Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters











Publication year range
1.
PLoS Negl Trop Dis ; 12(12): e0006975, 2018 12.
Article in English | MEDLINE | ID: mdl-30557313

ABSTRACT

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.


Subject(s)
Cytokines/analysis , Dengue Virus/immunology , Dengue/immunology , Asymptomatic Infections , Cohort Studies , Cross Reactions , Dengue/virology , Humans , Leukocytes, Mononuclear , Thailand/epidemiology
2.
Infect Immun ; 86(12)2018 12.
Article in English | MEDLINE | ID: mdl-30249744

ABSTRACT

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.


Subject(s)
Ascariasis/physiopathology , Hypersensitivity/parasitology , Lung/pathology , Respiratory Hypersensitivity/parasitology , Animals , Ascariasis/immunology , Ascaris/pathogenicity , Bronchoalveolar Lavage Fluid/immunology , Disease Models, Animal , Female , Interleukin-13/immunology , Interleukin-4/immunology , Interleukin-5/immunology , Larva/pathogenicity , Lung/parasitology , Mice , Mice, Inbred BALB C , Ovalbumin , Th2 Cells/immunology
3.
PLoS Pathog ; 14(8): e1007273, 2018 08.
Article in English | MEDLINE | ID: mdl-30153307

ABSTRACT

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.


Subject(s)
Immunity , Milk Proteins/immunology , Trichuriasis/prevention & control , Trichuris/immunology , Animals , Antibodies, Helminth/metabolism , Antigens, Helminth/genetics , Antigens, Helminth/immunology , Antigens, Helminth/pharmacology , Immunity/drug effects , Immunity/genetics , Male , Mice , Mice, Inbred AKR , Mice, Knockout , Mice, SCID , Milk Proteins/genetics , Milk Proteins/pharmacology , Trichuriasis/immunology , Trichuris/genetics , Vaccination/methods
4.
PLoS Negl Trop Dis ; 11(7): e0005769, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28708895

ABSTRACT

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.


Subject(s)
Antibodies, Helminth/blood , Antigens, Helminth/therapeutic use , Ascariasis/prevention & control , Th2 Cells/immunology , Vaccines/therapeutic use , Adjuvants, Immunologic , Animals , Antigens, Helminth/immunology , Ascaris suum , Cholera Toxin/immunology , Female , Immunoglobulin G/blood , Interleukin-4/immunology , Interleukin-5/immunology , Larva , Mice , Mice, Inbred BALB C , Recombinant Proteins/therapeutic use , Saccharomyces cerevisiae , Sequence Analysis , Vaccination
5.
J Immunol Methods ; 440: 74-82, 2017 01.
Article in English | MEDLINE | ID: mdl-27840065

ABSTRACT

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.


Subject(s)
Antigens, Helminth/immunology , Ascaris suum/immunology , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Enzyme-Linked Immunosorbent Assay/instrumentation , Helminth Proteins/immunology , Spleen/immunology , Animals , Antigens, Helminth/administration & dosage , Biomarkers/metabolism , Cells, Cultured , Culture Media, Conditioned/metabolism , Cytokines/blood , Encephalomyelitis, Autoimmune, Experimental/blood , Enzyme-Linked Immunosorbent Assay/methods , Equipment Design , Female , Helminth Proteins/administration & dosage , Immunization , Mice, Inbred BALB C , Reproducibility of Results , Spleen/metabolism , Time Factors , Workflow
6.
Vaccine ; 34(26): 3001-3005, 2016 06 03.
Article in English | MEDLINE | ID: mdl-27040400

ABSTRACT

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.


Subject(s)
Anemia/prevention & control , Hookworm Infections/prevention & control , Vaccines/therapeutic use , Anemia/parasitology , Animals , Biomedical Research/trends , Clinical Trials as Topic , Dogs , Hookworm Infections/complications , Humans , Necator americanus
7.
Vaccine ; 34(26): 2988-2991, 2016 06 03.
Article in English | MEDLINE | ID: mdl-27036511

ABSTRACT

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.


Subject(s)
Schistosomiasis/prevention & control , Vaccines/therapeutic use , Animals , Antigens, Bacterial/immunology , Antigens, Helminth/immunology , Biomedical Research/trends , Clinical Trials as Topic , Fatty Acid Transport Proteins/immunology , Glutathione Transferase/immunology , Helminth Proteins/immunology , Humans , Primates , Schistosoma haematobium , Schistosoma mansoni , Tetraspanins/immunology , Vaccines, Synthetic/therapeutic use
8.
Vaccine ; 34(26): 2992-2995, 2016 06 03.
Article in English | MEDLINE | ID: mdl-26973063

ABSTRACT

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.


Subject(s)
Leishmaniasis Vaccines/therapeutic use , Leishmaniasis/prevention & control , Animals , Biomedical Research/trends , Clinical Trials as Topic , Humans , Leishmaniasis, Cutaneous/prevention & control , Leishmaniasis, Visceral/prevention & control , Psychodidae/parasitology , Vaccines, Synthetic/therapeutic use
10.
Hum Vaccin Immunother ; 12(4): 976-87, 2016 04 02.
Article in English | MEDLINE | ID: mdl-26890466

ABSTRACT

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.


Subject(s)
Chagas Disease/therapy , Protozoan Vaccines/therapeutic use , Trypanosoma cruzi/immunology , Vaccines, DNA/therapeutic use , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Protozoan/immunology , CD8-Positive T-Lymphocytes/immunology , Chagas Cardiomyopathy/therapy , Chagas Disease/immunology , Disease Models, Animal , Female , Heart/parasitology , Immunity, Cellular , Immunoglobulin G/biosynthesis , Immunoglobulin G/immunology , Immunotherapy/methods , Interferon-gamma/immunology , Mice , Mice, Inbred BALB C , Nanoparticles , Oligodeoxyribonucleotides/immunology , Parasitemia/therapy , Protozoan Vaccines/immunology , Th1 Cells/immunology , Vaccines, DNA/adverse effects , Vaccines, DNA/immunology
SELECTION OF CITATIONS
SEARCH DETAIL