Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 551
Filter
Add more filters

Publication year range
1.
Toxicol Mech Methods ; 34(7): 813-820, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38717917

ABSTRACT

For nearly 90 years, aluminum (Al) salts have been utilized as vaccination adjuvants. Nevertheless, there is a risk of adverse effects associated with the amount of nanoaluminum used in various national pediatric immunization regimens. This study aimed to investigate the possible genotoxic effects of nanoaluminum incorporated in human vaccines on the brains of newborn albino rats and whether nanocurcumin has a potential protective effect against this toxicity. Fifty newborn albino rats were randomly assigned to 5 groups, with 10 in each group. Groups 1 and 2 received "high" and "low" Al injections corresponding to either the American or Scandinavian pediatric immunization schedules, respectively, as opposed to the control rats (group 5) that received saline injections. Groups 3 and 4 received the same regimens as groups 1 and 2 in addition to oral nanocurcumin. The expression of both the cell breakdown gene tumor protein (P53) and the cell stress gene uncoupling protein 2 (UCP2) was significantly greater in groups 1 and 2 than in group 5. Groups 1 and 2 exhibited severe DNA fragmentation, which was observed as DNA laddering. Nanocurcumin significantly reduced the expression of the P53 and UCP2 genes in groups 3 and 4, with very low or undetectable DNA laddering in both groups. Vaccination with nanoaluminum adjuvants can cause genotoxic effects, which can be mediated by the inflammatory response and oxidative stress, and nanocurcumin can protect against these toxic effects through the modulation of oxidative stress regulators and gene expression.


Subject(s)
Adjuvants, Immunologic , Curcumin , Animals , Rats , Adjuvants, Immunologic/toxicity , Aluminum Compounds/toxicity , Animals, Newborn , Brain/drug effects , Brain/metabolism , Brain/pathology , Curcumin/pharmacology , Curcumin/chemistry , DNA Damage/drug effects , DNA Fragmentation/drug effects , Nanoparticles/toxicity , Rats, Wistar , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Vaccines/toxicity
2.
Drug Chem Toxicol ; 46(3): 557-565, 2023 May.
Article in English | MEDLINE | ID: mdl-35484852

ABSTRACT

The hemolytic activity, in vitro as well as in vivo toxicity, and immunomodulatory potential of saponins-rich fraction of Asparagus adscendens Roxb. fruit (AA-SRF) have been assessed in this study in order to explore AA-SRF as an alternative safer adjuvant to standard Quil-A saponin. The AA-SRF showed lower hemolytic activity (HD50 = 301.01 ± 1.63 µg/ml) than Quil-A (HD50 = 17.15 ± 2.12 µg/ml). The sulforhodamine B assay also revealed that AA-SRF was less toxic to VERO cells (IC50≥200 ± 4.32 µg/ml) than Quil-A (IC50 = 60 ± 2.78 µg/ml). The AA-SRF did not lead to mortality in mice up to 1.6 mg and was much safer than Quil-A for in vivo use. Conversely, mice were subcutaneously immunized with OVA 100 µg alone or along with Alum (200 µg) or Quil-A (10 µg) or AA-SRF (50 µg/100 µg/200 µg) on days 0 and 14. The AA-SRF at 100 µg dose best supported the LPS/Con A primed splenocyte proliferation activity, elevated the serum OVA-specific total IgG antibody, IL-12, CD4 titer and upsurged CD3/CD19 expression in spleen as well as lymph node sections which in turn advocated its adjuvant potential. Thus, AA-SRF can be further studied for use as a safe alternative adjuvant in vaccines.


Subject(s)
Adjuvants, Immunologic , Asparagus Plant , Saponins , Animals , Mice , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/toxicity , Chlorocebus aethiops , Fruit , Immunoglobulin G , Ovalbumin , Saponins/immunology , Saponins/pharmacology , Saponins/toxicity , Vero Cells
3.
Crit Rev Toxicol ; 52(6): 403-419, 2022 07.
Article in English | MEDLINE | ID: mdl-36112128

ABSTRACT

Aluminum (Al) salts are commonly used as adjuvants in human and veterinary vaccines for almost a century. Despite this long history of use and the very large number of exposed individuals, data in the literature concerning the fate of these molecules after injection and their potential effects on the nervous system is limited. In the context of (i) an increase of exposure to Al salts through vaccination; (ii) the absence of safety values determined by health regulators; (iii) the lack of robustness of the studies used as references to officially claim Al adjuvant innocuity; (iv) the publication of several animal studies investigating Al salts clearance/biopersistence and neurotoxicity; we have examined in this review all published studies performed on animals and assessing Al adjuvants kinetics, biodistribution, and neuromodulation since the first work of A. Glenny in the 1920s. The diversity of methodological approaches, results, and potential weaknesses of the 31 collected studies are exposed. A large range of protocols has been used, including a variety of exposure schedule and analyses methods, making comparisons between studies uneasy. Nevertheless, published data highlight that when biopersistence, translocation, or neuromodulation were assessed, they were documented whatever the different in vivo models and methods used. Moreover, the studies pointed out the crucial importance of the different Al adjuvant physicochemical properties and host genetic background on their kinetics, biodistribution, and neuromodulatory effects. Regarding the state of the art on this key public health topic, further studies are clearly needed to determine the exact safety level of Al salts.


