Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
PLoS Pathog ; 14(8): e1007273, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30153307

RESUMEN

Human whipworm (Trichuris trichiura) infects approximately 1 in 15 people worldwide, representing the leading infectious cause of colitis and subsequent, inflammatory bowel disease (IBD). Current control measures focused on mass deworming have had limited success due to low drug efficacies. Vaccination would be an ideal, cost-effective strategy to induce protective immunity, leading to control of infection and transmission. Here we report the identification of whey acidic protein, a whipworm secretory protein, as a strong immunogen for inducing protective efficacy in a surrogate mouse T. muris infection model. The recombinant WAP protein (rTm-WAP49), as well as a single, highly conserved repeat within WAP (fragment 8) expressed as an Na-GST-1 fusion protein (rTm-WAP-F8+Na-GST-1), generate a strong T helper type 2 (Th2) immune response when delivered as subcutaneous vaccines formulated with Montanide ISA 720. Oral challenge with T. muris infective eggs following vaccination led to a significant reduction in worm burden of 48% by rTm-WAP49 and 33% by rTm-WAP-F8+Na-GST-1. The cellular immune correlates of protection included significant antigen-specific production of Th2 cytokines IL-4, IL-9, and IL-13 by cells isolated from the vaccine-draining inguinal lymph nodes, parasite-draining mesenteric lymph nodes, and spleen in mice vaccinated with either rTm-WAP49 or rTm-WAP-F8+Na-GST-1. The humoral immune correlates included a high antigen-specific ratio of IgG1 to IgG2a, without eliciting an IgE-mediated allergic response. Immunofluorescent staining of adult T. muris with WAP antisera identified the worm's pathogenic stichosome organ as the site of secretion of native Tm-WAP protein into the colonic mucosa. Given the high sequence conservation for the WAP proteins from T. muris and T. trichiura, the results presented here support the WAP protein to be further evaluated as a potential human whipworm vaccine candidate.


Asunto(s)
Inmunidad , Proteínas de la Leche/inmunología , Tricuriasis/prevención & control , Trichuris/inmunología , Animales , Anticuerpos Antihelmínticos/metabolismo , Antígenos Helmínticos/genética , Antígenos Helmínticos/inmunología , Antígenos Helmínticos/farmacología , Inmunidad/efectos de los fármacos , Inmunidad/genética , Masculino , Ratones , Ratones Endogámicos AKR , Ratones Noqueados , Ratones SCID , Proteínas de la Leche/genética , Proteínas de la Leche/farmacología , Tricuriasis/inmunología , Trichuris/genética , Vacunación/métodos
2.
Infect Immun ; 86(12)2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30249744

RESUMEN

Ascaris lumbricoides (roundworm) is the most common helminth infection globally and a cause of lifelong morbidity that may include allergic airway disease, an asthma phenotype. We hypothesize that Ascaris larval migration through the lungs leads to persistent airway hyperresponsiveness (AHR) and type 2 inflammatory lung pathology despite resolution of infection that resembles allergic airway disease. Mice were infected with Ascaris by oral gavage. Lung AHR was measured by plethysmography and histopathology with hematoxylin and eosin (H&E) and periodic acid-Schiff (PAS) stains, and cytokine concentrations were measured by using Luminex Magpix. Ascaris-infected mice were compared to controls or mice with allergic airway disease induced by ovalbumin (OVA) sensitization and challenge (OVA/OVA). Ascaris-infected mice developed profound AHR starting at day 8 postinfection (p.i.), peaking at day 12 p.i. and persisting through day 21 p.i., despite resolution of infection, which was significantly increased compared to controls and OVA/OVA mice. Ascaris-infected mice had a robust type 2 cytokine response in both the bronchoalveolar lavage (BAL) fluid and lung tissue, similar to that of the OVA/OVA mice, including interleukin-4 (IL-4) (P < 0.01 and P < 0.01, respectively), IL-5 (P < 0.001 and P < 0.001), and IL-13 (P < 0.001 and P < 0.01), compared to controls. By histopathology, Ascaris-infected mice demonstrated early airway remodeling similar to, but more profound than, that in OVA/OVA mice. We found that Ascaris larval migration causes significant pulmonary damage, including AHR and type 2 inflammatory lung pathology that resembles an extreme form of allergic airway disease. Our findings indicate that ascariasis may be an important cause of allergic airway disease in regions of endemicity.


