived CHIKV challenge, whereas when co-injected with pEnv+pnsP2, 90% of the mice survived viral challenge. Mice also exhibited a delayed onset signs of illness, and a marked decrease in morbidity, suggesting a nsP2 mediated adjuvant effect. Co-injection of the pnsP2 adjuvant with pEnv also qualitatively and quantitatively increased antigen specific neutralizing antibody responses compared to vaccination with pEnv alone. In sum, these novel data imply that the addition of nsP2 to the pEnv vaccine enhances anti-CHIKV-Env immune responses and maybe useful to include in future CHIKV clinical vaccination strategies.


Assuntos
Adjuvantes Imunológicos/metabolismo , Infecções por Alphavirus/prevenção & controle , Vírus Chikungunya/imunologia , Vacinas de DNA/imunologia , Proteínas do Envelope Viral/imunologia , Proteínas não Estruturais Virais/metabolismo , Adjuvantes Imunológicos/genética , Infecções por Alphavirus/imunologia , Infecções por Alphavirus/patologia , Animais , Febre de Chikungunya , Vírus Chikungunya/genética , Modelos Animais de Doenças , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Índice de Gravidade de Doença , Análise de Sobrevida , Vacinas de DNA/administração & dosagem , Vacinas de DNA/genética , Proteínas do Envelope Viral/genética , Proteínas não Estruturais Virais/genética
12.
Hum Vaccin Immunother ; 8(11): 1668-81, 2012 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-23151448

RESUMO

There is no licensed vaccine or cure for human cytomegalovirus (CMV), a ubiquitous ß-herpesvirus infecting 60-95% of adults worldwide. Infection can cause congenital abnormalities, result in severe disease in immunocompromised patients, and is a major impediment during successful organ transplantation. In addition, it has been associated with numerous inflammatory diseases and cancers, as well as being implicated in the development of essential hypertension, a major risk factor for heart disease. To date, limited data regarding the identification of immunogenic viral targets has frustrated CMV vaccine development. Based upon promising clinical data suggesting an important role for T cells in protecting against disease in the transplantation setting, we designed a novel panel of highly-optimized synthetic vaccines encoding major CMV proteins and evaluated their immune potential in murine studies. Vaccination induced robust CD8+ and CD4+ T cells of great epitopic breadth as extensively analyzed using a novel modified T cell assay described herein. Together with improved levels of CMV-specific T cells as driven by a vaccine, further immune evaluation of each target is warranted. The present model provides an important tool for guiding future immunization strategies against CMV.


Assuntos
Vacinas contra Citomegalovirus/imunologia , Citomegalovirus/imunologia , Linfócitos T/imunologia , Vacinas Sintéticas/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Eletroporação , Feminino , Citometria de Fluxo , Terapia Genética , Camundongos , Camundongos Endogâmicos C57BL
13.
Viral Immunol ; 24(4): 265-71, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21830898

RESUMO

Chikungunya virus (CHIKV) has caused large outbreaks worldwide in recent years. Acute-phase CHIKV infection has been reported to cause mild to severe febrile illness, and in some patients, this may be followed by long-lasting polyarthritis. The mainstay of treatment includes nonsteroidal anti-inflammatory drugs and other disease-modifying agents, the use of which is based on the assumption of an immunological interference mechanism in the pathogenesis. The present study has been designed to generate preliminary evidence to test this hypothesis. The levels of 30 cytokines were estimated in serum samples of acute CHIKV-infected patients, fully-recovered patients, patients with chronic CHIKV arthritis, and controls, using a quantitative multiplex bead ELISA. The levels of the proinflammatory cytokines IL-1 and IL-6 were elevated in acute patients, but IFN-γ/ß and TNF-α levels remained stable. IL-10, which might have an anti-inflammatory effect, was also elevated, indicating a predominantly anti-inflammatory response in the acute phase of infection. Elevation of MCP-1, IL-6, IL-8, MIP-1α, and MIP-1ß was most prominent in the chronic phase. These cytokines and chemokines have been shown to play important roles in other arthritides, including epidemic polyarthritis (EPA) caused by Ross River virus (RRV) and rheumatoid arthritis (RA).The immunopathogenesis of chronic CHIKV arthritis might have similarities to these arthritides. The novel intervention strategies being developed for EPA and RA, such as IL-6 and IL-8 signaling blockade, may also be considered for chronic CHIKV arthritis.