Subject(s)
Aluminum , Salts , Animals , Humans , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/toxicity , Aluminum/toxicity , Kinetics , Tissue Distribution
4.
Clin Exp Pharmacol Physiol ; 49(3): 406-418, 2022 03.
Article in English | MEDLINE | ID: mdl-34796981

ABSTRACT

Cardiac dysfunction is one of the leading causes of death in epilepsy. The anti-arrhythmic drug, amiodarone, is under investigation for its therapeutic effects in epilepsy. We aimed to evaluate the possible effects of amiodarone on cardiac injury during status epilepticus, as it can cause prolongation of the QT interval. Five rat groups were enrolled in the study; three control groups (1) Control, (2) Control-lithium and (3) Control-Amio, treated with 150 mg/kg/intraperitoneal amiodarone, (4) Epilepsy model, induced by sequential lithium/pilocarpine administration, and (5) the epilepsy-Amio group. The model group expressed a typical clinical picture of epileptiform activity confirmed by the augmented electroencephalogram alpha and beta spikes. The anticonvulsive effect of amiodarone was prominent, it diminished (p < 0.001) the severity of seizures and hence, deaths and reduced serum noradrenaline levels. In the model group, the electrocardiogram findings revealed tachycardia, prolongation of the corrected QT (QTc) interval, depressed ST segments and increased myocardial oxidative stress. The in-vitro myocardial performance (contraction force and - (df/dt)max ) was also reduced. Amiodarone decreased (p < 0.001) the heart rate, improved ST segment depression, and myocardial contractility with no significant change in the duration of the QTc interval. Amiodarone preserved the cardiac histological structure and reduced the myocardial injury markers represented by serum Troponin-I, oxidative stress and IL-1. Amiodarone pretreatment prevented the anticipated cardiac injury induced during epilepsy. Amiodarone possessed an anticonvulsive potential, protected the cardiac muscle and preserved its histological architecture. Therefore, amiodarone could be recommended as a protective therapy against cardiac dysfunction during epileptic seizures with favourable effect on seizure activity.


Subject(s)
Amiodarone/therapeutic use , Anti-Arrhythmia Agents/therapeutic use , Epilepsy/complications , Heart Diseases/drug therapy , Heart Diseases/etiology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/toxicity , Animals , Biomarkers/blood , Epilepsy/chemically induced , Glutathione/blood , Interleukin-1/metabolism , Lithium Chloride/administration & dosage , Lithium Chloride/toxicity , Male , Malondialdehyde/blood , Muscarinic Agonists/administration & dosage , Muscarinic Agonists/toxicity , Myocardial Contraction/drug effects , Pilocarpine/administration & dosage , Pilocarpine/toxicity , Rats , Rats, Wistar , Superoxide Dismutase/blood , Troponin I/blood
5.
Am J Pathol ; 190(2): 286-294, 2020 02.
Article in English | MEDLINE | ID: mdl-31734231

ABSTRACT

Murine models of Mycobacterium tuberculosis (Mtb) infection demonstrate progression of M1-like (proinflammatory) and M2-like (anti-inflammatory) macrophage morphology following primary granuloma formation. The Mtb cell wall cording factor, trehalose 6,6'-dimycolate (TDM), is a physiologically relevant and useful molecule for modeling early macrophage-mediated events during establishment of the tuberculosis-induced granuloma pathogenesis. Here, it is shown that TDM is a major driver of the early M1-like macrophage response as seen during initiation of the granulomas of primary pathology. Proinflammatory cytokines tumor necrosis factor-α, IL-1ß, IL-6, and IL-12p40 are produced in lung tissue after administration of TDM to mice. Furthermore, CD11b+CD45+ macrophages with a high surface expression of the M1-like markers CD38 and CD86 were found present in regions of pathology in lungs of mice at 7 days post-TDM introduction. Conversely, only low phenotypic marker expression of M2-like markers CD206 and EGR-2 were present on macrophages. These findings suggest that TDM plays a role in establishment of the M1-like shift in the microenvironment during primary tuberculosis.


Subject(s)
Adjuvants, Immunologic/toxicity , Cord Factors/toxicity , Granuloma/pathology , Inflammation Mediators/metabolism , Macrophages/pathology , Mycobacterium/metabolism , Pneumonia/pathology , Animals , Female , Granuloma/chemically induced , Granuloma/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Pneumonia/chemically induced , Pneumonia/metabolism
6.
Int J Toxicol ; 40(5): 442-452, 2021 10.
Article in English | MEDLINE | ID: mdl-34281421