Asunto(s)
Ascariasis/fisiopatología , Hipersensibilidad/parasitología , Pulmón/patología , Hipersensibilidad Respiratoria/parasitología , Animales , Ascariasis/inmunología , Ascaris/patogenicidad , Líquido del Lavado Bronquioalveolar/inmunología , Modelos Animales de Enfermedad , Femenino , Interleucina-13/inmunología , Interleucina-4/inmunología , Interleucina-5/inmunología , Larva/patogenicidad , Pulmón/parasitología , Ratones , Ratones Endogámicos BALB C , Ovalbúmina , Células Th2/inmunología
3.
PLoS Negl Trop Dis ; 12(12): e0006975, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30557313

RESUMEN

BACKGROUND: Hyperendemic circulation of all four types of dengue virus (DENV-1-4) has expanded globally, fueling concern for increased incidence of severe dengue. While the majority of DENV infections are subclinical, epidemiologic studies suggest that type-cross-reactive immunity can influence disease outcome in subsequent infections. The mechanisms controlling these differential clinical outcomes remain poorly defined. METHODOLOGY/PRINCIPAL FINDINGS: Blood samples were collected from a cohort of school-aged Thai children who subsequently experienced a subclinical DENV infection or developed dengue illness. PBMC collected prior to infection were stimulated in vitro with DENV and the secretion of 30 cytokines was measured using a multiplexed, bead-based array. Significant differences were found in cytokine production based on both the type of DENV used for stimulation and the occurrence of clinical illness. Secretion of IL-15 and MCP-1 was significantly higher by PBMC of subjects who later developed symptomatic DENV infection. In addition, IL-6 was produced by PBMC from all subjects who subsequently developed symptomatic infection, versus 59% of subjects who had subclinical infection. Secretion of IL-12, IL-2R, MIP-1α, RANTES, GM-CSF, and TNFα was significantly lower by PBMC from subjects with symptomatic infection. CONCLUSIONS/SIGNIFICANCE: These data demonstrate significant differences in pre-existing immune responses to DENV associated with the clinical outcome of subsequent infection. The finding of higher levels of some cytokines in subjects with symptomatic infection and higher levels of other cytokines in subjects with subclinical infection supports the existence of both protective and pathologic immune profiles. Clinical-immunological correlations identified in the context of natural DENV infection may be useful for evaluating immune responses to dengue vaccines.


Asunto(s)
Citocinas/análisis , Virus del Dengue/inmunología , Dengue/inmunología , Infecciones Asintomáticas , Estudios de Cohortes , Reacciones Cruzadas , Dengue/virología , Humanos , Leucocitos Mononucleares , Tailandia/epidemiología
4.
J Immunol Methods ; 440: 74-82, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27840065

RESUMEN

Luminex® technology provides a powerful methodology for multiplex cytokine detection but remains constrained by high costs and a minimum of 25-50µL sample volume requirement per assay-well often hindering analysis of limited biological samples. Here we compare the results of Luminex-based cytokine multiplexing assay performed using conventional 96-well microtiter plates and a particular 96-well wall-less plate based on Droparray® technology ("DA-Bead"). The application of the DA-Bead plate allows 80% reduction of sample and reagent volume, thus an opportunity for significant cost savings in Luminex reagents with no change to the workflow. To compare the DA-Bead method to the conventional method, two different types of samples were tested with two different commercially available Luminex kits and the results for each method were compared. The first type was splenocyte culture supernatants from murine spleens which were harvested from mice immunized with Ascaris suum protein As24 and followed by cell stimulation ex vivo at various time points with this same antigen. Cytokine levels in these supernatants were evaluated using a Bio-Plex® TH1/TH2 8-plex kit. The second sample type was plasma from mice from an experimental autoimmune encephalomyelitis (EAE) study, and these samples were evaluated using a Milliplex® TH17 25-plex kit. The data showed that the DA-Bead method for analysis was comparable to, if not superior to, the conventional method in terms of consistency/precision, accuracy, sensitivity and dynamic range and these results are not specific to sample type, reagents, or commercial vendor.


