Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 677
Filter
1.
Int J Nanomedicine ; 19: 4181-4197, 2024.
Article in English | MEDLINE | ID: mdl-38766656

ABSTRACT

Purpose: The committed differentiation fate regulation has been a difficult problem in the fields of stem cell research, evidence showed that nanomaterials could promote the differentiation of stem cells into specific cell types. Layered double hydroxide (LDH) nanoparticles possess the regulation function of stem cell fate, while the underlying mechanism needs to be investigated. In this study, the process of embryonic stem cells (ESCs) differentiate to neural progenitor cells (NPCs) by magnesium aluminum LDH (MgAl-LDH) was investigated. Methods: MgAl-LDH with diameters of 30, 50, and 100 nm were synthesized and characterized, and their effects on the cytotoxicity and differentiation of NPCs were detected in vitro. Dot blot and MeRIP-qPCR were performed to detect the level of m6A RNA methylation in nanoparticles-treated cells. Results: Our work displayed that LDH nanoparticles of three different sizes were biocompatible with NPCs, and the addition of MgAl-LDH could significantly promote the process of ESCs differentiate to NPCs. 100 nm LDH has a stronger effect on promoting NPCs differentiation compared to 30 nm and 50 nm LDH. In addition, dot blot results indicated that the enhanced NPCs differentiation by MgAl-LDH was closely related to m6A RNA methylation process, and the major modification enzyme in LDH controlled NPCs differentiation may be the m6A RNA methyltransferase METTL3. The upregulated METTL3 by LDH increased the m6A level of Sox1 mRNA, enhancing its stability. Conclusion: This work reveals that MgAl-LDH nanoparticles can regulate the differentiation of ESCs into NPCs by increasing m6A RNA methylation modification of Sox1.


Subject(s)
Cell Differentiation , Nanoparticles , Neural Stem Cells , Cell Differentiation/drug effects , Animals , Neural Stem Cells/drug effects , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Mice , Nanoparticles/chemistry , Methylation/drug effects , Hydroxides/chemistry , Hydroxides/pharmacology , Methyltransferases/metabolism , Methyltransferases/genetics , Particle Size , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/cytology , Adenosine/pharmacology , Adenosine/chemistry , Adenosine/analogs & derivatives , Aluminum Hydroxide/chemistry , Aluminum Hydroxide/pharmacology , Magnesium Hydroxide/chemistry , Magnesium Hydroxide/pharmacology
2.
Biomaterials ; 308: 122569, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38626556

ABSTRACT

In subunit vaccines, aluminum salts (Alum) are commonly used as adjuvants, but with limited cellular immune responses. To overcome this limitation, CpG oligodeoxynucleotides (ODNs) have been used in combination with Alum. However, current combined usage of Alum and CpG is limited to linear mixtures, and the underlying interaction mechanism between CpG and Alum is not well understood. Thus, we propose to chemically conjugate Alum nanoparticles and CpG (with 5' or 3' end exposed) to design combination adjuvants. Our study demonstrates that compared to the 3'-end exposure, the 5'-end exposure of CpG in combination adjuvants (Al-CpG-5') enhances the activation of bone-marrow derived dendritic cells (BMDCs) and promotes Th1 and Th2 cytokine secretion. We used the SARS-CoV-2 receptor binding domain (RBD) and hepatitis B surface antigen (HBsAg) as model antigens to demonstrate that Al-CpG-5' enhanced antigen-specific antibody production and upregulated cytotoxic T lymphocyte markers. Additionally, Al-CpG-5' allows for coordinated adaptive immune responses even at lower doses of both CpG ODNs and HBsAg antigens, and enhances lymph node transport of antigens and activation of dendritic cells, promoting Tfh cell differentiation and B cell activation. Our novel Alum-CPG strategy points the way towards broadening the use of nanoadjuvants for both prophylactic and therapeutic vaccines.


Subject(s)
Adjuvants, Immunologic , Aluminum Hydroxide , Aluminum Oxide , Dendritic Cells , Hepatitis B Surface Antigens , Nanoparticles , Oligodeoxyribonucleotides , Adjuvants, Immunologic/pharmacology , Animals , Nanoparticles/chemistry , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Oligodeoxyribonucleotides/chemistry , Oligodeoxyribonucleotides/pharmacology , Hepatitis B Surface Antigens/immunology , Hepatitis B Surface Antigens/metabolism , Aluminum Hydroxide/chemistry , Aluminum Hydroxide/pharmacology , Mice , Mice, Inbred C57BL , Female , Cytokines/metabolism , Alum Compounds/chemistry , Alum Compounds/pharmacology
3.
JCI Insight ; 8(23)2023 Dec 08.
Article in English | MEDLINE | ID: mdl-38063196