ABSTRACT

AV7909 is a next-generation anthrax vaccine under development for post-exposure prophylaxis following suspected or confirmed Bacillus anthracis exposure, when administered in conjunction with the recommended antibacterial regimen. AV7909 consists of the FDA-approved BioThrax® vaccine (anthrax vaccine adsorbed) and an immunostimulatory Toll-like receptor 9 agonist oligodeoxynucleotide adjuvant, CPG 7909. The purpose of this study was to evaluate the potential systemic and local toxicity of AV7909 when administered via repeat intramuscular injection to the right thigh muscle (biceps femoris) to male and female Sprague Dawley rats. The vaccine was administered on Days 1, 15, and 29 and the animals were assessed for treatment-related effects followed by a 2-week recovery period to evaluate the persistence or reversibility of any toxic effects. The AV7909 vaccine produced no apparent systemic toxicity based on evaluation of clinical observations, body weights, body temperature, clinical pathology, and anatomic pathology. Necrosis and inflammation were observed at the injection sites as well as in regional lymph nodes and adjacent tissues and were consistent with immune stimulation. Antibodies against B. anthracis protective antigen (PA) were detected in rats treated with the AV7909 vaccine, confirming relevance of this animal model for the assessment of systemic toxicity of AV7909. In contrast, sera of rats that received saline or soluble CPG 7909 alone were negative for anti-PA antibodies. Overall, 3 intramuscular immunizations of Sprague Dawley rats with AV7909 were well tolerated, did not induce mortality or any systemic adverse effects, and did not result in any delayed toxicity.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Anthrax Vaccines/administration & dosage , Oligodeoxyribonucleotides/administration & dosage , Adjuvants, Immunologic/toxicity , Animals , Anthrax Vaccines/toxicity , Antibodies, Bacterial/blood , Antibodies, Neutralizing/blood , Antigens, Bacterial/immunology , Bacterial Toxins/immunology , Female , Injection Site Reaction/blood , Injection Site Reaction/etiology , Injection Site Reaction/immunology , Injection Site Reaction/pathology , Injections, Intramuscular , Male , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Oligodeoxyribonucleotides/toxicity , Post-Exposure Prophylaxis , Rats, Sprague-Dawley
7.
J Neuroinflammation ; 17(1): 34, 2020 Jan 24.
Article in English | MEDLINE | ID: mdl-31980031

ABSTRACT

BACKGROUND: Chronic pain usually accompanied by tissue damage and inflammation. However, the pathogenesis of chronic pain remains unclear. METHODS: We investigated the role of nerve growth factor (NGF) in chronic inflammatory pain induced by complete Freund's adjuvant (CFA), explored the methylation status of CpG islands in the promoter region of the NGF gene, and clarified the function and mechanism of C/EBPα-NGF signaling pathway from epigenetic perspective in the chronic inflammatory pain model. RESULTS: CFA induced significant hyperalgesia and continuous upregulation of NGF mRNA and protein levels in the L4-6 dorsal root ganglions (DRGs) in rats. Hypomethylation of CpG islands occurred in the NGF gene promoter region after CFA treatment. At the same time, the miR-29b expression level was significantly increased, while the DNA methyltransferase 3b (DNMT3b) level reduced significantly. Moreover, CFA treatment promoted binding of C/EBPα to the NGF gene promoter region and C/EBPα siRNA treatment obviously decreased expression of NGF levels and also alleviate inflammatory hyperalgesia significantly in rats. CONCLUSION: Collectively, the results indicated that CFA leads to the upregulation of miR-29b level, which represses the expression of DNMT3b, enhances the demethylation of the NGF gene promoter region, and promotes the binding of C/EBPα with the NGF gene promoter, thus results in the upregulation of NGF gene expression and maintenance of chronic inflammatory pain.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , DNA Methylation , Gene Expression Regulation/physiology , Hyperalgesia , Inflammation , Nerve Growth Factor/physiology , Adjuvants, Immunologic/toxicity , Animals , Freund's Adjuvant/toxicity , Hyperalgesia/genetics , Hyperalgesia/metabolism , Inflammation/chemically induced , Inflammation/genetics , Inflammation/metabolism , Male , MicroRNAs/metabolism , Promoter Regions, Genetic/genetics , Rats , Rats, Sprague-Dawley
8.
FASEB J ; 33(6): 6829-6837, 2019 06.
Article in English | MEDLINE | ID: mdl-30811953

ABSTRACT

Although prostaglandins (PGs) are known to be involved in the progression of arthritis, the role of PGD2 remains unclear. In this study, we evaluated the role of PGD2 in joint inflammation using genetically modified mice. Injection of complete Freund's adjuvant (CFA) increased the production of PGD2 and induced paw swelling and cartilage erosion in wild-type (WT) mice. These phenomena were accompanied with an increase in the mRNA levels of TNF-α, IL-6, IL-1ß, and matrix-degrading metalloproteinase-9. Knockdown of hematopoietic PGD synthase (H-PGDS) abolished the PGD2 production and exacerbated all of the arthritic manifestations in the inflamed paw. Immunostaining revealed that infiltrating macrophages strongly expressed H-PGDS in the CFA-injected paw. Morphologic studies revealed vascular hyperpermeability and angiogenesis in the inflamed WT paw. H-PGDS deficiency was accelerated, whereas daily administration of a PGD2 receptor D prostanoid (DP) agonist attenuated the CFA-induced hyperpermeability and angiogenesis. We further confirmed that DP deficiency exacerbated, whereas the administration of the DP agonist improved, the CFA-induced arthritic manifestations. The findings demonstrate that H-PGDS-derived PGD2 ameliorates joint inflammation by attenuating vascular permeability and subsequent angiogenesis and indicates the therapeutic potential of a DP agonist for arthritis.-Tsubosaka, Y., Maehara, T., Imai, D., Nakamura, T., Kobayashi, K., Nagata, N., Fujii, W., Murata, T. Hematopoietic prostaglandin D synthase-derived prostaglandin D2 ameliorates adjuvant-induced joint inflammation in mice.