Asunto(s)
Antígenos Helmínticos/inmunología , Ascaris suum/inmunología , Citocinas/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Ensayo de Inmunoadsorción Enzimática/instrumentación , Proteínas del Helminto/inmunología , Bazo/inmunología , Animales , Antígenos Helmínticos/administración & dosificación , Biomarcadores/metabolismo , Células Cultivadas , Medios de Cultivo Condicionados/metabolismo , Citocinas/sangre , Encefalomielitis Autoinmune Experimental/sangre , Ensayo de Inmunoadsorción Enzimática/métodos , Diseño de Equipo , Femenino , Proteínas del Helminto/administración & dosificación , Inmunización , Ratones Endogámicos BALB C , Reproducibilidad de los Resultados , Bazo/metabolismo , Factores de Tiempo , Flujo de Trabajo
5.
PLoS Negl Trop Dis ; 11(7): e0005769, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28708895

RESUMEN

BACKGROUND: Ascariasis remains the most common helminth infection in humans. As an alternative or complementary approach to global deworming, a pan-anthelminthic vaccine is under development targeting Ascaris, hookworm, and Trichuris infections. As16 and As14 have previously been described as two genetically related proteins from Ascaris suum that induced protective immunity in mice when formulated with cholera toxin B subunit (CTB) as an adjuvant, but the exact protective mechanism was not well understood. METHODOLOGY/PRINCIPAL FINDINGS: As16 and As14 were highly expressed as soluble recombinant proteins (rAs16 and rAs14) in Pichia pastoris. The yeast-expressed rAs16 was highly recognized by immune sera from mice infected with A. suum eggs and elicited 99.6% protection against A. suum re-infection. Mice immunized with rAs16 formulated with ISA720 displayed significant larva reduction (36.7%) and stunted larval development against A. suum eggs challenge. The protective immunity was associated with a predominant Th2-type response characterized by high titers of serological IgG1 (IgG1/IgG2a > 2000) and high levels of IL-4 and IL-5 produced by restimulated splenocytes. A similar level of protection was observed in mice immunized with rAs16 formulated with alum (Alhydrogel), known to induce mainly a Th2-type immune response, whereas mice immunized with rAs16 formulated with MPLA or AddaVax, both known to induce a Th1-type biased response, were not significantly protected against A. suum infection. The rAs14 protein was not recognized by A. suum infected mouse sera and mice immunized with rAs14 formulated with ISA720 did not show significant protection against challenge infection, possibly due to the protein's inaccessibility to the host immune system or a Th1-type response was induced which would counter a protective Th2-type response. CONCLUSIONS/SIGNIFICANCE: Yeast-expressed rAs16 formulated with ISA720 or alum induced significant protection in mice against A. suum egg challenge that associates with a Th2-skewed immune response, suggesting that rAS16 could be a feasible vaccine candidate against ascariasis.


Asunto(s)
Anticuerpos Antihelmínticos/sangre , Antígenos Helmínticos/uso terapéutico , Ascariasis/prevención & control , Células Th2/inmunología , Vacunas/uso terapéutico , Adyuvantes Inmunológicos , Animales , Antígenos Helmínticos/inmunología , Ascaris suum , Toxina del Cólera/inmunología , Femenino , Inmunoglobulina G/sangre , Interleucina-4/inmunología , Interleucina-5/inmunología , Larva , Ratones , Ratones Endogámicos BALB C , Proteínas Recombinantes/uso terapéutico , Saccharomyces cerevisiae , Análisis de Secuencia , Vacunación
6.
Vaccine ; 34(26): 2992-2995, 2016 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-26973063

RESUMEN

A number of leishmaniasis vaccine candidates are at various stages of pre-clinical and clinical development. Leishmaniasis is a vector-borne neglected tropical disease (NTD) caused by a protozoan parasite of the genus Leishmania and transmitted to humans by the bite of a sand fly. Visceral leishmaniasis (VL, kala-azar) is a high mortality NTD found mostly in South Asia and East Africa, while cutaneous leishmaniasis (CL) is a disfiguring NTD highly endemic in the Middle East, Central Asia, North Africa, and the Americas. Estimates attribute 50,000 annual deaths and 3.3 million disability-adjusted life years to leishmaniasis. There are only a few approved drug treatments, no prophylactic drug and no vaccine. Ideally, an effective vaccine against leishmaniasis will elicit long-lasting immunity and protect broadly against VL and CL. Vaccines such as Leish-F1, F2 and F3, developed at IDRI and designed based on selected Leishmania antigen epitopes, have been in clinical trials. Other groups, including the Sabin Vaccine Institute in collaboration with the National Institutes of Health are investigating recombinant Leishmania antigens in combination with selected sand fly salivary gland antigens in order to augment host immunity. To date, both VL and CL vaccines have been shown to be cost-effective in economic modeling studies.


Asunto(s)
Vacunas contra la Leishmaniasis/uso terapéutico , Leishmaniasis/prevención & control , Animales , Investigación Biomédica/tendencias , Ensayos Clínicos como Asunto , Humanos , Leishmaniasis Cutánea/prevención & control , Leishmaniasis Visceral/prevención & control , Psychodidae/parasitología , Vacunas Sintéticas/uso terapéutico
7.
Vaccine ; 34(26): 3001-3005, 2016 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-27040400

RESUMEN

A human hookworm vaccine is under development and in clinical trials in Africa and the Americas. The vaccine contains the Na-APR-1 and Na-GST-1 antigens. It elicits neutralizing antibodies that interfere with establishment of the adult hookworm in the gut and the ability of the parasite to feed on blood. The vaccine target product profile is focused on the immunization of children to prevent hookworm infection and anemia caused by Necator americanus. It is intended for use in low- and middle-income countries where hookworm is highly endemic and responsible for at least three million disability-adjusted life years. So far, the human hookworm vaccine is being developed in the non-profit sector through the Sabin Vaccine Institute Product Development Partnership (PDP), in collaboration with the HOOKVAC consortium of European and African partners. We envision the vaccine to be incorporated into health systems as part of an elimination strategy for hookworm infection and other neglected tropical diseases, and as a means to reduce global poverty and address the Sustainable Development Goals.


Asunto(s)
Anemia/prevención & control , Infecciones por Uncinaria/prevención & control , Vacunas/uso terapéutico , Anemia/parasitología , Animales , Investigación Biomédica/tendencias , Ensayos Clínicos como Asunto , Perros , Infecciones por Uncinaria/complicaciones , Humanos , Necator americanus
8.
Vaccine ; 34(26): 2988-2991, 2016 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-27036511

RESUMEN

Several candidate human schistosomiasis vaccines are in different stages of preclinical and clinical development. The major targets are Schistosoma haematobium (urogenitial schistosomiasis) and Schistosoma mansoni (intestinal schistosomiasis) that account for 99% of the world's 252 million cases, with 90% of these cases in Africa. Two recombinant S. mansoni vaccines - Sm-TSP-2 and Sm-14 are in Phase 1 trials, while Smp80 (calpain) is undergoing testing in non-human primates. Sh28GST, also known as Bilhvax is in advanced clinical development for S. haematobium infection. The possibility remains that some of these vaccines may cross-react to target both schistosome species. These vaccines were selected on the basis of their protective immunity in preclinical challenge models, through human immune-epidemiological studies or both. They are being advanced through a combination of academic research institutions, non-profit vaccine product development partnerships, biotechnology companies, and developing country vaccine manufacturers. In addition, new schistosome candidate vaccines are being identified through bioinformatics, OMICs approaches, and moderate throughput screening, although the full potential of reverse vaccinology for schistosomiasis has not yet been realized. The target product profiles of these vaccines vary but many focus on vaccinating children, in some cases following mass treatment with praziquantel, also known as vaccine-linked chemotherapy. Several regulatory pathways have been proposed, some of which rely on World Health Organization prequalification.


Asunto(s)
Esquistosomiasis/prevención & control , Vacunas/uso terapéutico , Animales , Antígenos Bacterianos/inmunología , Antígenos Helmínticos/inmunología , Investigación Biomédica/tendencias , Ensayos Clínicos como Asunto , Proteínas de Transporte de Ácidos Grasos/inmunología , Glutatión Transferasa/inmunología , Proteínas del Helminto/inmunología , Humanos , Primates , Schistosoma haematobium , Schistosoma mansoni , Tetraspaninas/inmunología , Vacunas Sintéticas/uso terapéutico
9.
Hum Vaccin Immunother ; 12(4): 976-87, 2016 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-26890466

RESUMEN

Chagas disease, caused by Trypanosoma cruzi, results in an acute febrile illness that progresses to chronic chagasic cardiomyopathy in 30% of patients. Current treatments have significant side effects and poor efficacy during the chronic phase; therefore, there is an urgent need for new treatment modalities. A robust TH1-mediated immune response correlates with favorable clinical outcomes. A therapeutic vaccine administered to infected individuals could bolster the immune response, thereby slowing or stopping the progression of chagasic cardiomyopathy. Prior work in mice has identified an efficacious T. cruzi DNA vaccine encoding Tc24. To elicit a similar protective cell-mediated immune response to a Tc24 recombinant protein, we utilized a poly(lactic-co-glycolic acid) nanoparticle delivery system in conjunction with CpG motif-containing oligodeoxynucleotides as an immunomodulatory adjuvant. In a BALB/c mouse model, the vaccine produced a TH1-biased immune response, as demonstrated by a significant increase in antigen-specific IFNγ-producing splenocytes, IgG2a titers, and proliferative capacity of CD8(+) T cells. When tested for therapeutic efficacy, significantly reduced systemic parasitemia was seen during peak parasitemia. Additionally, there was a significant reduction in cardiac parasite burden and inflammatory cell infiltrate. This is the first study demonstrating immunogenicity and efficacy of a therapeutic Chagas vaccine using a nanoparticle delivery system.


Asunto(s)
Enfermedad de Chagas/terapia , Vacunas Antiprotozoos/uso terapéutico , Trypanosoma cruzi/inmunología , Vacunas de ADN/uso terapéutico , Adyuvantes Inmunológicos/administración & dosificación , Animales , Anticuerpos Antiprotozoarios/inmunología , Linfocitos T CD8-positivos/inmunología , Cardiomiopatía Chagásica/terapia , Enfermedad de Chagas/inmunología , Modelos Animales de Enfermedad , Femenino , Corazón/parasitología , Inmunidad Celular , Inmunoglobulina G/biosíntesis , Inmunoglobulina G/inmunología , Inmunoterapia/métodos , Interferón gamma/inmunología , Ratones , Ratones Endogámicos BALB C , Nanopartículas , Oligodesoxirribonucleótidos/inmunología , Parasitemia/terapia , Vacunas Antiprotozoos/inmunología , Células TH1/inmunología , Vacunas de ADN/efectos adversos , Vacunas de ADN/inmunología
10.
Expert Rev Vaccines ; 13(3): 321-31, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24392641

RESUMEN

The Sabin Vaccine Institute Product Development Partnership is developing a Pan-anthelmintic vaccine that simultaneously targets the major soil-transmitted nematode infections, in other words, ascariasis, trichuriasis and hookworm infection. The approach builds off the current bivalent Human Hookworm Vaccine now in clinical development and would ultimately add both a larval Ascaris lumbricoides antigen and an adult-stage Trichuris trichiura antigen from the parasite stichosome. Each selected antigen would partially reproduce the protective immunity afforded by UV-attenuated Ascaris eggs and Trichuris stichosome extracts, respectively. Final antigen selection will apply a ranking system that includes the evaluation of expression yields and solubility, feasibility of process development and the absence of circulating antigen-specific IgE among populations living in helminth-endemic regions. Here we describe a five year roadmap for the antigen discovery, feasibility and antigen selection, which will ultimately lead to the scale-up expression, process development, manufacture, good laboratory practices toxicology and preclinical evaluation, ultimately leading to Phase 1 clinical testing.