Assuntos
Infecções por Alphavirus/imunologia , Infecções por Alphavirus/patologia , Artrite Infecciosa/imunologia , Artrite Infecciosa/patologia , Vírus Chikungunya/patogenicidade , Citocinas/imunologia , Adulto , Infecções por Alphavirus/complicações , Doença Crônica , Citocinas/sangue , Ensaio de Imunoadsorção Enzimática , Humanos , Pessoa de Meia-Idade
14.
J Immunol ; 187(6): 2932-43, 2011 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-21856939

RESUMO

Recent evidence demonstrates that HIV-1 infection leads to the attenuation of cellular immune responses, which has been correlated with the increased expression of programmed death (PD)-1 on virus-specific CD8(+) T cells. PD-1 is induced upon T cell activation, and its prolonged expression facilitates CD8(+) T cell inhibitory signals when bound to its B7 family ligands, PD-ligand (L)1/2, which are expressed on APCs. Importantly, early reports demonstrated that blockade of the PD-1/PD-L interaction by Abs may help to counter the development of immune exhaustion driven by HIV viral persistence. To better understand the regulation of the PD-1 pathway during HIV infection, we examined the ability of the virus to induce PD-L expression on macrophages and dendritic cells. We found a direct relationship between the infection of APCs and the expression of PD-L1 in which virus-mediated upregulation induced a state of nonresponsiveness in uninfected HIV-specific T cells. Furthermore, this exhaustion phenotype was revitalized by the blockade of PD-L1, after which T cells regained their capacity for proliferation and the secretion of proinflammatory cytokines IFN-γ, IL-2, and IL-12 upon restimulation. In addition, we identify a critical role for the PI3K/serine-threonine kinase signaling pathway in PD-L1 upregulation of APCs by HIV, because inhibition of these intracellular signal transducer enzymes significantly reduced PD-L1 induction by infection. These data identify a novel mechanism by which HIV exploits the immunosuppressive PD-1 pathway and suggest a new role for virus-infected cells in the local corruption of immune responses required for viral suppression.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Linfócitos T CD8-Positivos/imunologia , Infecções por HIV/imunologia , Ativação Linfocitária/imunologia , Transdução de Sinais/imunologia , Células Apresentadoras de Antígenos/metabolismo , Antígenos CD/biossíntese , Antígenos CD/imunologia , Proteínas Reguladoras de Apoptose/biossíntese , Proteínas Reguladoras de Apoptose/imunologia , Western Blotting , Linfócitos T CD8-Positivos/metabolismo , Separação Celular , Ativação Enzimática/imunologia , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Infecções por HIV/metabolismo , HIV-1/imunologia , Humanos , Ligantes , Fosfatidilinositol 3-Quinases/imunologia , Fosfatidilinositol 3-Quinases/metabolismo , Receptor de Morte Celular Programada 1 , Proteínas Serina-Treonina Quinases/imunologia , Proteínas Serina-Treonina Quinases/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Regulação para Cima
15.
PLoS Negl Trop Dis ; 5(1): e928, 2011 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-21264351

RESUMO

Chikungunya virus (CHIKV) is an emerging mosquito-borne alphavirus indigenous to tropical Africa and Asia. Acute illness is characterized by fever, arthralgias, conjunctivitis, rash, and sometimes arthritis. Relatively little is known about the antigenic targets for immunity, and no licensed vaccines or therapeutics are currently available for the pathogen. While the Aedes aegypti mosquito is its primary vector, recent evidence suggests that other carriers can transmit CHIKV thus raising concerns about its spread outside of natural endemic areas to new countries including the U.S. and Europe. Considering the potential for pandemic spread, understanding the development of immunity is paramount to the development of effective counter measures against CHIKV. In this study, we isolated a new CHIKV virus from an acutely infected human patient and developed a defined viral challenge stock in mice that allowed us to study viral pathogenesis and develop a viral neutralization assay. We then constructed a synthetic DNA vaccine delivered by in vivo electroporation (EP) that expresses a component of the CHIKV envelope glycoprotein and used this model to evaluate its efficacy. Vaccination induced robust antigen-specific cellular and humoral immune responses, which individually were capable of providing protection against CHIKV challenge in mice. Furthermore, vaccine studies in rhesus macaques demonstrated induction of nAb responses, which mimicked those induced in convalescent human patient sera. These data suggest a protective role for nAb against CHIKV disease and support further study of envelope-based CHIKV DNA vaccines.


Assuntos
Infecções por Alphavirus/prevenção & controle , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Vírus Chikungunya/imunologia , Vacinas de DNA/imunologia , Infecções por Alphavirus/virologia , Animais , Vírus Chikungunya/genética , Vírus Chikungunya/isolamento & purificação , Modelos Animais de Doenças , Eletroporação , Feminino , Humanos , Macaca mulatta , Camundongos , Camundongos Endogâmicos BALB C , Testes de Neutralização/métodos , Vacinação/métodos , Vacinas de DNA/administração & dosagem , Vacinas de DNA/genética , Virologia/métodos
16.
Vaccine ; 29(39): 6755-62, 2011 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-21238574

RESUMO

Protection against infection is the hallmark of immunity and the basis of effective vaccination. For a variety of reasons there is a great demand to develop new, safer and more effective vaccine platforms. In this regard, while 'first-generation' DNA vaccines were poorly immunogenic, new genetic 'optimization' strategies and the application of in vivo electroporation (EP) have dramatically boosted their potency. We developed a highly optimized plasmid DNA vaccine that expresses the lymphocytic choriomeningitis virus (LCMV) nucleocapsid protein (NP) and evaluated it using the LCMV challenge model, a gold standard for studying infection and immunity. When administered intramuscularly with EP, robust NP-specific cellular and humoral immune responses were elicited, the magnitudes of which approached those following acute LCMV infection. Furthermore, these responses were capable of providing 100% protection against a high-dose, normally lethal virus challenge. This is the first non-infectious vaccine conferring complete protective immunity up to 8 weeks after vaccination and demonstrates the potential of 'next-generation' DNA vaccines.


Assuntos
Imunidade Celular , Imunidade Humoral , Coriomeningite Linfocítica/prevenção & controle , Proteínas do Nucleocapsídeo/imunologia , Vacinas de DNA/imunologia , Vacinas Virais/imunologia , Animais , Formação de Anticorpos , ELISPOT , Feminino , Vetores Genéticos , Células HEK293 , Humanos , Dose Letal Mediana , Coriomeningite Linfocítica/imunologia , Coriomeningite Linfocítica/virologia , Vírus da Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/patogenicidade , Camundongos , Camundongos Endogâmicos C57BL , Plasmídeos/genética , Plasmídeos/metabolismo , Transfecção , Vacinação/métodos , Vacinas de DNA/administração & dosagem , Vacinas Virais/administração & dosagem
17.
Curr Opin Virol ; 1(4): 233-40, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22440782

RESUMO

The safety, stability, and ability for repeat homologous vaccination makes the DNA vaccine platform an excellent candidate for an effective HIV-1 vaccine. However, the immunogenicity of early DNA vaccines did not translate from small animal models into larger non-human primates and was markedly lower than viral vectors. In addition to improvements to the DNA vector itself, delivery with electroporation, the inclusion of molecular adjuvants, and heterologous prime-boost strategies have dramatically improved the immunogenicity of DNA vaccines for HIV and currently makes them a leading platform with many areas warranting further research and clinical development.