Subject(s)
Arthritis, Experimental/prevention & control , Inflammation/prevention & control , Intramolecular Oxidoreductases/physiology , Joint Diseases/prevention & control , Neovascularization, Pathologic/prevention & control , Prostaglandin D2/pharmacology , Adjuvants, Immunologic/toxicity , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Capillary Permeability , Collagen/toxicity , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Joint Diseases/chemically induced , Joint Diseases/metabolism , Joint Diseases/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Pathologic/chemically induced , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology
9.
Anesthesiology ; 132(4): 867-880, 2020 04.
Article in English | MEDLINE | ID: mdl-32011337

ABSTRACT

BACKGROUND: Recent emerging evidence suggests that extra-adrenal synthesis of aldosterone occurs (e.g., within the failing heart and in certain brain areas). In this study, the authors investigated evidence for a local endogenous aldosterone production through its key processing enzyme aldosterone synthase within peripheral nociceptive neurons. METHODS: In male Wistar rats (n = 5 to 8 per group) with Freund's complete adjuvant hind paw inflammation, the authors examined aldosterone, aldosterone synthase, and mineralocorticoid receptor expression in peripheral sensory neurons using quantitative reverse transcriptase-polymerase chain reaction, Western blot, immunohistochemistry, and immunoprecipitation. Moreover, the authors explored the nociceptive behavioral changes after selective mineralocorticoid receptor antagonist, canrenoate-K, or specific aldosterone synthase inhibitor application. RESULTS: In rats with Freund's complete adjuvant-induced hind paw inflammation subcutaneous and intrathecal application of mineralocorticoid receptor antagonist, canrenoate-K, rapidly and dose-dependently attenuated nociceptive behavior (94 and 48% reduction in mean paw pressure thresholds, respectively), suggesting a tonic activation of neuronal mineralocorticoid receptors by an endogenous ligand. Indeed, aldosterone immunoreactivity was abundant in peptidergic nociceptive neurons of dorsal root ganglia and colocalized predominantly with its processing enzyme aldosterone synthase and mineralocorticoid receptors. Moreover, aldosterone and its synthesizing enzyme were significantly upregulated in peripheral sensory neurons under inflammatory conditions. The membrane mineralocorticoid receptor consistently coimmunoprecipitated with endogenous aldosterone, confirming a functional link between mineralocorticoid receptors and its endogenous ligand. Importantly, inhibition of endogenous aldosterone production in peripheral sensory neurons by a specific aldosterone synthase inhibitor attenuated nociceptive behavior after hind paw inflammation (a 32% reduction in paw pressure thresholds; inflammation, 47 ± 2 [mean ± SD] vs. inflammation + aldosterone synthase inhibitor, 62 ± 2). CONCLUSIONS: Local production of aldosterone by its processing enzyme aldosterone synthase within peripheral sensory neurons contributes to ongoing mechanical hypersensitivity during local inflammation via intrinsic activation of neuronal mineralocorticoid receptors.


Subject(s)
Cytochrome P-450 CYP11B2/biosynthesis , Hyperalgesia/metabolism , Pain Measurement/methods , Sensory Receptor Cells/metabolism , Adjuvants, Immunologic/toxicity , Aldosterone/biosynthesis , Animals , Freund's Adjuvant/toxicity , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Male , Mineralocorticoid Receptor Antagonists/pharmacology , Mineralocorticoid Receptor Antagonists/therapeutic use , Pain Measurement/drug effects , Physical Stimulation/adverse effects , Rats , Rats, Wistar , Sensory Receptor Cells/drug effects
10.
Org Biomol Chem ; 18(3): 425-430, 2020 01 22.
Article in English | MEDLINE | ID: mdl-31774425

ABSTRACT

Herein, we report on the synthesis of a series of enantiomerically pure linear, iso-branched, and α-branched monoacyl glycerides (MAGs) in 63-72% overall yield. The ability of the MAGs to signal through human macrophage inducible C-type lectin (hMincle) using NFAT-GFP reporter cells was explored, as was the ability of the compounds to activate human monocytes. From these studies, MAGs with an acyl chain length ≥C22 were required for Mincle activation and the production of interleukin-8 (IL-8) by human monocytes. Moreover, the iso-branched MAGs led to a more pronounced immune response compared to linear MAGs, while an α-branched MAG containing a C-32 acyl chain activated cells to a higher degree than trehalose dibehenate (TDB), the prototypical Mincle agonist. Across the compound classes, the activity of the sn-1 substituted isomers was greater than the sn-3 counterparts. None of the representative compounds were cytotoxic, thus mitigating cytotoxicity as a potential mediator of cellular activity. Taken together, 6h (sn-1, iC26+1), 8a (sn-1, C32) and 8b (sn-3, C32) exhibited the best immunostimulatory properties and thus, have potential as vaccine adjuvants.