Asunto(s)
Ascariasis/prevención & control , Infecciones por Uncinaria/prevención & control , Vacunas Antiprotozoos/inmunología , Tricuriasis/prevención & control , Ancylostomatoidea/inmunología , Animales , Antihelmínticos , Antígenos Helmínticos/inmunología , Ascariasis/inmunología , Ascaris lumbricoides/inmunología , Infecciones por Uncinaria/inmunología , Humanos , Suelo/parasitología , Tricuriasis/inmunología , Trichuris/inmunología
11.
Transl Res ; 162(3): 144-55, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23578479

RESUMEN

Neglected tropical diseases (NTDs) are a significant source of morbidity and socioeconomic burden among the world's poor. Virtually all of the 2.4 billion people who live on less than $2 per d, more than a third of the world's population, are at risk for these debilitating NTDs. Although chemotherapeutic measures exist for many of these pathogens, they are not sustainable countermeasures on their own because of rates of reinfection, risk of drug resistance, and inconsistent maintenance of drug treatment programs. Preventative and therapeutic NTD vaccines are needed as long-term solutions. Because there is no market in the for-profit sector of vaccine development for these pathogens, much of the effort to develop vaccines is driven by nonprofit entities, mostly through product development partnerships. This review describes the progress of vaccines under development for many of the NTDs, with a specific focus on those about to enter or that are currently in human clinical trials. Specifically, we report on the progress on dengue, hookworm, leishmaniasis, schistosomiasis, Chagas disease, and onchocerciasis vaccines. These products will be some of the first with specific objectives to aid the world's poorest populations.


Asunto(s)
Dengue/prevención & control , Enfermedades Parasitarias/prevención & control , Vacunas/uso terapéutico , Humanos , Vacunas/clasificación
12.
Vaccine ; 31 Suppl 2: B227-32, 2013 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-23598487

RESUMEN

Hookworm infection is one of the world's most common neglected tropical diseases and a leading cause of iron deficiency anemia in low- and middle-income countries. A Human Hookworm Vaccine is currently being developed by the Sabin Vaccine Institute and is in phase 1 clinical testing. The candidate vaccine is comprised of two recombinant antigens known as Na-GST-1 and Na-APR-1, each of which is an important parasite enzyme required for hookworms to successfully utilize host blood as a source of energy. The recombinant proteins are formulated on Alhydrogel(®) and are being tested in combination with a synthetic Toll-like receptor 4 agonist. The aim of the vaccine is to induce anti-enzyme antibodies that will reduce both host blood loss and the number of hookworms attached to the gut. Transfer of the manufacturing technology to the Oswaldo Cruz Foundation (FIOCRUZ)/Bio-Manguinhos (a Brazilian public sector developing country vaccine manufacturer) is planned, with a clinical development plan that could lead to registration of the vaccine in Brazil. The vaccine would also need to be introduced in the poorest regions of Africa and Asia, where hookworm infection is highly endemic. Ultimately, the vaccine could become an essential tool for achieving hookworm control and elimination, a key target in the 2012 London Declaration on Neglected Tropical Diseases.