Assuntos
Vacinas contra a AIDS/imunologia , Infecções por HIV/imunologia , HIV-1/imunologia , Vacinação/métodos , Vacinas de DNA/imunologia , Vacinas contra a AIDS/genética , Animais , Infecções por HIV/genética , Infecções por HIV/prevenção & controle , HIV-1/genética , Humanos , Vacinas de DNA/genética
18.
Expert Rev Vaccines ; 9(7): 747-63, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20624048

RESUMO

DNA vaccination has been of great interest since its discovery in the 1990s due to its ability to elicit both humoral and cellular immune responses. DNA vaccines consist of a DNA plasmid containing a transgene that encodes the sequence of a target protein from a pathogen under the control of a eukaryotic promoter. This revolutionary technology has proven to be effective in animal models and four DNA vaccine products have recently been approved for veterinary use. Although few DNA vaccines against bacterial infections have been tested, the results are encouraging. Because of their versatility, safety and simplicity a wider range of organisms can be targeted by these vaccines, which shows their potential advantages to public health. This article describes the mechanism of action of DNA vaccines and their potential use for targeting bacterial infections. In addition, it provides an updated summary of the methods used to enhance immunogenicity from codon optimization and adjuvants to delivery techniques including electroporation and use of nanoparticles.


Assuntos
Infecções Bacterianas/prevenção & controle , Vacinas Bacterianas/imunologia , Vacinas de DNA/imunologia , Adjuvantes Imunológicos/administração & dosagem , Animais , Antígenos de Bactérias/genética , Códon , Modelos Animais de Doenças , Sistemas de Liberação de Medicamentos , Expressão Gênica , Vetores Genéticos , Humanos , Plasmídeos , Regiões Promotoras Genéticas
19.
PLoS Negl Trop Dis ; 4(4): e623, 2010 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-20436958

RESUMO

Chikungunya virus is a mosquito-borne emerging pathogen that has a major health impact in humans and causes fever disease, headache, rash, nausea, vomiting, myalgia, and arthralgia. Indigenous to tropical Africa, recent large outbreaks have been reported in parts of South East Asia and several of its neighboring islands in 2005-07 and in Europe in 2007. Furthermore, positive cases have been confirmed in the United States in travelers returning from known outbreak areas. Currently, there is no vaccine or antiviral treatment. With the threat of an emerging global pandemic, the peculiar problems associated with the more immediate and seasonal epidemics warrant the development of an effective vaccine. In this review, we summarize the evidence supporting these concepts.


Assuntos
Infecções por Alphavirus/epidemiologia , Infecções por Alphavirus/virologia , Vírus Chikungunya/isolamento & purificação , Doenças Transmissíveis Emergentes/epidemiologia , Doenças Transmissíveis Emergentes/virologia , África/epidemiologia , Infecções por Alphavirus/prevenção & controle , Sudeste Asiático/epidemiologia , Vírus Chikungunya/imunologia , Doenças Transmissíveis Emergentes/prevenção & controle , Europa (Continente)/epidemiologia , Humanos , Viagem , Estados Unidos/epidemiologia , Vacinas Virais/imunologia
20.
Cytometry A ; 77(3): 275-84, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20104580

RESUMO

The capacity for robust proliferation upon re-infection is a hallmark of adaptive immunity and the basis of vaccination. A widely used animal model for the study of human disease is the rhesus macaque (RM), where capacity for proliferation can be assessed ex vivo using carboxyfluorescein succinimidyl ester (CFSE)-based dilution assays. However, we show over the course of the standard ex vivo proliferation assay that CFSE-labeling at commonly used dye concentrations induces significant cell death, but that this phenomenon is dose-dependent. Here, we describe an alternative semiquantitative method for estimating T cell proliferative responses that avoids the putative biases associated with chemical modification. RM peripheral blood mononuclear cells were stimulated ex vivo with cognate peptides for 5 days, immunostained for intracellular Ki-67, and then analyzed by flow cytometry. We describe a gating strategy using Ki-67 and side light scatter, also a marker of blastogenesis, which correlates strongly with data from CFSE dilution. We show that this method is a valid tool for measuring RM antigen-specific cellular proliferation ex vivo and can be used as an alternative to CFSE dilution assays.


Assuntos
Citometria de Fluxo/métodos , Antígeno Ki-67/biossíntese , Linfócitos T/citologia , Animais , Antígenos/química , Linfócitos T CD8-Positivos/imunologia , Proliferação de Células , Corantes/farmacologia , Relação Dose-Resposta a Droga , Fluoresceínas/química , Deleção de Genes , Leucócitos Mononucleares/citologia , Macaca mulatta , Succinimidas/química , Linfócitos T/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...