Subject(s)
Adjuvants, Immunologic/pharmacology , Lectins, C-Type/agonists , Monoglycerides/pharmacology , Receptors, Immunologic/agonists , Adjuvants, Immunologic/chemical synthesis , Adjuvants, Immunologic/toxicity , Cell Line, Tumor , Humans , Molecular Structure , Monoglycerides/chemical synthesis , Monoglycerides/toxicity , Stereoisomerism , Structure-Activity Relationship
11.
J Immunol ; 201(1): 167-182, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29794016

ABSTRACT

IL-36α (gene symbol Il1f6), a member of the IL-36 family, is closely associated with inflammatory diseases, including colitis and psoriasis. In this study, we found that Il1f6-/- mice developed milder psoriasiform dermatitis upon treatment with imiquimod, a ligand for TLR ligand 7 (TLR7) and TLR8, whereas Il1f6-/- mice showed similar susceptibility to dextran sodium sulfate-induced colitis to wild-type mice. These effects were observed in both cohoused and separately housed conditions, and antibiotic treatment did not cancel the resistance of Il1f6-/- mice to imiquimod-induced dermatitis. Bone marrow (BM) cell transfer revealed that IL-36α expression in skin-resident cells is important for the pathogenesis of dermatitis in these mice. Following stimulation with IL-36α, the expression of Il1f6 and Il1f9 (IL-36γ), but not Il1f8 (IL-36ß), was enhanced in murine BM-derived Langerhans cells (BMLCs) and murine primary keratinocytes but not in fibroblasts from mice. Upon stimulation with agonistic ligands of TLRs and C-type lectin receptors (CLRs), Il1f6 expression was induced in BMLCs and BM-derived dendritic cells. Furthermore, IL-36α stimulation resulted in significantly increased gene expression of psoriasis-associated Th17-related cytokines and chemokines such as IL-1α, IL-1ß, IL-23, CXCL1, and CXCL2 in BMLCs and fibroblasts, and IL-1α, IL-1ß, IL-17C, and CXCL2 in keratinocytes. Collectively, these results suggest that TLR/CLR signaling-induced IL-36α plays an important role for the development of psoriasiform dermatitis by enhancing Th17-related cytokine/chemokine production in skin-resident cells via a local autoamplification loop.


Subject(s)
Adjuvants, Immunologic/toxicity , Chemokines/biosynthesis , Colitis/pathology , Imiquimod/toxicity , Interleukin-1/metabolism , Keratinocytes/metabolism , Psoriasis/pathology , Skin/pathology , Th17 Cells/immunology , Animals , Bone Marrow Cells/cytology , Bone Marrow Transplantation , Cells, Cultured , Colitis/chemically induced , Dendritic Cells/metabolism , Dextran Sulfate/toxicity , Fibroblasts/metabolism , Interleukin-1/genetics , Langerhans Cells/metabolism , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Psoriasis/drug therapy , Psoriasis/genetics , Skin/cytology , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 8/metabolism
12.
Angew Chem Int Ed Engl ; 59(44): 19610-19617, 2020 10 26.
Article in English | MEDLINE | ID: mdl-32876984

ABSTRACT

Aluminum-containing adjuvants used in vaccine formulations suffer from low cellular immunity, severe aggregation, and accumulation in the brain. Conventional aluminosilicates widely used in the chemical industry focus mainly on acidic sites for catalytic applications, but they are rarely used as adjuvants. Reported here is an innovative "ligand-assisted steric hindrance" strategy to create a high density of six-coordinate VI Al-OH groups with basicity on dendritic mesoporous silica nanoparticles as new nanoadjuvants. Compared to four-coordinate IV Al-modified counterparts, VI Al-OH-rich aluminosilicate nanoadjuvants enhance cellular delivery of antigens and provoke stronger cellular immunity. Moreover, the aluminum accumulation in the brain is more reduced than that with a commercial adjuvant. These results show that coordination chemistry can be used to control the adjuvanticity, providing new understanding in the development of next-generation vaccine adjuvants.


Subject(s)
Adjuvants, Immunologic/pharmacology , Aluminum Silicates/pharmacology , Coordination Complexes/pharmacology , Nanoparticles/chemistry , Silicon Dioxide/pharmacology , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/toxicity , Aluminum/chemistry , Aluminum/pharmacology , Aluminum/toxicity , Aluminum Silicates/chemistry , Aluminum Silicates/toxicity , Animals , Antigens/immunology , B-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/drug effects , Coordination Complexes/chemistry , Coordination Complexes/toxicity , Female , Lymphocyte Activation/drug effects , Mice , Nanoparticles/toxicity , Ovalbumin/immunology , Porosity , RAW 264.7 Cells , Silicon Dioxide/chemistry , Silicon Dioxide/toxicity
13.
Fish Shellfish Immunol ; 85: 31-43, 2019 Feb.
Article in English | MEDLINE | ID: mdl-29510253