Asunto(s)
Investigación Biomédica/tendencias , Infecciones por Uncinaria/prevención & control , Vacunas/uso terapéutico , Academias e Institutos , Ancylostomatoidea/enzimología , Ancylostomatoidea/inmunología , Animales , Antígenos Helmínticos/inmunología , Ensayos Clínicos Fase I como Asunto , Salud Global , Humanos , Sector Público , Asociación entre el Sector Público-Privado , Proteínas Recombinantes/inmunología , Transferencia de Tecnología , Receptor Toll-Like 4/agonistas
14.
Viral Immunol ; 23(5): 477-85, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20883162

RESUMEN

Cytotoxic T lymphocytes (CTL) are hypothesized to play a role in clearance during primary dengue virus (DENV) infections, and contribute to immunopathology during secondary heterologous infections in humans. We previously reported skewed T-cell responses to secondary DENV infection in BALB/c (H-2(d)) mice, reproducing characteristics of human DENV infection. To set the stage for using widely available transgenic and knockout mice, we extended these studies to identify DENV-specific T-cell responses in C57BL/6 (H-2(b)) mice. We identified dominant CD8+ T-cell responses to H-2D(b)-restricted epitopes on the DENV NS4a (aa 249-265) and NS5 (aa 521-537) proteins. High frequencies of IFN-γ- and TNF-α-producing T cells directed at both epitopes were detected following primary infection with all four DENV serotypes, and were augmented by secondary DENV infections. In vivo cytotoxicity assays demonstrated rapid clearance of target cells pulsed with the NS4a peptide; in contrast, NS5 peptide-pulsed target cells were poorly cleared in vivo. These data characterize two H-2(b)-restricted T-cell epitopes displaying divergent in vivo function. These results should facilitate further studies of the in vivo effects of DENV-specific T cells, including the use of genetically modified mouse strains.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Virus del Dengue/inmunología , Dengue/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Pruebas Inmunológicas de Citotoxicidad , Epítopos de Linfocito T/inmunología , Antígenos H-2/inmunología , Antígeno de Histocompatibilidad H-2D , Interferón gamma/metabolismo , Ratones , Ratones Endogámicos C57BL , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas no Estructurales Virales/inmunología
15.
Viral Immunol ; 22(3): 215-9, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19435418

RESUMEN

Secondary dengue virus (DENV) infection is a major factor contributing to the risk for severe disease, an effect that depends upon the sequence of infection with different DENV serotypes. We previously reported sequence-dependent effects of secondary DENV infection on CD8+ T-cell responses in mice. To further evaluate the effect of infection sequence, we analyzed DENV-specific CD4+ T-cell responses and their relationship to the CD8+ T-cell response. Serotype cross-reactivity of CD4+ T-cell responses also depended upon the sequence of serotypes in this model. Furthermore, adoptive transfer of memory CD4+ T cells altered the response of memory CD8+ T cells to secondary infection. These data demonstrate the interaction of different components of the T-cell response in determining the immunological outcome of secondary DENV infection.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Dengue/inmunología , Memoria Inmunológica , Animales , Antígenos Virales/inmunología , Reacciones Cruzadas , Virus del Dengue/inmunología , Humanos , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Serotipificación
16.
Nat Med ; 15(8): 879-85, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19525963

RESUMEN

African green monkeys (genus Chlorocebus) can be infected with species-specific simian immunodeficiency virus (SIVagm) but do not develop AIDS. These natural hosts of SIV, like sooty mangabeys, maintain high levels of SIV replication but have evolved to avoid immunodeficiency. Elucidating the mechanisms that allow natural hosts to coexist with SIV without overt disease may provide crucial information for understanding AIDS pathogenesis. Here we show that many CD4(+) T cells from African green monkeys downregulate CD4 in vivo as they enter the memory pool; that downregulation of CD4 by memory T cells is independent of SIV infection; that the CD4(-) memory T cells maintain functions that are normally attributed to CD4(+) T cells, including production of interleukin-2 (IL-2), production of IL-17, expression of forkhead box P3 and expression of CD40 ligand; that loss of CD4 expression protects these T cells from infection by SIVagm in vivo; and that these CD4(-) T cells can maintain major histocompatibility complex class II restriction. These data show that the absence of SIV-induced disease progression in natural host species may be partially explained by preservation of a subset of T cells that maintain CD4(+) T cell function while being resistant to SIV infection in vivo.