ABSTRACT

IL-12 is a potent cytokine that can promote innate and adaptive anticancer immunity, but its clinical development has been limited by toxicity when delivered systemically. Intratumoral (i.t.) administration can expand the therapeutic window of IL-12 and other cytokines but is in turn limited by rapid drug clearance from the tumor, which reduces efficacy, necessitates frequent administration, and increases systemic accumulation. To address these limitations, we developed an anchored IL-12 designated ANK-101, composed of an engineered IL-12 variant that forms a stable complex with the FDA-approved vaccine adjuvant aluminum hydroxide (Alhydrogel). Following i.t. administration of murine ANK-101 (mANK-101) in early intervention syngeneic mouse tumors, the complex formed a depot that was locally retained for weeks as measured by IVIS or SPECT/CT imaging, while unanchored protein injected i.t. was cleared within hours. One or 2 i.t. injections of mANK-101 induced single-agent antitumor activity across a diverse range of syngeneic tumors, including models resistant to checkpoint blockade at doses where unanchored IL-12 had no efficacy. Local treatment with mANK-101 further induced regressions of noninjected lesions, especially when combined with systemic checkpoint blockade. Antitumor activity was associated with remodeling of the tumor microenvironment, including prolonged IFN-γ and chemokine expression, recruitment and activation of T and NK cells, M1 myeloid cell skewing, and increased antigen processing and presentation. Subcutaneous administration of ANK-101 in cynomolgus macaques was well tolerated. Together, these data demonstrate that ANK-101 has an enhanced efficacy and safety profile and warrants future clinical development.


Subject(s)
Interleukin-12 , Neoplasms , Mice , Animals , Aluminum Hydroxide/pharmacology , Tumor Microenvironment , Cytokines
4.
Biomed Res Int ; 2023: 4499407, 2023.
Article in English | MEDLINE | ID: mdl-37854793

ABSTRACT

The present study is aimed at investigating the long-term effects of the aluminum hydroxide administration in the small intestine, lung, liver, and kidney of male BALB/c mice. The mice received via orogastric gavage phosphate buffered or 10 mg/kg aluminum hydroxide 3 times a week for 6 months. Administration of aluminum hydroxide decreased hemoglobin, hematocrit, and erythrocyte. In the blood, kidney and liver function markers were evaluated, and long-term administration of aluminum hydroxide led to an increase in AST levels and a decrease in urea levels. The animals exposed to aluminum showed higher lipid and protein oxidation in all the organs analyzed. In relation to the enzymes involved in antioxidant defense, the lungs showed lower superoxide dismutase (SOD) and catalase activity and a lower reduced and oxidized glutathione (GSH/GSSG) ratio. In the liver, aluminum administration led to a decrease in catalase activity and the GSH/GSSG ratio. Lower catalase activity was observed in the small intestine, as well as in the lungs and liver. In addition to alterations in antioxidant defense, increased levels of the chemokine CCL-2 were observed in the lungs, lower levels of IL-10 in the liver and small intestine, and decreased levels of IL-6 in the intestine of the animals that received aluminum hydroxide for 6 months. Long-term exposure to aluminum promoted steatosis in the liver. In the kidneys, mice treated with aluminum presented a decreased glomerular density than in the naive control group. In the small intestine, exposure caused villi shortening. Our results indicate that long-term oral administration of aluminum hydroxide provokes systemic histological damage, inflammation, and redox imbalance.


Subject(s)
Antioxidants , Glutathione , Mice , Male , Animals , Antioxidants/pharmacology , Glutathione Disulfide/metabolism , Glutathione/metabolism , Catalase/metabolism , Aluminum Hydroxide/pharmacology , Mice, Inbred BALB C , Aluminum/pharmacology , Oxidation-Reduction , Superoxide Dismutase/metabolism , Liver/metabolism , Inflammation/chemically induced , Inflammation/metabolism , Oxidative Stress
5.
Antimicrob Agents Chemother ; 67(4): e0149522, 2023 04 18.
Article in English | MEDLINE | ID: mdl-36943038

ABSTRACT

Tebipenem pivoxil hydrobromide (TBP-PI-HBr) is a novel oral carbapenem prodrug being developed for the treatment of serious bacterial infections. This open-label, 3-period, fixed sequence study evaluated the effect of gastric acid-reducing agents, aluminum hydroxide/magnesium hydroxide/simethicone, and omeprazole on the pharmacokinetics (PK) of tebipenem (TBP), the active moiety, following coadministration with immediate release TBP-PI-HBr during fasting. In Period 1, subjects received a single oral dose of TBP-PI-HBr 600 mg (2 × 300 mg tablets). In Period 2, subjects received a single oral dose of aluminum hydroxide 800 mg/magnesium hydroxide 800 mg/simethicone 80 mg suspension co-administered with a single dose of TBP-PI-HBr 600 mg. In Period 3, subjects received a single oral dose of omeprazole 40 mg once daily over 5 days, followed by single dose administration of TBP-PI-HBr 600 mg on day 5. In each period, whole blood samples were obtained prior to, and up to 24 h, following TBP-PI-HBr dose administration in order to characterize TBP PK. A 7-day washout was required between periods. Twenty subjects were enrolled and completed the study. Following co-administration of TBP-PI-HBr with either aluminum hydroxide/magnesium hydroxide/simethicone or omeprazole, total TBP exposure (area under the curve [AUC]) was approximately 11% (geometric mean ratio 89.2, 90% confidence interval: 83,2, 95.7) lower, and Cmax was 22% (geometric mean ratio 78.4, 90% confidence interval: 67.9, 90.6) and 43% (geometric mean ratio 56.9, 90% confidence interval: 49.2, 65.8) lower, respectively, compared to administration of TBP-PI-HBr alone. Mean TBP elimination half-life (t1/2) was generally comparable across treatments (range: 1.0 to 1.5 h). Concomitant administration of TBP-PI-HBr with omeprazole or aluminum hydroxide/magnesium hydroxide/simethicone is not expected to impact the efficacy of TBP-PI-HBr, as there is minimal impact on TBP plasma AUC, which is the pharmacodynamic driver of efficacy. Co-administration was generally safe and well tolerated.


Subject(s)
Antacids , Anti-Ulcer Agents , Adult , Humans , Administration, Oral , Aluminum Hydroxide/pharmacology , Antacids/pharmacology , Cross-Over Studies , Drug Interactions , Magnesium Hydroxide/pharmacology , Omeprazole/pharmacology , Proton Pump Inhibitors/pharmacology , Simethicone
6.
Sci Rep ; 13(1): 3198, 2023 02 23.
Article in English | MEDLINE | ID: mdl-36823452

ABSTRACT

Aluminum compounds are the most widely used adjuvants in veterinary and human vaccines. Despite almost a century of use and substantial advances made in recent decades about their fate and biological effects, the exact mechanism of their action has been continuously debated, from the initial "depot-theory" to the direct immune system stimulation, and remains elusive. Here we investigated the early in vitro response of primary human PBMCs obtained from healthy individuals to aluminum oxyhydroxide (the most commonly used adjuvant) and a whole vaccine, in terms of internalization, conventional and non-conventional autophagy pathways, inflammation, ROS production, and mitochondrial metabolism. During the first four hours of contact, aluminum oxyhydroxide particles, with or without adsorbed vaccine antigen, (1) were quickly recognized and internalized by immune cells; (2) increased and balanced two cellular clearance mechanisms, i.e. canonical autophagy and LC3-associated phagocytosis; (3) induced an inflammatory response with TNF-α production as an early event; (4) and altered mitochondrial metabolism as assessed by both decreased maximal oxygen consumption and reduced mitochondrial reserve, thus potentially limiting further adaptation to other energetic requests. Further studies should consider a multisystemic approach of the cellular adjuvant mechanism involving interconnections between clearance mechanism, inflammatory response and mitochondrial respiration.


Subject(s)
Aluminum , Vaccines , Humans , Aluminum Hydroxide/pharmacology , Adjuvants, Immunologic/pharmacology , Macrophages
7.
Brain Res ; 1803: 148241, 2023 03 15.
Article in English | MEDLINE | ID: mdl-36639094

ABSTRACT

The current study was designed to assess the possible neuroprotective effect of borax (BX) against the toxicity of aluminum hydroxide [AH, Al (OH)3] on brain of rainbow trout (Oncorhynchus mykiss) with multibiomarker approaches. For this purpose, the presence of the neuroprotective action by BX against the AH exposure was assessed by the activities of catalase (CAT), glutathione peroxidase (GPx), superoxide dismutase (SOD), myeloperoxidase (MPO), acetylcholinesterase (AChE). In addition, we evaluated glutathione (GSH), malondialdehyde (MDA), DNA damage (8-OHdG), apoptosis (caspase 3), tumor necrosis factor alpha (TNF-α), interleukin 6 (IL-6), nuclear factor erythroid-2 (Nrf-2), and brain-derived neurotrophic factor (BDNF) levels in 96 h semi-static treatment. In the 48th and 96th hour samplings, apoptosis induced by AH in the Nrf-2/BDNF/AChE pathways in rainbow trout brain tissue was revealed by DNA damage, enzyme inhibitions and lipid peroxidations. On the contrary applications of BX supported antioxidant capacity without leading apoptosis, lipid peroxidation, inflammatory response and DNA damage. BX also increased the BDNF levels and AChE activity. Moreover, BX exerted a neuroprotective effect against AH-induced neurotoxicity via down-regulating cytokine-related pathways, minimising DNA damage, apoptosis as well as up-regulating GSH, AChE, BDNF and antioxidant enzyme levels. It can be concluded that the combination of borax with AH modulated the toxic effects of AH.


Subject(s)
Antioxidants , Neuroprotective Agents , Animals , Antioxidants/pharmacology , Antioxidants/metabolism , Acetylcholinesterase/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Aluminum Hydroxide/metabolism , Aluminum Hydroxide/pharmacology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/metabolism , Superoxide Dismutase/metabolism , Brain/metabolism , Oxidative Stress , Glutathione/metabolism
8.
J Ethnopharmacol ; 302(Pt A): 115913, 2023 Feb 10.
Article in English | MEDLINE | ID: mdl-36347302

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Jingfang Granule (JFG) is a Traditional Chinese Medicine prescription to empirically treat skin disease such as urticaria in clinical practice. However, the potential mechanisms of JFG on urticaria are not fully defined. AIM OF STUDY: The aim of this study is to investigate the mechanisms of JFG in treating urticaria through an OVA/aluminum hydroxide induced urticaria mice model. MATERIALS AND METHODS: KM mice were injected intraperitoneally (i.p.) with OVA/aluminium hydroxide to establish the model with urticaria. After the mice were administered JFG, itching degree and hematoxylin and eosin (H&E) staining were used to assess the protective effect of JFG on mice with urticaria. The regulatory networks were investigated by proteomics and central carbon metabolomics. Spleen T lymphocyte subsets were detected by flow cytometry. Peripheral blood cytokines were detected using ELISA kits or Cytometric Bead Array (CBA) kits. The protein expression of skin tissue was detected by western blot or immunohistochemical staining. RESULTS: JFG significantly relived skin tissue lesions and skin pruritus in mice with urticaria. Meanwhile, JFG significantly decreased IgE, IL-1ß, IL-6, IL-4, TNF-α and IL-17A levels and increased IFN-γ levels in the serum of urticaria mice by inhibiting the expression of inflammation associated proteins including TLR4 and p-NF-κB p65, p-ERK1/2, p-JNK and p-p38, NLRP3, ASC and cleaved caspase-1. The results of proteomics, central carbon metabolomics, western blot and immunohistochemical staining confirmed that JFG inhibited Glycolysis/Gluconeogenesis and Pentose phosphate pathway in the skin tissue of urticaria mice by activating the LKB1/AMPK/SIRT1 axis and then downregulating the protein expressions of Glut1, TORC2, p-CREB, PEPCK, HNF4α and G6Pase. CONCLUSION: The current study demonstrates that JFG is effective in treating OVA/aluminum hydroxide-induced skin lesions and inflammation in mice, and JFG exhibits the clinical benefits via modulating LKB1/AMPK/SIRT1 axis, which in turn inhibits Glycolysis/Gluconeogenesis and Pentose phosphate pathway.


Subject(s)
Sirtuin 1 , Urticaria , Animals , Mice , Sirtuin 1/metabolism , AMP-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Signal Transduction , Aluminum Hydroxide/pharmacology , Inflammation/drug therapy , Carbon , Glucose/pharmacology
9.
Zhongguo Zhong Yao Za Zhi ; 47(20): 5467-5472, 2022 Oct.
Article in Chinese | MEDLINE | ID: mdl-36471961

ABSTRACT

This study explored the curative effect of Jingfang Mixture on urticaria mice induced by aluminum hydroxide/ovalbumin, and discussed its mechanism. Sixty male Kunming mice were randomly divided into a normal group, a model group, three Jingfang Mixture(low-dose, medium-dose, and high-dose) groups, and a positive drug(cetirizine hydrochloride) group. The urticarial model in mice was induced by the intraperitoneal injection of the mixed solution of ovalbumin and aluminum hydroxide. The degrees of pruritus were observed after the second immunization. Pathological changes were detected by hematoxylin and eosin(HE) staining. Levels of interleukin 1ß(IL-1ß) and tumor necrosis factor α(TNF-α) in the serum were detected by enzyme linked immunosorbent assay(ELISA). Expressions of NOD-like receptor protein 3(NLRP3) and IL-1ß were detected by immunohistochemistry(IHC). Expressions of nuclear factor kappa-B(NF-κB p65), NLRP3, apoptosis-associated speck-like protein containing a CARD(ASC), cysteinyl aspartate-specific proteases 1(caspase-1), and IL-1ß proteins were detected by Western blot. The results showed that, except for the normal group, the mice in all groups had different degrees of pruritus. Compared with the model group, the Jingfang Mixture groups and the positive drug group prolonged the scratching latency of mice(P<0.05), and significantly reduced the number of scratching(P<0.05). In addition, the Jingfang Mixture groups and the positive drug group improved the pathological morphology of skin tissue. The expression levels of IL-1ß and TNF-α in serum were significantly reduced(P<0.05), and the number of NLRP3 and IL-1ß positive cells was decreased(P<0.01). The expressions of p-NF-κB p65, NLRP3, ASC, cleaved caspase-1, and IL-1ß protein were significantly down-regulated(P<0.05). The results of the above study indicate that Jingfang Mixture inhibit the inflammatory response in urticaria mice, and the mechanism may be related to the inhibition of activating NF-κB/NLRP3/IL-1ß signaling pathway.


Subject(s)
NF-kappa B , Urticaria , Animals , Male , Mice , NF-kappa B/genetics , NF-kappa B/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Tumor Necrosis Factor-alpha/metabolism , Ovalbumin , Aluminum Hydroxide/pharmacology , Signal Transduction , Caspase 1/genetics , Caspase 1/metabolism , Pruritus
10.
Vet Res Commun ; 46(4): 1097-1109, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35927371

ABSTRACT

Edwardsiella tarda is considered one of the important bacterial fish pathogens. The outer membrane proteins (OMPs) of E. tarda are structurally and functionally conserved, and immunogenic. This study assessed the effects of the OMPs of E. tarda CGH9 as a vaccine without aluminium hydroxide [AH] (T1) and with AH adjuvant (T2) on the respiratory burst (ROB) activity, lymphocyte proliferation of head kidney (HK) leukocytes, and serum antibody production in pangas catfish Pangasius pangasius. The ROB activity and lymphocyte proliferation of HK leukocytes increased in both vaccinated groups compared to the control. Nonetheless, the T2 group showed a gradual increase in ROB activity and lymphocyte proliferation of HK leukocytes up to 3-weeks post-vaccination (wpv). The serum antibody production in the T1 group decreased initially for up to 2-wpv and increased from 3-wpv; whereas, in the T2 group, the serum-specific antibody levels were significantly high from 1-wpv compared to control. Simultaneously, the protective efficacy in terms of relative percentage survival in the T2 group after injecting with a lethal dose of E. tarda CGH9 was high (89.00±15.56) compared to the T1 group (78.00±0.00). Furthermore, the catfish administered with a booster dose of E. tarda OMPs with or without AH adjuvant showed no additional increase in immune response or protective immunity. These results suggested that E. tarda OMPs and AH adjuvant complex has a higher potential to induce protective immunity, which may be a good choice as a vaccine to combat E. tarda infection in catfish.


Subject(s)
Catfishes , Enterobacteriaceae Infections , Fish Diseases , Animals , Edwardsiella tarda , Aluminum Hydroxide/pharmacology , Membrane Proteins , Bacterial Vaccines , Fish Diseases/microbiology , Enterobacteriaceae Infections/prevention & control , Enterobacteriaceae Infections/veterinary , Antibodies, Bacterial , Adjuvants, Immunologic/pharmacology , Immunity
11.
Vaccine ; 40(38): 5601-5607, 2022 09 09.
Article in English | MEDLINE | ID: mdl-35999078

ABSTRACT

Safety and potency assessment for batch release testing of established vaccines still relies partly on animal tests. An important avenue to move to batch release without animal testing is the consistency approach. This approach is based on thorough characterization of the vaccine to identify critical quality attributes that inform the use of a comprehensive set of non-animal tests to release the vaccine, together with the principle that the quality of subsequent batches follows from their consistent production. Many vaccine antigens are by themselves not able to induce a protective immune response. The antigens are therefore administered together with adjuvant, most often by adsorption to aluminium salts. Adjuvant function is an important component of vaccine potency, and an important quality attribute of the final product. Aluminium adjuvants are capable of inducing NLRP3 inflammasome activation. The aim of this study was to develop and evaluate an in vitro assay for NLRP3 inflammasome activation by aluminium-adjuvanted vaccines. We evaluated the effects of Diphtheria-Tetanus-acellular Pertussis combination vaccines from two manufacturers and their respective adjuvants, aluminium phosphate (AP) and aluminium hydroxide (AH), in an in vitro assay for NLRP3 inflammasome activation. All vaccines and adjuvants tested showed a dose-dependent increase in IL-1ß production and a concomitant decrease in cell viability, suggesting NLRP3 inflammasome activation. The results were analysed by benchmark dose modelling, showing a similar 50% effective dose (ED50) for the two vaccine batches and corresponding adjuvant of manufacturer A (AP), and a similar ED50 for the two vaccine batches and corresponding adjuvant of manufacturer B (AH). This suggests that NLRP3 inflammasome activation is determined by the adjuvant only. Repeated freeze-thaw cycles reduced the adjuvant biological activity of AH, but not AP. Inflammasome activation may be used to measure adjuvant biological activity as an important quality attribute for control or characterization of the adjuvant.


Subject(s)
Diphtheria-Tetanus-acellular Pertussis Vaccines , Diphtheria , Tetanus , Whooping Cough , Adjuvants, Immunologic/pharmacology , Aluminum , Aluminum Hydroxide/pharmacology , Antibodies, Bacterial , Cell Line , Diphtheria/prevention & control , Diphtheria-Tetanus-Pertussis Vaccine , Humans , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Pertussis Vaccine , Tetanus/prevention & control , Whooping Cough/prevention & control
12.
J Environ Sci Health B ; 57(5): 350-357, 2022.
Article in English | MEDLINE | ID: mdl-35380503

ABSTRACT

The presence of antibiotic-resistant bacteria (ARB) and antibiotic-resistance genes (ARGs) in swine wastewater may present a threat to the environment and public health. Conventional swine wastewater treatment processes generally fail to effectively reduce the content of ARGs. Therefore, it is necessary to develop a highly efficient and low-cost treatment method to solve this environmental problem. In doing so, we evaluated the application of three common coagulants in the treatment of swine wastewater. Using metagenomics, we evaluated the removal efficiency of ARG loads, as well as the effect of coagulation on the structure and diversity of swine wastewater, and on the bacterial community. The results showed that the three coagulants could effectively reduce the physicochemical pollution indexes of swine wastewater (e.g., TP, NTU, COD). After treatment, the loads of a variety of antibiotics in the swine wastewater were significantly reduced, with the exception of NFX and SMD, which were all close to 100%. At the same time, in evaluating the total number of microbial colonies and the total number of fecal Escherichia coli bacteria under the three conditions, Polyaluminum Chloride (PAC) ranked first among the three coagulants with 89.18%, 93.07%, 89.92%, 98.76%, 99.60%, and 98.68%. Metagenomic analysis revealed that the abundance of cfcC, tetX, mphE, msrE, tet36, and other ARGs in the water sample after the LST treatment was significantly lower than that of the original swine wastewater sample. These findings demonstrate the feasibility of using coagulants to treat swine wastewater, which is of great significance for improving water quality and reducing the potential impacts of ARGs.


Subject(s)
Wastewater , Water Purification , Aluminum Hydroxide/pharmacology , Angiotensin Receptor Antagonists , Angiotensin-Converting Enzyme Inhibitors , Animals , Anti-Bacterial Agents/pharmacology , Drug Resistance, Microbial/genetics , Genes, Bacterial , Iron , Sulfates , Swine , Water Purification/methods
13.
Mol Microbiol ; 117(5): 1173-1195, 2022 05.
Article in English | MEDLINE | ID: mdl-35344242

ABSTRACT

Aluminum salts have been successfully utilized as adjuvants to enhance the immunogenicity of vaccine antigens since the 1930s. However, the cellular mechanisms behind the immune adjuvanticity effect of these materials in antigen-presenting cells are poorly understood. In this study, we investigated the uptake and trafficking of aluminum oxy-hydroxide (AlOOH), in RAW 264.7 murine and U-937 human macrophages-like cells. Furthermore, we determined the impact that the adsorption to AlOOH particulates has on the trafficking of a Bordetella pertussis vaccine candidate, the genetically detoxified pertussis toxin (gdPT). Our results indicate that macrophages internalize AlOOH by constitutive macropinocytosis assisted by the filopodial protrusions that capture the adjuvant particles. Moreover, we show that AlOOH has the capacity to nonspecifically adsorb IgG, engaging opsonic phagocytosis, which is a feature that may allow for more effective capture and uptake of adjuvant particles by antigen-presenting cells (APCs) at the site of vaccine administration. We found that AlOOH traffics to endolysosomal compartments that hold degradative properties. Importantly, while we show that gdPT escapes degradative endolysosomes and traffics toward the retrograde pathway, as reported for the wild-type pertussis toxin, the adsorption to AlOOH diverts gdPT to traffic to the adjuvant's lysosome-type compartments, which may be key for MHC-II-driven antigen presentation and activation of CD4+ T cell. Thus, our findings establish a direct link between antigen adsorption to AlOOH and the intracellular trafficking of antigens within antigen-presenting cells and bring to light a new potential mechanism for aluminum adjuvancy. Moreover, the in-vitro single-cell approach described herein provides a general framework and tools for understanding critical attributes of other vaccine formulations.


Subject(s)
Aluminum Hydroxide , Aluminum , Adjuvants, Immunologic/pharmacology , Aluminum/pharmacology , Aluminum Hydroxide/pharmacology , Animals , Humans , Lysosomes , Macrophages , Mice , Pertussis Toxin/genetics , Pertussis Toxin/pharmacology , Pertussis Vaccine/pharmacology
14.
J Pharm Sci ; 111(4): 982-990, 2022 04.
Article in English | MEDLINE | ID: mdl-35090866

ABSTRACT

Aluminum hydroxide (Al(OH)3) and aluminum phosphate (AlPO4) are widely used adjuvants in human vaccines. However, a rationale to choose one or the other is lacking since the differences between molecular mechanisms of action of these adjuvants are unknown. In the current study, we compared the innate immune response induced by both adjuvants in vitro and in vivo. Proteome analysis of human primary monocytes was used to determine the immunological pathways activated by these adjuvants. Subsequently, analysis of immune cells present at the site of injection and proteome analysis of the muscle tissue revealed the differentially regulated processes related to the innate immune response in vivo. Incubation with Al(OH)3 specifically enhanced the activation of antigen processing and presentation pathways in vitro. In vivo experiments showed that only intramuscular (I.M.) immunization with Al(OH)3 attracted neutrophils, while I.M. immunization with AlPO4 attracted monocytes/macrophages to the site of injection. In addition, only I.M. immunization with Al(OH)3 enhanced the process of hemostasis after 96 hours, possibly related to neutrophilic extracellular trap formation. Both adjuvants differentially regulated various immune system-related processes. The results show that Al(OH)3 and AlPO4 act differently on the innate immune system. We speculate that these different regulations affect the interaction with cells, due to the different physicochemical properties of both adjuvants.


Subject(s)
Aluminum Hydroxide , Proteome , Adjuvants, Immunologic/pharmacology , Adjuvants, Pharmaceutic , Aluminum , Aluminum Compounds , Aluminum Hydroxide/pharmacology , Humans , Immunity, Innate , Phosphates
15.
Sci Rep ; 11(1): 15448, 2021 07 29.
Article in English | MEDLINE | ID: mdl-34326377

ABSTRACT

Herpes simplex virus is among the most prevalent sexually transmitted infections. Acyclovir is a potent, selective inhibitor of herpes viruses and it is indicated for the treatment and management of recurrent cold sores on the lips and face, genital herpes, among other diseases. The problem of the oral bioavailability of acyclovir is limited because of the low permeability across the gastrointestinal membrane. The use of nanoparticles of pseudoboehmite as a drug delivery system in vitro assays is a promising approach to further the permeability of acyclovir release. Here we report the synthesis of high purity pseudoboehmite from aluminium nitrate and ammonium hydroxide containing nanoparticles, using the sol-gel method, as a drug delivery system to improve the systemic bioavailability of acyclovir. The presence of pseudoboehmite nanoparticles were verified by infrared spectroscopy, transmission electron microscopy, and X-ray diffraction techniques. In vivo tests were performed with Wistar rats to compare the release of acyclovir, with and without the addition of pseudoboehmite. The administration of acyclovir with the addition of pseudoboehmite increased the drug content by 4.6 times in the plasma of Wistar rats after 4 h administration. We determined that the toxicity of pseudoboehmite is low up to 10 mg/mL, in gel and the dried pseudoboehmite nanoparticles.


Subject(s)
Acyclovir/administration & dosage , Aluminum Hydroxide/chemistry , Aluminum Oxide/chemistry , Antiviral Agents/administration & dosage , Drug Delivery Systems/methods , Nanogels/chemistry , Acyclovir/blood , Acyclovir/pharmacokinetics , Administration, Oral , Aluminum Hydroxide/pharmacology , Aluminum Oxide/pharmacology , Animals , Antiviral Agents/blood , Antiviral Agents/pharmacokinetics , Biological Availability , Caco-2 Cells , Cell Survival/drug effects , Drug Liberation , Herpes Simplex/drug therapy , Herpes Simplex/virology , Humans , Models, Animal , Rats , Rats, Wistar , Simplexvirus/drug effects
16.
Arch Razi Inst ; 76(5): 1213-1220, 2021 11.
Article in English | MEDLINE | ID: mdl-35355760

ABSTRACT

Newcastle disease (ND) and Avian influenza (AI) are the major problems and the most economically important viral diseases in the poultry industry; therefore, vaccination against these diseases is considered one of the most effective ways of prevention. Extensive studies have been conducted to improve the performance of vaccines, and one of the major achievements of these studies is the preparation of adjuvants as stimulants of the immune system and one of the most important compounds in killed vaccines. An immunogenicity comparison of three adjuvants including, ISA70VG, Nano-Aluminum Hydroxide (Nano-Alum), and MF59 alone or with Nano-Selenium (Nano-Se), was performed using bivalent Newcastle plus Avian Influenza (ND+AI) killed vaccine. In this study, 105 specific-pathogen-free chicks (Ross-308) were divided into 7 treatments, including T1 (control group), T2 (ISA70VG), T3 (ISA70VG plus Nano-Se), T4 (Nano-Alum Hydroxide), T5 (Nano-Alum+Nano-Se), T6 (MF59), and T7 (MF59+Nano-Se). The vaccine was injected subcutaneously on day 21 in the back of the neck area. The blood samples were taken on days 14, 21, 28, 35, 42, and 49 post-vaccination. Serums of the samples were titrated by the haemagglutination inhibition (HI) test against Newcastle and Avian influenza. Based on the results, the highest HI test titers were observed for the T2 and T3 treatments, while the T6 and T7 treatments had the lowest titers. Moreover, regardless of the type of the adjuvants, adding Nano-Se increased the antibody titer in the vaccinated groups. In conclusion, a combination of the ISA70VG adjuvant and Nano-Se induced excellent antibody titers using bivalent ND+AI killed vaccine.


Subject(s)
Influenza Vaccines , Influenza in Birds , Selenium , Aluminum Hydroxide/pharmacology , Animals , Chickens , Immunity, Humoral , Influenza in Birds/prevention & control , Newcastle disease virus , Selenium/pharmacology
17.
Front Immunol ; 12: 803647, 2021.
Article in English | MEDLINE | ID: mdl-35095889

ABSTRACT

The newly emerged severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is causing a spread of coronavirus disease 2019 (COVID-19) globally. In order to end the COVID-19 pandemic, an effective vaccine against SARS-CoV-2 must be produced at low cost and disseminated worldwide. The spike (S) protein of coronaviruses plays a pivotal role in the infection to host cells. Therefore, targeting the S protein is one of the most rational approaches in developing vaccines and therapeutic agents. In this study, we optimized the expression of secreted trimerized S protein of SARS-CoV-2 using a silkworm-baculovirus expression vector system and evaluated its immunogenicity in mice. The results showed that the S protein forming the trimeric structure was the most stable when the chicken cartilage matrix protein was used as the trimeric motif and could be purified in large amounts from the serum of silkworm larvae. The purified S protein efficiently induced antigen-specific antibodies in mouse serum without adjuvant, but its ability to induce neutralizing antibodies was low. After examining several adjuvants, the use of Alum adjuvant was the most effective in inducing strong neutralizing antibody induction. We also examined the adjuvant effect of paramylon from Euglena gracilis when administered with the S protein. Our results highlight the effectiveness and suitable construct design of the S protein produced in silkworms for the subunit vaccine development against SARS-CoV-2.


Subject(s)
Alum Compounds/pharmacology , Aluminum Hydroxide/pharmacology , Bombyx/genetics , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology , Adjuvants, Immunologic/pharmacology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , COVID-19/immunology , COVID-19 Vaccines/immunology , Cell Line , Chickens/genetics , Chickens/immunology , Chlorocebus aethiops , Euglena gracilis/immunology , Euglenozoa Infections/immunology , Female , Humans , Mice , Mice, Inbred BALB C , Pandemics/prevention & control , SARS-CoV-2/immunology , Vaccination/methods , Vero Cells
18.
J Leukoc Biol ; 109(1): 223-232, 2021 01.
Article in English | MEDLINE | ID: mdl-32745316

ABSTRACT

The efficacy of certain vaccines is improved by the use of adjuvants. Nowadays, the development of new, effective, and safe adjuvants that stimulate the innate immune response is researched. In this context, medicinal plants appear as a suitable alternative. Minthostachys verticillata essential oil (EO) has demonstrated the ability to modulate mechanisms of the innate immune response. Thus, the present work aimed to evaluate the EO adjuvant effect on humoral and cellular immunity, coadministered with OVA as antigen. The chemical analysis of EO by gas chromatography-mass spectrometry revealed a predominant pulegone-menthone chemotype. EO (1.25, 2.5, or 5.0 mg/ml) did not alter the viability of murine fibroblasts (3T3 cell line) neither showed signs of toxicity in Balb/c mice inoculated subcutaneously. The serum of mice immunized with OVA + EO showed increased levels of anti-OVA-specific antibodies of IgG1 subclass compared with the mice immunized with OVA alone revealing an adjuvant effect of EO. The delayed type hypersensitivity showed that the combination OVA + Al(OH)3  + EO was the best to induce a cellular immune response that extended until 48 h postinjection of OVA. M. verticillata EO appears as a new, safe, and effective adjuvant, which should continue to be studied for their possible future incorporation into vaccine formulations.


Subject(s)
Adjuvants, Immunologic/pharmacology , Lamiaceae/immunology , Oils, Volatile/pharmacology , Ovalbumin/immunology , Plant Oils/pharmacology , Aluminum Hydroxide/immunology , Aluminum Hydroxide/pharmacology , Animals , Immunity, Cellular/drug effects , Immunity, Cellular/immunology , Immunoglobulin G/immunology , Male , Mice , Mice, Inbred BALB C , Ovalbumin/pharmacology
19.
FASEB J ; 34(10): 14024-14041, 2020 10.
Article in English | MEDLINE | ID: mdl-32860638

ABSTRACT

Aluminium salts have been used in vaccines for decades. However, the mechanisms underlying their adjuvant effect are still unclear. Neutrophils, the first immune cells at the injection site, can release cellular DNA together with granular material, so-called neutrophil extracellular traps (NETs). In mice, NETs apparently play a role in aluminium hydroxide (alum)-adjuvant immune response to vaccines. Although no experimental data exist, this effect is assumed to be operative also in humans. As a first step to verify this knowledge in humans, we demonstrate that the injection of alum particles into human skin biopsies ex vivo leads to similar tissue infiltration of neutrophils and NET-formation. Moreover, we characterized the mechanism leading to alum-induced NET-release in human neutrophils as rapid, NADPH oxidase-independent process involving charge, phagocytosis, phagolysosomal rupture, Ca2+ -flux, hyperpolarization of the mitochondrial membrane, and mitochondrial ROS. Extracellular flow and inhibition experiments suggested that no additional energy from oxidative phosphorylation or glycolysis is required for NET-release. This study suggests a so far unappreciated role for neutrophils in the initial phase of immune responses to alum-containing vaccines in humans and provides novel insights into bioenergetic requirements of NET-formation.


Subject(s)
Adjuvants, Immunologic/pharmacology , Aluminum Hydroxide/pharmacology , Extracellular Traps , Lysosomes/metabolism , Membrane Potential, Mitochondrial , Neutrophil Infiltration , Neutrophils/drug effects , Calcium/metabolism , Cells, Cultured , Glycolysis , Humans , Mitochondria/metabolism , NADPH Oxidases/metabolism , Neutrophils/cytology , Neutrophils/immunology , Oxidative Phosphorylation
20.
Front Immunol ; 11: 1673, 2020.
Article in English | MEDLINE | ID: mdl-32849580

ABSTRACT

Respiratory syncytial virus (RSV) remains the most common cause of lower respiratory tract infections in children worldwide. Development of a vaccine has been hindered by the risk of developing enhanced respiratory disease (ERD) upon natural exposure to the virus. Generation of higher quality neutralizing antibodies with stabilized pre-fusion F protein antigens has been proposed as a strategy to prevent ERD. We sought to test whether there was evidence of ERD in naïve BALB/c mice immunized with an unadjuvanted, stabilized pre-fusion F protein, and challenged with RSV line 19. We further sought to determine the extent to which formulation with a Th2-biased (alum) or a more Th1/Th2-balanced (Advax-SM) adjuvant influenced cellular responses and lung pathology. When exposed to RSV, mice immunized with pre-fusion F protein alone (PreF) exhibited increased airway eosinophilia and mucus accumulation. This was further exacerbated by formulation of PreF with Alum (aluminum hydroxide). Conversely, formulation of PreF with a Th1/Th2-balanced adjuvant, Advax-SM, not only suppressed RSV viral replication, but also inhibited airway eosinophilia and mucus accumulation. This was associated with lower numbers of lung innate lymphocyte cells (ILC2s) and CD4+ T cells producing IL-5+ or IL-13+ and increased IFNγ+ CD4+ and CD8+ T cells, in addition to RSV F-specific CD8+ T cells. These data suggest that in the absence of preimmunity, stabilized PreF antigens may still be associated with aberrant Th2 responses that induce lung pathology in response to RSV infection, and can be prevented by formulation with more Th1/Th2-balanced adjuvants that enhance CD4+ and CD8+ IFNγ+ T cell responses. This may support the use of stabilized PreF antigens with Th1/Th2-balanced adjuvants like, Advax-SM, as safer alternatives to alum in RSV vaccine candidates.


Subject(s)
Adjuvants, Immunologic/pharmacology , Aluminum Hydroxide/pharmacology , Lung/drug effects , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/pharmacology , Respiratory Syncytial Viruses/drug effects , Th2 Cells/drug effects , Viral Fusion Proteins/pharmacology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Female , Immunity, Humoral/drug effects , Immunization , Immunogenicity, Vaccine/drug effects , Lung/immunology , Lung/pathology , Lung/virology , Mice, Inbred BALB C , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/pathology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/immunology , Respiratory Syncytial Viruses/pathogenicity , Th1-Th2 Balance/drug effects , Th2 Cells/immunology , Th2 Cells/metabolism , Th2 Cells/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...