ABSTRACT

A key goal of a successful vaccine formulation is the strong induction of persistent protective immune responses without producing side-effects. Adjuvants have been proved to be successful in several species at inducing increased immune responses against poorly immunogenic antigens. Fish are not the exception and promising results of adjuvanted vaccine formulations in many species are needed. In this study, over a period of 300 days, we characterized the apparent damage and immune response in gilthead seabream immunized by intraperitoneal injection with the model antigen keyhole limpet hemocyanin (KLH) alone or formulated with Montanide ISA water-in-oil (761 or 763), or Imject™ aluminum hydroxide (aluminium), as adjuvants. Throughout the trial, external tissue damage was examined visually, but no change was observed. Internally, severe adhesions, increased fat tissue, and hepatomegaly were recorded, but, without impairing animal health. At 120 days post priming (dpp), histopathological evaluations of head-kidney, spleen and liver revealed the presence of altered melanomacrophage centers (MMC) in HK and spleen, but not in liver. Surprisingly, in all aluminium treated fish, classical stains unmasked a toxic effect on splenic-MMC, unequivocally characterized by a strong cell depletion. Furthermore, at 170 dpp transmission electron microscopy confirmed this data. Paradoxically, at the same time powerful immune responses were recorded in most vaccinated groups, including the aluminium treatment. Whatever the case, despite the observed adhesions and MMC depletion, fish physiology was not affected, and most side-effects were resolved after 300 dpp. Therefore, our data support adjuvant inclusion, but strongly suggest that use of aluminium must be further explored in detail before it might benefit the rational design of new vaccination strategies in aquaculture.


Subject(s)
Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/toxicity , Aluminum/pharmacology , Aluminum/toxicity , Macrophages/drug effects , Sea Bream/immunology , Animals , Hemocyanins/administration & dosage , Hemocyanins/immunology , Immunization/veterinary , Injections, Intraperitoneal/veterinary , Microscopy, Electron, Transmission/veterinary , Spleen/drug effects , Spleen/metabolism
14.
J Immunol ; 199(12): 3943-3951, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29093062

ABSTRACT

Understanding functions of Foxp3+ regulatory T cells (Tregs) during allergic airway inflammation remains incomplete. In this study, we report that, during cockroach Ag-induced allergic airway inflammation, Foxp3+ Tregs are rapidly mobilized into the inflamed lung tissues. However, the level of Treg accumulation in the lung was different depending on the type of inflammation. During eosinophilic airway inflammation, ∼30% of lung-infiltrating CD4 T cells express Foxp3, indicative of Tregs. On the contrary, only ∼10% of infiltrating CD4 T cells express Foxp3 during neutrophilic airway inflammation. Despite the different accumulation, the lung inflammation and inflammatory T cell responses were aggravated following Treg depletion, regardless of the type of inflammation, suggesting regulatory roles for Tregs. Interestingly, however, the extent to which inflammatory responses are aggravated by Treg depletion was significantly greater during eosinophilic airway inflammation. Indeed, lung-infiltrating Tregs exhibit phenotypic and functional features associated with potent suppression. Our results demonstrate that Tregs are essential regulators of inflammation, regardless of the type of inflammation, although the mechanisms used by Tregs to control inflammation may be shaped by environmental cues available to them.


Subject(s)
Alveolitis, Extrinsic Allergic/immunology , Lung/immunology , Neutrophil Infiltration/immunology , Pulmonary Eosinophilia/immunology , T-Lymphocytes, Regulatory/immunology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/toxicity , Allergens/administration & dosage , Allergens/immunology , Alum Compounds/administration & dosage , Alum Compounds/toxicity , Alveolitis, Extrinsic Allergic/etiology , Alveolitis, Extrinsic Allergic/pathology , Animals , Cockroaches/immunology , Disease Models, Animal , Forkhead Transcription Factors/analysis , Freund's Adjuvant/administration & dosage , Freund's Adjuvant/toxicity , Gene Expression Profiling , Genes, Reporter , Immunophenotyping , Inflammation , Insect Proteins/administration & dosage , Insect Proteins/immunology , Lung/pathology , Mice, Inbred C57BL , Pulmonary Eosinophilia/etiology , Pulmonary Eosinophilia/pathology , Specific Pathogen-Free Organisms
15.
Regul Toxicol Pharmacol ; 108: 104441, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31425728

ABSTRACT

PQ Birch represents an allergen-specific immunotherapy for the treatment of birch pollinosis. It consists of native birch pollen extract chemically modified with glutaldehyde adsorbed to L-tyrosine in its microcrystalline form with addition of the adjuvant Monophosphoryl Lipid A (MPL®). A nonclinical safety testing strategy was designed based upon interpretation of current legislation and regulatory intelligence and comprised genotoxicity studies (bacterial reverse mutation and Chinese hamster ovary micronucleus assays), a rat repeat dose toxicology study and a rabbit local tolerance study. No safety findings of concern were found. Thus, no evidence of genotoxicity was found. Relatively minor, immunostimulatory effects were seen following repeated subcutaneous dosing (once every 2 weeks for 13 weeks) as reversible increased white cell count (notably neutrophils), increased globulin level (resulting in decreased albumin/globulin [A/G] ratio) and increased fibrinogen, as well as minor dose site reaction in the form of inflammatory cell infiltrate. These findings are likely due to the immunostimulatory nature of MPL® and/or the presence of L-tyrosine within the adjuvanted vaccine. Similar dose site inflammatory changes to the injected formulation were also noted in the rabbit local tolerance study.


Subject(s)
Adjuvants, Immunologic/toxicity , Betula/immunology , Immunotherapy/adverse effects , Lipid A/analogs & derivatives , Pollen/immunology , Tyrosine/toxicity , Animals , CHO Cells , Cricetulus , Female , Lipid A/toxicity , Male , Mutagenicity Tests , Rabbits , Rats, Wistar , Rhinitis, Allergic, Seasonal/therapy , Salmonella typhimurium/drug effects , Salmonella typhimurium/growth & development , Skin/drug effects
16.
J Appl Toxicol ; 39(10): 1462-1469, 2019 10.
Article in English | MEDLINE | ID: mdl-31231859

ABSTRACT

PQ Grass represents an allergen-specific immunotherapy for pre-seasonal treatment of patients with seasonal allergic rhinitis (or rhinoconjunctivitis) with or without mild-to-moderate bronchial asthma. It consists of a native pollen extract for 13 grass species, chemically modified with glutaraldehyde, and adsorbed to l-tyrosine in a microcrystalline form with addition of the adjuvant Monophosphoryl Lipid A (MPL® ). Previous non-clinical safety testing, including rat repeat dose toxicity in adult and juvenile animals, rat reproductive toxicity and rabbit local tolerance studies showed no safety findings of concern. A new Good Laboratory Practice compliant rat subcutaneous repeat dose toxicity study to evaluate a higher clinical dose and modified posology (once every 2 weeks for 13 weeks) showed no signs of toxicity. As seen in previous studies, relatively minor, immunostimulatory effects were seen such as reversible increased white cell count (notably neutrophils), increased globulin level (resulting in decreased A/G ratio) and increased fibrinogen as well as minor dose site reaction in the form of inflammatory cell infiltrate. These findings are likely due to the immunostimulatory nature of MPL and/or the presence of l-tyrosine within the adjuvanted vaccine. This new toxicity study with PQ Grass therefore supports longer posology with higher dose levels.


Subject(s)
Adjuvants, Immunologic/toxicity , Adjuvants, Immunologic/therapeutic use , Hypersensitivity/drug therapy , Hypersensitivity/etiology , Immunotherapy/adverse effects , Immunotherapy/methods , Poaceae/adverse effects , Animals , Female , Humans , Male , Models, Animal , Rats, Wistar
17.
Immunopharmacol Immunotoxicol ; 41(1): 140-149, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30714433

ABSTRACT

Context: Influenza is a severe, life-threatening viral disease that can be prevented by vaccination. However, the anti-influenza human vaccine failed to show the required efficacy both in infants under 5 years old and in the elder population, who are among those with the highest risk of developing severe complications after influenza infection. Therefore, it is of high importance to improve the vaccine efficacy and ensure its safety in these susceptible populations. GK-1, a novel 18-aa peptide adjuvant, has been proved to increase the immunogenicity of the human influenza vaccine in both young and aged mice. Objective: A preclinical study of the toxicity profile of GK-1 following the World Health Organization guidelines to support its use was herein conducted. Material and methods: GK-1 was synthetically produced following Good Manufacturing Practices. The toxicological evaluation of GK-1 peptide was performed in rats after repeated dose-ranging trials by the subcutaneous route. The mutagenic potential of GK-1 was assessed by the micronucleus, chromosomal aberration, and Ames tests, in accordance with OECD Guidelines. Results: GK-1 did not show toxic effects at doses up to 12.5mg/kg, corresponding to 25 times the dose intended for human use. No indications of mutagenic potential were observed. GK-1 after dermal administration was well tolerated locally. Conclusion: The efficacy of GK-1 to improve influenza vaccine protection, along with the absence of toxicity and mutagenicity, as reported herein, support the evaluation of this peptide in a clinical trial as a novel adjuvant for human use.


Subject(s)
Adjuvants, Immunologic/toxicity , Chromosome Aberrations/drug effects , DNA Damage , Influenza Vaccines/immunology , Peptides, Cyclic/toxicity , Animals , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Female , Humans , Influenza, Human/prevention & control , Injections, Subcutaneous , Male , Mutagenicity Tests , Peptides, Cyclic/immunology , Rats, Wistar , Salmonella typhimurium/drug effects , Salmonella typhimurium/genetics , Toxicity Tests, Chronic
18.
Int J Toxicol ; 38(3): 163-172, 2019.
Article in English | MEDLINE | ID: mdl-31179828

ABSTRACT

A recombinant protective antigen (rPA) anthrax vaccine candidate (rPA7909) was developed as a next-generation vaccine indicated for postexposure prophylaxis of disease resulting from suspected or confirmed Bacillus anthracis exposure. The lyophilized form of rPA7909-vaccinated candidate contains 75 µg purified rPA, 750 µg aluminum (as Alhydrogel adjuvant), and 250 µg of an immunostimulatory Toll-like receptor 9 agonist oligodeoxynucleotide CpG 7909 in a 0.5 mL phosphate-buffered suspension. General toxicity and local reactogenicity were evaluated in Sprague Dawley rats vaccinated with the full human dose of rPA7909 by intramuscular injection. Animals were immunized on study days 1, 15, and 29. Control groups were administered diluent only or adjuvant control (excipients, CpG 7909, and Alhydrogel adjuvant in diluent) intramuscularly at the same dose volume and according to the same schedule used for rPA7909. Toxicity was assessed based on the results of clinical observations, physical examinations, body weights, injection site reactogenicity, ophthalmology, clinical pathology (hematology, coagulation, and serum chemistry), organ weights, and macroscopic and microscopic pathology evaluation. The immune response to rPA7909 vaccination was confirmed by measuring serum anti-PA immunoglobulin G levels. The rPA7909 vaccine produced no apparent systemic toxicity and only transient reactogenicity at the injection site. The injection site reaction from animals receiving the adjuvant control was very similar to those receiving rPA7909 with respect to the inflammation. The inflammatory response observed in the injection site and the draining lymph nodes was consistent with expected immune stimulation. The overall results indicated a favorable safety profile for rPA7909.


Subject(s)
Adjuvants, Immunologic/toxicity , Anthrax Vaccines/toxicity , Antigens, Bacterial/toxicity , Bacterial Toxins/toxicity , Oligodeoxyribonucleotides/toxicity , Animals , Antigens, Bacterial/immunology , Bacterial Toxins/immunology , Female , Freeze Drying , Immunoglobulin G/blood , Male , Rats, Sprague-Dawley , Recombinant Proteins/toxicity
19.
Int J Mol Sci ; 20(23)2019 Nov 25.
Article in English | MEDLINE | ID: mdl-31775224

ABSTRACT

The therapeutic application of antimicrobial peptides (AMPs), a potential type of peptide-based biomaterial, is impeded by their poor antimicrobial activity and potential cytotoxicity as a lack of understanding of their structure-activity relationships. In order to comprehensively enhance the antibacterial and clinical application potency of AMPs, a rational approach was applied to design amphiphilic peptides, including head-to-tail cyclic, linear and D-proline antimicrobial peptides using the template (IR)nP(IR)nP (n = 1, 2 and 3). Results showed that these amphiphilic peptides demonstrated antimicrobial activity in a size-dependent manner and that cyclic peptide OIR3, which contained three repeating units (IR)3, had greater antimicrobial potency and cell selectivity than liner peptide IR3, DIR3 with D-Pro and gramicidin S (GS). Surface plasmon resonance and endotoxin neutralization assays indicated that OIR3 had significant endotoxin neutralization capabilities, which suggested that the effects of OIR3 were mediated by binding to lipopolysaccharides (LPS). Using fluorescence spectrometry and electron microscopy, we found that OIR3 strongly promoted membrane disruption and thereby induced cell lysis. In addition, an LPS-induced inflammation assay showed that OIR3 inhibited the pro-inflammatory factor TNF-α in RAW264.7 cells. OIR3 was able to reduce oxazolone-induced skin inflammation in allergic dermatitis mouse model via the inhibition of TNF-α, IL-1ß and IL-6 mRNA expression. Collectively, the engineered head-to-tail cyclic peptide OIR3 was considerable potential candidate for use as a clinical therapeutic for the treatment of bacterial infections and skin inflammation.


Subject(s)
Anti-Bacterial Agents/pharmacology , Anti-Inflammatory Agents/pharmacology , Biocompatible Materials/pharmacology , Dermatitis/drug therapy , Peptides, Cyclic/pharmacology , Adjuvants, Immunologic/toxicity , Animals , Anti-Bacterial Agents/chemistry , Anti-Inflammatory Agents/chemistry , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/pharmacology , Biocompatible Materials/chemistry , Cell Death , Dermatitis/etiology , Dermatitis/pathology , Endotoxins/pharmacology , Hemolysis/drug effects , Humans , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred ICR , Oxazolone/toxicity , Peptides/chemistry , Peptides/pharmacology , Peptides, Cyclic/chemistry , RAW 264.7 Cells
20.
Pharm Dev Technol ; 24(1): 12-23, 2019 Jan.
Article in English | MEDLINE | ID: mdl-29172854

ABSTRACT

Catabolic conditions like acquired immunodeficiency syndrome, cancer, and burn can cause immunosuppression. Amino acids such as alanine and glutamine are essential for the activity of the immune system. Propolis is immunostimulant and the waste of propolis extraction has been reused with technological and therapeutic purposes. Therefore, this study describes the association of propolis byproduct extract (BPE) with pectin to prepare spray-dried microparticles containing the dipeptide l-alanyl-l-glutamine as stimulant systems of neutrophils. The use of a factorial design allowed selecting the best formulation, which was characterized by morphology, size, and entrapment efficiency analyses. In addition, the systems were characterized by thermal and X-ray diffraction analysis, Fourier-transform infrared spectroscopy, in vitro drug release, and in vitro cytotoxicity and stimulation test of neutrophils. Small well-structured microparticles with good entrapment efficiency values were achieved. Thermal stability of formulation was observed, and it was proved that pectin, BPE and l-alanyl-l-glutamine were dispersed throughout the matrix. The drug was released from the microparticles during 24 h governed by swelling and diffusion. The drug-loaded formulations showed a significant stimulating effect on neutrophils. These structures could increase the activity of immune cells, and other in vitro and in vivo studies should be performed in the future.


Subject(s)
Dipeptides/administration & dosage , Neutrophils/drug effects , Pectins/chemistry , Propolis/chemistry , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/toxicity , Chemistry, Pharmaceutical/methods , Dipeptides/pharmacology , Dipeptides/toxicity , Drug Carriers/chemistry , Drug Liberation , Humans , In Vitro Techniques , Microspheres , Neutrophils/metabolism , Spectroscopy, Fourier Transform Infrared , Time Factors , X-Ray Diffraction
SELECTION OF CITATIONS
SEARCH DETAIL