Asunto(s)
Antígenos CD4/metabolismo , Linfocitos T CD4-Positivos/fisiología , Chlorocebus aethiops/inmunología , Memoria Inmunológica/fisiología , Síndrome de Inmunodeficiencia Adquirida del Simio/prevención & control , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Regulación hacia Abajo/inmunología , Regulación hacia Abajo/fisiología , Inmunidad Innata/inmunología , Memoria Inmunológica/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Especificidad de la Especie
17.
J Infect Dis ; 197(4): 608-17, 2008 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-18275279

RESUMEN

Dengue virus is the causative agent of dengue fever and the more-severe dengue hemorrhagic fever (DHF). Human studies suggest that the increased risk of DHF during secondary infection is due to immunopathology partially mediated by cross-reactive memory T cells from the primary infection. To model T cell responses to sequential infections, we immunized mice with different sequences of dengue virus serotypes and measured the frequency of peptide-specific T cells after infection. The acute response after heterologous secondary infections was enhanced compared with the acute or memory response after primary infection. Also, the hierarchy of epitope-specific responses was influenced by the specific sequence of infection. Adoptive-transfer experiments showed that memory T cells responded preferentially to the secondary infection. These findings demonstrate that cross-reactive T cells from a primary infection alter the immune response during a heterologous secondary infection.


Asunto(s)
Linfocitos T CD8-positivos/virología , Virus del Dengue/inmunología , Dengue Grave/inmunología , Animales , Reacciones Cruzadas , Modelos Animales de Enfermedad , Memoria Inmunológica , Interferón gamma/inmunología , Ratones , Ratones Endogámicos BALB C , Serotipificación , Factor de Necrosis Tumoral alfa/inmunología
19.
J Immunol ; 170(11): 5786-92, 2003 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-12759463

RESUMEN

This study investigated the relationship between HIV-1 replication and virus (HIV-1; CMV)-specific CD4(+) T cell frequency and function in HIV-1-infected children. HIV-1 gag p55-specific CD4(+) T cell IFN-gamma responses were detected in the majority of children studied. p55-specific responses were detected less commonly and at lower frequencies in children with <50 copies/ml plasma HIV-1 RNA than in children with active HIV-1 replication. In children with <50 copies/ml plasma HIV-1, p55-specific responses were detected only in children with evidence of ongoing HIV-1 replication, indicating a direct relationship between HIV-1 replication and HIV-specific CD4(+) T cell frequencies. In contrast, p55-specific proliferative responses were detected more frequently in children with <50 copies/ml plasma HIV-1. CMV-specific CD4(+) responses were more commonly detected and at higher frequencies in CMV-coinfected children with suppressed HIV-1 replication. The lack of HIV-specific CD4(+) proliferative responses, along with the preservation of CMV-specific CD4(+) responses in children with controlled HIV-1 replication, suggests that viral replication may have deleterious effects on HIV-1 and other virus-specific CD4(+) responses. Vaccination to stimulate HIV-specific CD4(+) T cell responses in these children may synergize with antiretroviral therapy to improve the long-term control of viral replication, and may perhaps allow the eventual discontinuation of antiretroviral therapy.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/virología , Epítopos de Linfocito T/inmunología , Infecciones por VIH/inmunología , Infecciones por VIH/virología , VIH-1/inmunología , Replicación Viral/inmunología , Adolescente , Adulto , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/patología , División Celular/inmunología , Niño , Preescolar , Enfermedad Crónica , Citomegalovirus/inmunología , Regulación hacia Abajo/inmunología , Dosificación de Gen , Productos del Gen gag/inmunología , Infecciones por VIH/sangre , Infecciones por VIH/patología , VIH-1/genética , VIH-1/aislamiento & purificación , Humanos , Lactante , Interferón gamma/biosíntesis , Activación de Linfocitos , Recuento de Linfocitos , Linfopenia/inmunología , Linfopenia/patología , Linfopenia/virología , Fosfoproteínas/inmunología , Precursores de Proteínas/inmunología , Carga Viral , Proteínas de la Matriz Viral/inmunología
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda