Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Cell Rep ; 43(4): 113981, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38520688

ABSTRACT

Cholera toxin (CT), a bacterial exotoxin composed of one A subunit (CTA) and five B subunits (CTB), functions as an immune adjuvant. CTB can induce production of interleukin-1ß (IL-1ß), a proinflammatory cytokine, in synergy with a lipopolysaccharide (LPS), from resident peritoneal macrophages (RPMs) through the pyrin and NLRP3 inflammasomes. However, how CTB or CT activates these inflammasomes in the macrophages has been unclear. Here, we clarify the roles of inositol-requiring enzyme 1 alpha (IRE1α), an endoplasmic reticulum (ER) stress sensor, in CT-induced IL-1ß production in RPMs. In RPMs, CTB is incorporated into the ER and induces ER stress responses, depending on GM1, a cell membrane ganglioside. IRE1α-deficient RPMs show a significant impairment of CT- or CTB-induced IL-1ß production, indicating that IRE1α is required for CT- or CTB-induced IL-1ß production in RPMs. This study demonstrates the critical roles of IRE1α in activation of both NLRP3 and pyrin inflammasomes in tissue-resident macrophages.


Subject(s)
Cholera Toxin , Endoplasmic Reticulum Stress , Endoribonucleases , Interleukin-1beta , Protein Serine-Threonine Kinases , Interleukin-1beta/metabolism , Animals , Endoribonucleases/metabolism , Protein Serine-Threonine Kinases/metabolism , Endoplasmic Reticulum Stress/drug effects , Mice , Cholera Toxin/pharmacology , Cholera Toxin/metabolism , Inflammasomes/metabolism , Mice, Inbred C57BL , Macrophages/metabolism , Macrophages/drug effects , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/immunology , Lipopolysaccharides/pharmacology , Endoplasmic Reticulum/metabolism
2.
Front Immunol ; 14: 1116238, 2023.
Article in English | MEDLINE | ID: mdl-36891311

ABSTRACT

Background: Adjuvants are chemical or biological materials that enhance the efficacy of vaccines. A-910823 is a squalene-based emulsion adjuvant used for S-268019-b, a novel vaccine against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that is currently in clinical development. Published evidence has demonstrated that A-910823 can enhance the induction of neutralizing antibodies against SARS-CoV-2 in humans and animal models. However, the characteristics and mechanisms of the immune responses induced by A-910823 are not yet known. Methods and Results: To characterize A-910823, we compared the adaptive immune response profile enhanced by A-910823 with that of other adjuvants (AddaVax, QS21, aluminum salt-based adjuvants, and empty lipid nanoparticle [eLNP]) in a murine model. Compared with other adjuvants, A-910823 enhanced humoral immune responses to an equal or greater extent following potent T follicular helper (Tfh) and germinal center B (GCB) cell induction, without inducing a strong systemic inflammatory cytokine response. Furthermore, S-268019-b containing A-910823 adjuvant produced similar results even when given as a booster dose following primary administration of a lipid nanoparticle-encapsulated messenger RNA (mRNA-LNP) vaccine. Preparation of modified A-910823 adjuvants to identify which components of A-910823 play a role in driving the adjuvant effect and detailed evaluation of the immunological characteristics induced by each adjuvant showed that the induction of humoral immunity and Tfh and GCB cell induction in A-910823 were dependent on α-tocopherol. Finally, we revealed that the recruitment of inflammatory cells to the draining lymph nodes and induction of serum cytokines and chemokines by A-910823 were also dependent on the α-tocopherol component. Conclusions: This study demonstrates that the novel adjuvant A-910823 is capable of robust Tfh cell induction and humoral immune responses, even when given as a booster dose. The findings also emphasize that α-tocopherol drives the potent Tfh-inducing adjuvant function of A-910823. Overall, our data provide key information that may inform the future production of improved adjuvants.


Subject(s)
COVID-19 , Immunity, Humoral , Humans , Animals , Mice , T Follicular Helper Cells , alpha-Tocopherol/pharmacology , Squalene/pharmacology , Emulsions , SARS-CoV-2 , Adjuvants, Immunologic/pharmacology , Adjuvants, Pharmaceutic
3.
BMC Cancer ; 22(1): 744, 2022 Jul 07.
Article in English | MEDLINE | ID: mdl-35799134

ABSTRACT

BACKGROUND: Cytosine-phosphate-guanine oligodeoxynucleotide (CpG ODN) (K3)-a novel synthetic single-stranded DNA immune adjuvant for cancer immunotherapy-induces a potential Th1-type immune response against cancer cells. We conducted a phase I study of CpG ODN (K3) in patients with lung cancer to assess its safety and patients' immune responses. METHODS: The primary endpoint was the proportion of dose-limiting toxicities (DLTs) at each dose level. Secondary endpoints included safety profile, an immune response, including dynamic changes in immune cell and cytokine production, and progression-free survival (PFS). In a 3 + 3 dose-escalation design, the dosage levels for CpG ODN (K3) were 5 or 10 mg/body via subcutaneous injection and 0.2 mg/kg via intravenous administration on days 1, 8, 15, and 29. RESULTS: Nine patients (eight non-small-cell lung cancer; one small-cell lung cancer) were enrolled. We found no DLTs at any dose level and observed no serious treatment-related adverse events. The median observation period after registration was 55 days (range: 46-181 days). Serum IFN-α2 levels, but not inflammatory cytokines, increased in six patients after the third administration of CpG ODN (K3) (mean value: from 2.67 pg/mL to 3.61 pg/mL after 24 hours). Serum IFN-γ (mean value, from 9.07 pg/mL to 12.7 pg/m after 24 hours) and CXCL10 levels (mean value, from 351 pg/mL to 676 pg/mL after 24 hours) also increased in eight patients after the third administration. During the treatment course, the percentage of T-bet-expressing CD8+ T cells gradually increased (mean, 49.8% at baseline and 59.1% at day 29, p = 0.0273). Interestingly, both T-bet-expressing effector memory (mean, 52.7% at baseline and 63.7% at day 29, p = 0.0195) and terminally differentiated effector memory (mean, 82.3% at baseline and 90.0% at day 29, p = 0.0039) CD8+ T cells significantly increased. The median PFS was 398 days. CONCLUSIONS: This is the first clinical study showing that CpG ODN (K3) activated innate immunity and elicited Th1-type adaptive immune response and cytotoxic activity in cancer patients. CpG ODN (K3) was well tolerated at the dose settings tested, although the maximum tolerated dose was not determined. TRIAL REGISTRATION: UMIN-CTR number 000023276. Registered 1 September 2016, https://upload.umin.ac.jp/cgi-open-bin/ctr/ctr_view.cgi?recptno=R000026649.


Subject(s)
Antineoplastic Agents , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Adaptive Immunity , Adjuvants, Immunologic/adverse effects , Antineoplastic Agents/pharmacology , CD8-Positive T-Lymphocytes , Carcinoma, Non-Small-Cell Lung/drug therapy , Cytosine , Guanine , Humans , Lung Neoplasms/drug therapy , Oligodeoxyribonucleotides/adverse effects , Phosphates , Toll-Like Receptor 9
4.
Front Immunol ; 13: 847616, 2022.
Article in English | MEDLINE | ID: mdl-35663999

ABSTRACT

Adjuvants are important vaccine components, composed of a variety of chemical and biological materials that enhance the vaccine antigen-specific immune responses by stimulating the innate immune cells in both direct and indirect manners to produce a variety cytokines, chemokines, and growth factors. It has been developed by empirical methods for decades and considered difficult to choose a single screening method for an ideal vaccine adjuvant, due to their diverse biochemical characteristics, complex mechanisms of, and species specificity for their adjuvanticity. We therefore established a robust adjuvant screening strategy by combining multiparametric analysis of adjuvanticity in vivo and immunological profiles in vitro (such as cytokines, chemokines, and growth factor secretion) of various library compounds derived from hot-water extracts of herbal medicines, together with their diverse distribution of nano-sized physical particle properties with a machine learning algorithm. By combining multiparametric analysis with a machine learning algorithm such as rCCA, sparse-PLS, and DIABLO, we identified that human G-CSF and mouse RANTES, produced upon adjuvant stimulation in vitro, are the most robust biological parameters that can predict the adjuvanticity of various library compounds. Notably, we revealed a certain nano-sized particle population that functioned as an independent negative parameter to adjuvanticity. Finally, we proved that the two-step strategy pairing the negative and positive parameters significantly improved the efficacy of screening and a screening strategy applying principal component analysis using the identified parameters. These novel parameters we identified for adjuvant screening by machine learning with multiple biological and physical parameters may provide new insights into the future development of effective and safe adjuvants for human use.


Subject(s)
Adjuvants, Vaccine , Vaccines , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/pharmacology , Adjuvants, Pharmaceutic , Animals , Cytokines , Herbal Medicine , Machine Learning , Mice
5.
Angew Chem Int Ed Engl ; 60(2): 961-969, 2021 01 11.
Article in English | MEDLINE | ID: mdl-32979004

ABSTRACT

Immune potentiators, termed adjuvants, trigger early innate immune responses to ensure the generation of robust and long-lasting adaptive immune responses of vaccines. Presented here is a study that takes advantage of a self-assembling small-molecule library for the development of a novel vaccine adjuvant. Cell-based screening of the library and subsequent structural optimization led to the discovery of a simple, chemically tractable deoxycholate derivative (molecule 6, also named cholicamide) whose well-defined nanoassembly potently elicits innate immune responses in macrophages and dendritic cells. Functional and mechanistic analyses indicate that the virus-like assembly enters the cells and stimulates the innate immune response through Toll-like receptor 7 (TLR7), an endosomal TLR that detects single-stranded viral RNA. As an influenza vaccine adjuvant in mice, molecule 6 was as potent as Alum, a clinically used adjuvant. The studies described here pave the way for a new approach to discovering and designing self-assembling small-molecule adjuvants against pathogens, including emerging viruses.


Subject(s)
Adjuvants, Immunologic/chemistry , Amides/chemistry , Amides/immunology , Amides/pharmacology , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Deoxycholic Acid/chemistry , Drug Evaluation, Preclinical , Fluorescent Dyes/chemistry , Immunity, Innate , Immunoglobulin G/blood , Influenza Vaccines/chemistry , Influenza Vaccines/immunology , Interleukin-6/metabolism , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nanostructures/chemistry , RAW 264.7 Cells , Structure-Activity Relationship , Toll-Like Receptor 7/metabolism
6.
Biologicals ; 55: 43-52, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30017557

ABSTRACT

Recently, many vaccine adjuvants have been developed; however, most of the newly developed adjuvants have been dropped out of preclinical and clinical trials owing to their unexpected toxicity. Thus, the development of highly quantitative and comparable screening methods for evaluating adjuvant safety is needed. In a previous study, we identified specific biomarkers for evaluating the safety of an intranasal influenza vaccine with CpG K3 adjuvant by comparing biomarker expression. We hypothesized that these biomarkers might be useful for screening newly developed adjuvant safety. We compared the expression of biomarkers in mouse lungs by the intranasal administration of 4 types of adjuvants: Alum, Pam3CSK4, NanoSiO2, and DMXAA with subvirion influenza vaccine. The control adjuvant alum did not show any significant increase in biomarker expression or preclinical parameters; however, NanoSiO2 and Pam3CSK4 increased the expression of biomarkers, such as Timp1 and Csf1. DMXAA at 300 µg induced the expression of over 80% of biomarkers. Hierarchical clustering analysis showed that 300 µg DMXAA was classified in the toxicity reference whole-particle influenza vaccine cluster. FACS analysis to confirm specific phenotypes that the number of T cells decreased in DMXAA-treated mouse lungs. Thus, our biomarkers are useful for initial adjuvant safety and toxicity screening.


Subject(s)
Adjuvants, Immunologic , Influenza A Virus, H1N1 Subtype , Influenza Vaccines , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/pharmacology , Administration, Intranasal , Animals , Drug Evaluation, Preclinical , Female , Influenza A Virus, H1N1 Subtype/chemistry , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/chemistry , Influenza Vaccines/immunology , Influenza Vaccines/pharmacology , Mice , Mice, Inbred BALB C
7.
Adv Exp Med Biol ; 1031: 55-94, 2017.
Article in English | MEDLINE | ID: mdl-29214566

ABSTRACT

Public health relies on technologies to produce and analyse data, as well as effectively develop and implement policies and practices. An example is the public health practice of epidemiology, which relies on computational technology to monitor the health status of populations, identify disadvantaged or at risk population groups and thereby inform health policy and priority setting. Critical to achieving health improvements for the underserved population of people living with rare diseases is early diagnosis and best care. In the rare diseases field, the vast majority of diseases are caused by destructive but previously difficult to identify protein-coding gene mutations. The reduction in cost of genetic testing and advances in the clinical use of genome sequencing, data science and imaging are converging to provide more precise understandings of the 'person-time-place' triad. That is: who is affected (people); when the disease is occurring (time); and where the disease is occurring (place). Consequently we are witnessing a paradigm shift in public health policy and practice towards 'precision public health'.Patient and stakeholder engagement has informed the need for a national public health policy framework for rare diseases. The engagement approach in different countries has produced highly comparable outcomes and objectives. Knowledge and experience sharing across the international rare diseases networks and partnerships has informed the development of the Western Australian Rare Diseases Strategic Framework 2015-2018 (RD Framework) and Australian government health briefings on the need for a National plan.The RD Framework is guiding the translation of genomic and other technologies into the Western Australian health system, leading to greater precision in diagnostic pathways and care, and is an example of how a precision public health framework can improve health outcomes for the rare diseases population.Five vignettes are used to illustrate how policy decisions provide the scaffolding for translation of new genomics knowledge, and catalyze transformative change in delivery of clinical services. The vignettes presented here are from an Australian perspective and are not intended to be comprehensive, but rather to provide insights into how a new and emerging 'precision public health' paradigm can improve the experiences of patients living with rare diseases, their caregivers and families.The conclusion is that genomic public health is informed by the individual and family needs, and the population health imperatives of an early and accurate diagnosis; which is the portal to best practice care. Knowledge sharing is critical for public health policy development and improving the lives of people living with rare diseases.


Subject(s)
Genomics/methods , Health Policy , Precision Medicine , Public Health , Rare Diseases/therapy , Genetic Predisposition to Disease , Genomics/organization & administration , Health Policy/legislation & jurisprudence , Humans , Phenotype , Policy Making , Predictive Value of Tests , Prognosis , Program Development , Program Evaluation , Public Health/legislation & jurisprudence , Rare Diseases/diagnosis , Rare Diseases/epidemiology , Rare Diseases/genetics
8.
Sci Immunol ; 2(12)2017 Jun 02.
Article in English | MEDLINE | ID: mdl-28783657

ABSTRACT

Although malaria is a life-threatening disease with severe complications, most people develop partial immunity and suffer from mild symptoms. However, incomplete recovery from infection causes chronic illness, and little is known of the potential outcomes of this chronicity. We found that malaria causes bone loss and growth retardation as a result of chronic bone inflammation induced by Plasmodium products. Acute malaria infection severely suppresses bone homeostasis, but sustained accumulation of Plasmodium products in the bone marrow niche induces MyD88-dependent inflammatory responses in osteoclast and osteoblast precursors, leading to increased RANKL expression and overstimulation of osteoclastogenesis, favoring bone resorption. Infection with a mutant parasite with impaired hemoglobin digestion that produces little hemozoin, a major Plasmodium by-product, did not cause bone loss. Supplementation of alfacalcidol, a vitamin D3 analog, could prevent the bone loss. These results highlight the risk of bone loss in malaria-infected patients and the potential benefits of coupling bone therapy with antimalarial treatment.

9.
Vaccine ; 35(5): 821-830, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28063707

ABSTRACT

Vaccines are administered to healthy humans, including infants, so the safety and efficacy must be very high. Therefore, evaluating vaccine safety in preclinical and clinical studies, according to World Health Organization guidelines, is crucial for vaccine development and clinical use. A change in the route of administration is considered to alter a vaccine's immunogenicity. Several adjuvants have also been developed and approved for use in vaccines. However, the addition of adjuvants to vaccines may cause unwanted immune responses, including facial nerve paralysis and narcolepsy. Therefore, a more accurate and comprehensive strategy must be used to develope next-generation vaccines for ensuring vaccine safety. Previously, we have developed a system with which to evaluate vaccine safety in rats using a systematic vaccinological approach and 20 marker genes. In this study, we developed a safety evaluation system for nasally administered influenza vaccines and adjuvanted influenza vaccines using these marker genes. Expression of these genes increased dose-dependent manner when mice were intranasally administered the toxicity reference vaccine. When the adjuvant CpG K3 or a CpG-K3-combined influenza vaccine was administered intranasally, marker gene expression increased in a CpG-K3-dose-dependent way. A histopathological analysis indicated that marker gene expression correlated with vaccine- or adjuvant-induced phenotypic changes in the lung and nasal mucosa. We believe that the marker genes expression analyses will be useful in preclinical testing, adjuvant development, and selecting the appropriate dose of adjuvant in nasal administration vaccines.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antibodies, Viral/biosynthesis , Influenza Vaccines/administration & dosage , Oligodeoxyribonucleotides/administration & dosage , Orthomyxoviridae Infections/prevention & control , Administration, Intranasal , Animals , Biomarkers/metabolism , Cytokines/genetics , Cytokines/immunology , Dose-Response Relationship, Immunologic , Drug Evaluation, Preclinical , Female , Gene Expression , Humans , Membrane Transport Proteins/genetics , Membrane Transport Proteins/immunology , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Vaccines, Inactivated
10.
Immunity ; 45(6): 1299-1310, 2016 12 20.
Article in English | MEDLINE | ID: mdl-28002730

ABSTRACT

Particulate pollution is thought to function as an adjuvant that can induce allergic responses. However, the exact cell types and immunological factors that initiate the lung-specific immune responses are unclear. We found that upon intratracheal instillation, particulates such as aluminum salts and silica killed alveolar macrophages (AMs), which then released interleukin-1α (IL-1α) and caused inducible bronchus-associated lymphoid tissue (iBALT) formation in the lung. IL-1α release continued for up to 2 weeks after particulate exposure, and type-2 allergic immune responses were induced by the inhalation of antigen during IL-1α release and iBALT formation, even long after particulate instillation. Recombinant IL-1α was sufficient to induce iBALTs, which coincided with subsequent immunoglobulin E responses, and IL-1-receptor-deficient mice failed to induce iBALT formation. Therefore, the AM-IL-1α-iBALT axis might be a therapeutic target for particulate-induced allergic inflammation.


Subject(s)
Bronchi/immunology , Interleukin-1alpha/immunology , Lymphoid Tissue/immunology , Macrophages, Alveolar/pathology , Particulate Matter/toxicity , Aluminum Compounds/toxicity , Animals , Female , Mice , Mice, Inbred C57BL , Silicon Dioxide/toxicity
11.
Int Immunol ; 28(7): 329-38, 2016 07.
Article in English | MEDLINE | ID: mdl-27006304

ABSTRACT

Accumulated evidence obtained from various clinical trials and animal studies suggested that cancer vaccines need better adjuvants than those that are currently licensed, which include the most commonly used alum and incomplete Freund's adjuvant, because of either a lack of potent anti-tumor immunity or the induction of undesired immunity. Several clinical trials using immunostimulatory adjuvants, particularly agonistic as well as non-agonistic ligands for TLRs, C-type lectin receptors, retinoic acid-inducible gene I-like receptors and stimulator of interferon genes, have revealed their therapeutic potential not only as vaccine adjuvants but also as anti-tumor agents. Recently, combinations of such immunostimulatory or immunomodulatory adjuvants have shown superior efficacy over their singular use, suggesting that seeking optimal combinations of the currently available or well-characterized adjuvants may provide a better chance for the development of novel adjuvants for cancer immunotherapy.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Alum Compounds/therapeutic use , Cancer Vaccines/immunology , Immunotherapy/methods , Lectins, C-Type/therapeutic use , Neoplasms/therapy , Oligodeoxyribonucleotides/therapeutic use , Animals , Clinical Trials as Topic , Humans , Immunomodulation , Neoplasms/immunology , Vaccination
12.
Vaccine ; 34(3): 306-12, 2016 Jan 12.
Article in English | MEDLINE | ID: mdl-26667613

ABSTRACT

Various particles such as Alum or silica are known to act as an adjuvant if co-administered with vaccine antigens. Several reports have demonstrated that the adjuvanticity is strongly affected by the physicochemical properties of particles such as the size, shape and surface charge, although the required properties and its relationship to the adjuvanticity are still controversial. Hydroxyapatite particle (HAp) composed of calcium phosphate has been shown to work as adjuvant in mice. However, the properties of HAp required for the adjuvanticity have not been fully characterized yet. In this study, we examined the role of size or shape of HAps in the antibody responses after immunization with antigen. HAps whose diameter ranging between 100 and 400 nm provided significantly higher antibody responses than smaller or larger ones. By comparison between sphere and rod shaped HAps, rod shaped HAps induced stronger inflammasome-dependent IL-1ß production than the sphere shaped ones in vitro. However, sphere- and rod-shaped HAp elicited comparable antibody response in WT mice. Vice versa, Nlrp3(-/-), Asc(-/-) or Caspase1(-/-) mice provided comparable level of antibody responses to HAp adjuvanted vaccination. Collectively, our results demonstrated that the size rather than shape is a more critical property, and IL-1ß production via NLRP3 inflammasome is dispensable for the adjuvanticity of HAps in mice.


Subject(s)
Adjuvants, Immunologic/pharmacology , Chemical Phenomena , Durapatite/pharmacology , Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Durapatite/administration & dosage , Female , Mice, Inbred C57BL , Nanoparticles/administration & dosage , Vaccines/administration & dosage
13.
Vaccine ; 30(52): 7658-61, 2012 Dec 14.
Article in English | MEDLINE | ID: mdl-23084848

ABSTRACT

The effectiveness of influenza vaccines is still controversial, and the role of adjuvants in such vaccines is briefly reviewed in this paper. Inactivated whole virus vaccines may include components that function as adjuvants, meaning that additive adjuvants are often not required. MF59 and AS03 showed higher adjuvanticity than aluminum salts in several clinical studies. Recent research has suggested that immune cell recruitment is the main mechanism underlying adjuvant actions in general, and that aluminum salts induce this recruitment via inflammation at the injected site. The aspect of how oil-based adjuvants, such as MF59 and AS03, recruit immune cells remains to be clarified.


Subject(s)
Adjuvants, Immunologic/pharmacology , Influenza Vaccines/immunology , Alum Compounds/pharmacology , Drug Combinations , Humans , Polysorbates/pharmacology , Squalene/pharmacology , Vaccines, Inactivated/immunology , alpha-Tocopherol/pharmacology
14.
J Allergy Clin Immunol ; 130(1): 184-94.e11, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22460070

ABSTRACT

BACKGROUND: We reported previously that serum levels of IL-33 are significantly increased in patients with allergic rhinitis (AR). However, very little is known about the role of IL-33 for the development of AR. OBJECTIVE: We thought to develop a novel murine model of ragweed pollen-specific AR and examined the pathologic role for ragweed-induced IL-33 in the development of AR manifestation using IL-33-deficient (il33(-/-)) mice. METHODS: Ragweed-immunized and ragweed-challenged mice were examined for early- and late-phase nasal responses. IL-33 protein expression in the nasal epithelial cells of the AR murine model and patients with AR were assessed by using confocal microscopy. RESULTS: After nasal challenge with ragweed pollen, ragweed-immunized wild-type mice manifested early-phase (sneezing) and late-phase (eosinophilic and basophilic accumulation) responses. In contrast, il33(-/-) and FcεRI(-/-) mice did not have both early- and late-phase AR responses. IL-33 protein was constitutively expressed in the nucleus of nasal epithelial cells and was promptly released into nasal fluids in response to nasal exposure to ragweed pollen. In human subjects we revealed constitutive expression of IL-33 protein in the nasal epithelial cells of healthy control subjects and downregulated expression of IL-33 protein in inflamed nasal epithelial cells of patients with AR. IL-33-stimulated mast cells and basophils contributed to the early- and late-phase AR manifestation through increasing histamine release and production of chemoattractants for eosinophils/basophils, respectively. CONCLUSIONS: Ragweed pollen-driven endogenous IL-33 contributed to the development of AR responses. IL-33 might present an important therapeutic target for the prevention of AR.


Subject(s)
Ambrosia/immunology , Disease Models, Animal , Hypersensitivity, Immediate/immunology , Interleukins/metabolism , Pollen/immunology , Rhinitis/immunology , Animals , Basophils/immunology , Eosinophils/immunology , Humans , Hypersensitivity, Immediate/pathology , Interleukins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Nasal Mucosa/immunology , Nasal Mucosa/pathology , Nasal Provocation Tests
15.
Parasitol Int ; 59(3): 380-6, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20493274

ABSTRACT

An effective malaria vaccine is a public health priority. Proteins expressed during the blood-stage of the parasite life cycle have been proposed as good vaccine candidates. No such blood-stage vaccine, however, is available against Plasmodium falciparum, the deadliest Plasmodium species. We show here that P. falciparum serine repeat antigen 5 (SERA5) is a potential vaccine immunogen. We have constructed a new recombinant molecule of SERA5, namely SE36, based on previously reported SE47' molecule by removing the serine repeats. Epidemiological study in the holo-endemic population of Solomon Islands shows highly significant correlation of sero-conversion and malaria protective immunity against this antigen. Animal experiments using non-human primates, and a human phase 1a clinical trial assessed SE36 vaccine immunogenicity. Vaccination of squirrel monkeys with SE36 protein and aluminum hydroxyl gel (SE36/AHG) conferred protection against high parasitemia and boosted serum anti-SE36 IgG after P. falciparum parasite challenge. SE36/AHG was highly immunogenic in chimpanzees, where serum anti-SE36 IgG titers last more than one year. Phase 1a clinical trial (current controlled trials, ISRCTN78679862) demonstrated the safety and immunogenicity of SE36/AHG with 30 healthy adults and 10 placebo controls. Three subcutaneous administrations of 50 and 100microg dose of SE36/AHG were well-tolerated, with no severe adverse events; and resulted in 100% sero-conversion in both dose arms. The current research results for SE36/AHG provide initial clinical validation for future trials and suggest clues/strategies for further vaccine development.


Subject(s)
Antigens, Protozoan/immunology , Immunoglobulin G/blood , Malaria Vaccines/immunology , Malaria, Falciparum/parasitology , Parasitemia/prevention & control , Adult , Animals , Antigens, Protozoan/genetics , Drug Evaluation, Preclinical , Humans , Malaria Vaccines/administration & dosage , Malaria Vaccines/genetics , Malaria, Falciparum/epidemiology , Malaria, Falciparum/prevention & control , Melanesia/epidemiology , Parasitemia/epidemiology , Parasitemia/parasitology , Plasmodium falciparum/immunology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Saimiri , Treatment Outcome , Vaccination
16.
Expert Rev Vaccines ; 8(8): 1099-107, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19627190

ABSTRACT

Optimal vaccine efficacy requires not only a protective antigen, but also a strong immune activator as an adjuvant. Most viral vaccines, such as influenza vaccines and nonviral genetic vaccines (e.g., DNA vaccines), contain nucleic acids, which appear to act as essential 'built-in' adjuvants. Specific receptors, including Toll-like receptors, retinoic acid-inducible protein-I-like receptors, and nucleotide-binding oligomerization domain-like receptors can detect specific nucleic acid patterns, depending on the immunized tissue, cell type and intracellular localization. The resulting immune activation is uniquely regulated by intra- and intercellular signaling pathways, which are indispensable for the ensuing vaccine immunogenicity, such as antigen-specific T- and B-cell responses. Thus, elucidation and manipulation of immune signaling and interactions by nucleic acid adjuvants are essential for maximizing the immunogenicity and safety of viral and DNA vaccines.


Subject(s)
Immunity, Innate , Vaccines, DNA/immunology , Viral Vaccines/immunology , Adjuvants, Immunologic/pharmacology , Humans , Nucleic Acids/immunology , Nucleic Acids/pharmacology
17.
J Clin Immunol ; 27(4): 363-71, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17370119

ABSTRACT

Successful vaccines contain an adjuvant component that activates the innate immune system, thereby eliciting antigen-specific immune responses. Many adjuvants appear to be ligands for toll-like receptors (TLR), which are thus promising targets for the development of novel adjuvants to elicit vaccine immunogenicity. However, recent evidence suggests that some adjuvants activate the innate immune system in a TLR-independent manner possibly through other pattern recognition receptors and signaling machinery. In particular, newly identified intracellular retinoic-acid-inducible gene (RIG)-like receptors, NOD-like receptors, or even as yet unknown recognition machinery for the adjuvant may regulate TLR-independent vaccine immunogenicity. To develop optimal vaccines, it will be critical to understand how TLR-dependent and TLR-independent innate immune activation, by various adjuvants, control the consequent adaptive immune responses to vaccine.


Subject(s)
Adjuvants, Immunologic , Toll-Like Receptors/physiology , Vaccines , Humans , Immunity, Innate/drug effects , Immunization , Signal Transduction/immunology , Toll-Like Receptors/immunology , Vaccines/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL