Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
Front Behav Neurosci ; 18: 1341901, 2024.
Article in English | MEDLINE | ID: mdl-38698886

ABSTRACT

Prion diseases, such as scrapie, entail the accumulation of disease-specific prion protein (PrPSc) within the brain. Toll-like receptors (TLRs) are crucial components of the pattern recognition system. They recognize pathogen-associated molecular patterns (PAMPs) and play a central role in orchestrating host innate immune responses. The expression levels of Toll-like receptors (TLRs) in the central nervous system (CNS) were not well-defined. To establish a model of prion diseases in BALB/C mice, the 22L strain was employed. The features of the 22L strain were analyzed, and the cerebellum exhibited severe pathological changes. TLR1-13 levels in the cerebellum were measured using quantitative polymerase chain reaction (qPCR) at time points of 60, 90, 120, and the final end point (145 days post-infection). During the pathogenesis, the expression levels of Toll-like receptors (TLRs) 1, 2, 7, 8, and 9 increased in a time-dependent manner. This trend mirrored the expression patterns of PrPSc (the pathological isoform of the prion protein) and glial fibrillary acidic protein. Notably, at the end point, TLR1-13 levels were significantly elevated. Protein level of TLR7 and TLR9 showed increasing at the end point of the 22L-infected mice. A deeper understanding of the increased Toll-like receptors (TLRs) in prion diseases could shed light on their role in initiating immune responses at various stages during pathogenesis. This insight is particularly relevant when considering TLRs as potential therapeutic targets for prion diseases.

2.
Toxicol Lett ; 396: 19-27, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38642674

ABSTRACT

Ricin toxin (RT) is highly cytotoxic and can release a considerable amount of pro-inflammatory factors due to depurination, causing excessive inflammation that may aggravate the harm to the body. Pyroptosis, a type of gasdermin-mediated cell death, is a contributor to the exacerbation of inflammation. Accumulating evidence indicate that pyroptosis plays a significant role in the pathogen infection and tissue injury, suggesting a potential correlation between pyroptosis and RT-induced inflammation. Here, we aim to demonstrate this correlation and explore its molecular mechanisms. Results showed that RT triggers mouse alveolar macrophage MH-S cells pyroptosis by activating caspase-3 and cleaving Gasgermin E (GSDME). In contrast, inhibition of caspase-3 with Z-DEVD-FMK (inhibitor of caspase-3) or knockdown of GSDME attenuates this process, suggesting the essential role of caspase-3/GSDME-mediated pyroptosis in contributing to RT-induced inflammation. Collectively, our study enhances our understanding of a novel mechanism of ricin cytotoxicity, which may emerge as a potential target in immunotherapy to control the RT-induced inflammation.


Subject(s)
Caspase 3 , Inflammation , Pyroptosis , Ricin , Pyroptosis/drug effects , Ricin/toxicity , Animals , Mice , Caspase 3/metabolism , Inflammation/chemically induced , Inflammation/metabolism , Cell Line , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/metabolism , Gasdermins
3.
Toxicol Appl Pharmacol ; 485: 116890, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38492674

ABSTRACT

Ricin (ricin toxin, RT) has the potential to cause damage to multiple organs and systems. Currently, there are no existing antidotes, vaccinations, or effective therapies to prevent or treat RT intoxication. Apart from halting protein synthesis, RT also induces oxidative stress, inflammation and autophagy. To explore the mechanisms of RT-induced inflammatory injury and specific targets of prevention and treatment for RT poisoning, we characterized the role of cross-talk between autophagy and NLRP3 inflammasome in RT-induced damage and elucidated the underlying mechanisms. We showed that RT-induced inflammation was attributed to activation of the TLR4/MyD88/NLRP3 signaling and ROS production, evidenced by increased ASC speck formation and attenuated TXNIP/TRX-1 interaction, as well as pre-treatment with MCC950, MyD88 knockdown and NAC significantly reduced IL-1ß, IL-6 and TNF-α mRNA expression. In addition, autophagy is also enhanced in RT-triggered MLE-12 cells. RT elevated the levels of ATG5, p62 and Beclin1 protein, provoked the accumulation of LC3 puncta detected by immunofluorescence staining. Treatment with rapamycin (Rapa) reversed the RT-caused TLR4/MyD88/NLRP3 signaling activation, ASC specks formation as well as the levels of IL-1ß, IL-6 and TNF-α mRNA. In conclusion, RT promoted NLRP3 inflammasome activation and autophgay. Inflammation induced by RT was attenuated by autophagy activation, which suppressed the NLRP3 inflammasome. These findings suggest Rapa as a potential therapeutic drug for the treatment of RT-induced inflammation-related diseases.


Subject(s)
Autophagy , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Ricin , Signal Transduction , Autophagy/drug effects , Animals , Inflammasomes/metabolism , Inflammasomes/drug effects , Ricin/toxicity , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Mice , Signal Transduction/drug effects , Inflammation/metabolism , Inflammation/chemically induced , Cell Line , Toll-Like Receptor 4/metabolism , Myeloid Differentiation Factor 88/metabolism , Myeloid Differentiation Factor 88/genetics , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism
4.
Front Immunol ; 15: 1326033, 2024.
Article in English | MEDLINE | ID: mdl-38318188

ABSTRACT

Melittin, a main component of bee venom, is a cationic amphiphilic peptide with a linear α-helix structure. It has been reported that melittin can exert pharmacological effects, such as antitumor, antiviral and anti-inflammatory effects in vitro and in vivo. In particular, melittin may be beneficial for the treatment of diseases for which no specific clinical therapeutic agents exist. Melittin can effectively enhance the therapeutic properties of some first-line drugs. Elucidating the mechanism underlying melittin-mediated biological function can provide valuable insights for the application of melittin in disease intervention. However, in melittin, the positively charged amino acids enables it to directly punching holes in cell membranes. The hemolysis in red cells and the cytotoxicity triggered by melittin limit its applications. Melittin-based nanomodification, immuno-conjugation, structural regulation and gene technology strategies have been demonstrated to enhance the specificity, reduce the cytotoxicity and limit the off-target cytolysis of melittin, which suggests the potential of melittin to be used clinically. This article summarizes research progress on antiviral, antitumor and anti-inflammatory properties of melittin, and discusses the strategies of melittin-modification for its future potential clinical applications in preventing drug resistance, enhancing the selectivity to target cells and alleviating cytotoxic effects to normal cells.


Subject(s)
Bee Venoms , Melitten , Melitten/pharmacology , Melitten/chemistry , Melitten/metabolism , Antimicrobial Peptides , Bee Venoms/pharmacology , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Antiviral Agents
6.
Toxicol Lett ; 383: 177-191, 2023 Jul 01.
Article in English | MEDLINE | ID: mdl-37392970

ABSTRACT

γ-bungarotoxin (γ-BGT) is an RGD motif-containing protein, derived from the venom of Bungarus multicinctus, leading to acute death in mice. These RGD motif-containing proteins from snake venom belonging to the disintegrin family can interfere with vascular endothelial homeostasis by directly binding cell surface integrins. Targeting integrins that generate vascular endothelial dysfunction may contribute to γ-BGT poisoning, however, the underlying mechanisms have not been investigated in detail. In this study, the results showed that γ-BGT played a role in -promoting the permeability of the vascular endothelial barrier. Depending on its selective binding to integrin α5 in vascular endothelium (VE), γ-BGT initiated downstream events, including focal adhesion kinase dephosphorylation and cytoskeleton remodeling, resulting in the intercellular junction interruption. Those alternations facilitated paracellular permeability of VE and barrier dysfunction. Proteomics profiling identified that as a downstream effector of the integrin α5 / FAK signaling pathway cyclin D1 partially mediated the cellular structural changes and barrier dysfunction. Furthermore, VE-released plasminogen activator urokinase and platelet-derived growth factor D could serve as potential diagnostic biomarkers for γ-BGT-induced vascular endothelial dysfunction. Our results indicate the mechanisms through which γ-BGT as a novel disintegrin directly interacts with the VE, with consequences for barrier dysfunction.


Subject(s)
Bungarotoxins , Endothelium, Vascular , Integrin alpha5 , Snake Venoms , Animals , Mice , Bungarotoxins/toxicity , Disintegrins/pharmacology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Integrin alpha5/metabolism , Integrins/metabolism , Oligopeptides , Snake Venoms/toxicity
7.
Toxicol Lett ; 383: 152-161, 2023 Jul 01.
Article in English | MEDLINE | ID: mdl-37390852

ABSTRACT

DNA-encoded monoclonal antibodies (DMAbs) and in vivo expression of antibody therapeutics presents an innovative alternative to conventional delivery methods. Therefore, in order to prevent the lethal dose of ricin toxin (RT) and to avoid human anti-mouse antibody (HAMA) reaction, we developed the human neutralizing antibody 4-4E against RT and constructed DMAb-4-4E. The human neutralizing antibody 4-4E could neutralize RT in vitro and in vivo, while the mice in RT group all died. Using intramuscular electroporation (IM EP), antibodies were rapidly expressed in vivo within 7 days and were enriched in intestine and gastrocnemius muscle mostly. Besides, we found that DMAbs have shown a broad protective efficacy of RT poisoning prophylaxis. Driven by plasmids for IgG expression, mice were survived and the blood glucose level of mice in DMAb-IgG group returned to normal at 72 h post RT challenge, and the RT group died within 48 h. Furthermore, hindrance of protein disulfide isomerase (PDI) and accumulation of RT in endosomes were found in IgG-protected cells, revealing the possible mechanism of neutralization details. These data support the further study of RT-neutralizing monoclonal antibodies (mAbs) in the development.


Subject(s)
Foodborne Diseases , Plant Poisoning , Poisoning , Ricin , Toxins, Biological , Animals , Mice , Humans , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing , Ricin/toxicity , Immunoglobulin G , Mice, Inbred BALB C , Poisoning/prevention & control
8.
Front Cell Infect Microbiol ; 13: 1155293, 2023.
Article in English | MEDLINE | ID: mdl-37207187

ABSTRACT

Introduction: The constantly mutating SARS-CoV-2 has been infected an increasing number of people, hence the safe and efficacious treatment are urgently needed to combat the COVID-19 pandemic. Currently, neutralizing antibodies (Nabs), targeting the receptor-binding domain (RBD) of the SARS-CoV-2 spike protein are potentially effective therapeutics against COVID-19. As a new form of antibody, bispecific single chain antibodies (BscAbs) can be easily expressed in E. coli and exhibits broad-spectrum antiviral activity. Methods: In this study, we constructed two BscAbs 16-29, 16-3022 and three single chain variable fragments (scFv) S1-16, S2-29 and S3022 as a comparison to explore their antiviral activity against SARS-CoV-2. The affinity of the five antibodies was characterized by ELISA and SPR and the neutralizing activity of them was analyzed using pseudovirus or authentic virus neutralization assay. Bioinformatics and competitive ELISA methods were used to identify different epitopes on RBD. Results: Our results revealed the potent neutralizing activity of two BscAbs 16-29 and 16-3022 against SARS-CoV-2 original strain and Omicron variant infection. In addition, we also found that SARS-CoV RBD-targeted scFv S3022 could play a synergistic role with other SARS-CoV-2 RBD-targeted antibodies to enhance neutralizing activity in the form of a BscAb or in cocktail therapies. Discussion: This innovative approach offers a promising avenue for the development of subsequent antibody therapies against SARSCoV-2. Combining the advantages of cocktails and single-molecule strategies, BscAb therapy has the potential to be developed as an effective immunotherapeutic for clinical use to mitigate the ongoing pandemic.


Subject(s)
COVID-19 , Single-Chain Antibodies , Humans , SARS-CoV-2/genetics , Escherichia coli , Pandemics , Antibodies, Monoclonal , Antibodies, Neutralizing , Single-Chain Antibodies/genetics , Antibodies, Viral/therapeutic use , Antiviral Agents
9.
Hum Vaccin Immunother ; 19(1): 2158670, 2023 12 31.
Article in English | MEDLINE | ID: mdl-37067182

ABSTRACT

Melanoma stem cells (MSCs)-based vaccine strategies have been a potent immunotherapeutic approach for melanoma treatment, which aimed at inducing specific anti-tumor immunity and targeting cancer stem-like cells. As the main cancer-fighting immune cells, CD8+T cells play an important role in vaccine-induced antitumor immunity. Here, we developed a novel MSC vaccine that induces CD8+T cells to target melanoma stem cells specifically. The MSC vaccine was prepared for our study in order to determine the effectiveness of antitumor immunity. The proportion and activity of CD8+T cells were examined in the spleen after immunization, in particular, the expression and cytotoxicity of the immune checkpoint of spleen lymphocytes were detected by flow cytometry and ELISA, moreover, tumor size and the number of lung metastasis nodules were observed and the specific killing effect of the vaccine was evaluated in immunized mice. We found that the MSC vaccine could promote DCs maturation, activate CD8+T cells, suppress the expression of CTLA-4, PD-1, and Tim-3, and increase the expression of IFN-γ and GzmB of CD8+T cells. Melanoma growth and metastasis were inhibited by the vaccine's specific targeted killing effect. The vaccines based on melanoma stem cells (MSCs) delay the progression of melanoma by inducing anti-tumor immune responses in CD8+T cells.


Subject(s)
Cancer Vaccines , Lung Neoplasms , Melanoma , Mice , Animals , Melanoma/drug therapy , CD8-Positive T-Lymphocytes , Lung Neoplasms/drug therapy , Immunization , Stem Cells , Mice, Inbred C57BL
10.
Toxicon ; 224: 107046, 2023 Mar 01.
Article in English | MEDLINE | ID: mdl-36702354

ABSTRACT

Acting as microRNA (miRNA) sponges, circular RNAs (circRNAs) have been discovered to be critical modulators of inflammatory processes. Ricin Toxin (RT) is highly toxic to mammalian cells and low doses of RT can induce acute inflammation. However, current researches on the underlying mechanism and function of circRNA/miRNA network in RT-induced inflammation are limited. Previously, we found miR-221-5p was aberrant and associated with the inflammation of RT induction. In this study, based on the circRNA high-throughput sequencing (circRNA-seq), we obtained a novel circRNA termed circNLRP3 and revealed that circNLRP3 can sponge miR-221-5p, release its target mRNA A20, and further suppress NF-κB signaling pathway to alleviated RT-induced TNF-α production. Our findings elucidated a possible mechanistic link between the circNLRP3/miR-221-5p/A20 axis and RT-induced inflammatory response, which may broaden our understanding of RT poisoning.


Subject(s)
MicroRNAs , Ricin , Animals , RNA, Circular , Tumor Necrosis Factor-alpha , MicroRNAs/genetics , Inflammation , Mammals/genetics , Mammals/metabolism
11.
Toxicon ; 214: 47-53, 2022 Jul 30.
Article in English | MEDLINE | ID: mdl-35595086

ABSTRACT

Ricin toxin (RT) belongs to the ribosome-inactivating protein (RIP) family of toxins and is considered to be a moderate threat by the US Center of Disease Control and Prevention (CDC). RT poses a great potential threat to the public, but there has been a lack of effective treatment options so far. Over the past few decades, researches on the prevention and treatment of RT poisoning have been investigated, among which neutralizing antibodies targeting RT specifically have always been a research hotspot. In this review, we have summarized the mechanism of action of RT, the research results and the design strategies of RT neutralizing antibodies, and discussed the key issues in the development of RT neutralizing antibody researches.


Subject(s)
Ricin , Antibodies, Neutralizing , Ricin/toxicity
12.
Toxicon ; 212: 11-18, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35390424

ABSTRACT

Ricin toxin (RT) is one of the most lethal type II ribosome-inactivating proteins (RIP), and is classified as a potential bioterror agent due to its severe cytotoxicity and high availability. The toxicity of RT is dependent on both dose and route of exposure. Increasing evidence demonstrates that sub-lethal RT induces acute inflammation and increases the release of pro-inflammatory cytokines. However, current studies on mechanism of RT-induced inflammation are limited. In this study, to evaluate the relationship between miRNAs and RT-induced inflammation, RNA sequencing (RNA-Seq) was used to analyze the expression of miRNAs and mRNAs in RT-treated RAW264.7 macrophage cells. A total of 14 significantly differently expressed (DE) miRNAs and 323 miRNA-mRNA interaction pairs were predicted by bioinformatics analysis. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis revealed that majority of those interaction pairs were involved in PI3K/Akt pathway. In addition, overexpression of miR-221-5p promoted the inflammatory response by inhibiting the mRNA expression of COL4a5. This work contributes to our understanding of RT-induced inflammation and demonstrates the potential role of miRNAs in innate immunity, which may be regarded as potential targets in developing therapies for RT poisoning.


Subject(s)
MicroRNAs , Ricin , Collagen Type IV/toxicity , Humans , Inflammation/chemically induced , Inflammation/metabolism , MicroRNAs/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , Ricin/toxicity , Signal Transduction
13.
Front Pharmacol ; 12: 767900, 2021.
Article in English | MEDLINE | ID: mdl-34744746

ABSTRACT

Increasing studies have concentrated on investigating circular RNAs (circRNAs) as pivotal regulators in the progression of numerous diseases and biological processes and abundant evidence shows that circRNAs are participated in the regulation of innate immune responses. Several studies showed that Ricin Toxin (RT) could induce inflammatory injury. There was no research on the particular functions and underlying mechanisms of circRNAs in RT-induced inflammation. In this study, RNA sequencing performed on RT-treated and normal RAW264.7 macrophage cells was used to investigated the differentially expressed circRNAs. Based on the dataset, the expression of circEpc1 (mmu_circ_0,000,842) was identified higher in RT-treated cells. Moreover, gain-and-loss function assays showed that circEpc1 function as a promoter in RT-induced inflammation in vivo and in vitro. Mechanistically, circEpc1 acted as a miR-5114 sponge to relieve the suppressive effect of miR-5114 on its target NOD2 and thereby activating NF-κB and MAPK signaling pathways. Our results illuminated a link between RT-induced inflammation and the circEpc1 regulatory loop and provided novel insight into the functions of circRNA in innate immune, which may emerge as a potential target in immunotherapy to control the RT-induced inflammatory injury.

14.
Toxicon ; 203: 129-138, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34673083

ABSTRACT

Ricin toxin (RT) is a ribosome-inactivating protein derived from the beans of the castor oil plant. Our previous studies have reported that RT can induce the production of inflammatory cytokines and cause inflammatory injury in RAW264.7 cells. In order to explore the various biological processes that long noncoding RNA (lncRNA), circular RNA (circRNA) and micro RNA (miRNA) as endogenous non-coding RNAs (ceRNAs) may participate in the pro-inflammatory mechanism, RT (20 ng/mL) treated and normal RAW264.7 cells were firstly sequenced by RNA-seq. By comparing the differentially expressed genes, we obtained 10 hub genes and enriched the inflammatory-related signaling pathways. Based on our results, we concluded a lncRNA/circRNA-miRNA-mRNA network. Finally, we verified the key genes and pathways by qRT-PCR, WB and ELISA. From the experiment results, an opening MAPK signaling pathway in TNF signaling pathway via TNFR2 was found involved in RT-induced inflammation. This work provides a reference for searching for ceRNA targets or therapeutic drugs in RT-induced inflammatory injury in the future.


Subject(s)
MicroRNAs , RNA, Long Noncoding , Ricin , Animals , Gene Regulatory Networks , Inflammation/chemically induced , Mice , MicroRNAs/genetics , RAW 264.7 Cells , RNA, Circular , RNA, Long Noncoding/genetics , RNA, Messenger , Ricin/toxicity
15.
Aging (Albany NY) ; 13(14): 18515-18526, 2021 07 22.
Article in English | MEDLINE | ID: mdl-34292876

ABSTRACT

Agomelatine is a non-selective melatonin receptor agonist and an atypical antidepressant with anti-inflammatory, neuroprotective, and cardioprotective effects. The renin-angiotensin system modulates blood pressure and vascular homeostasis. Angiotensin II (Ang II) and its receptor Ang II type I receptor (AT1R) are recognized as contributors to the pathogenesis of cardiovascular and cardiometabolic diseases, including diabetes, obesity, and atherosclerosis. The recruitment and attachment of monocytes to the vascular endothelium is a major event in the early stages of atherosclerosis and other cardiovascular diseases. In the present study, we demonstrate that agomelatine reduced Ang II-induced expression of AT1R while significantly inhibiting the attachment of monocytes to endothelial cells induced by Ang II and mediated by ICAM-1 and VCAM-1. Additionally, Ang II inhibited the expression of the chemokines CXCL1, MCP-1, and CCL5, which are critical in the process of immune cell recruitment and invasion. Agomelatine also suppressed the expression of TNF-α, IL-8, and IL-12, which are proinflammatory cytokines that promote endothelial dysfunction and atherogenesis. Importantly, we demonstrate that the inhibitory effect of agomelatine against the expression of adhesion molecules is mediated through the downregulation of Egr-1 signaling. Together, our findings provide evidence of a novel mechanism of agomelatine that may be practicable in the treatment and prevention of cardiovascular diseases.


Subject(s)
Acetamides/pharmacology , Angiotensin II/pharmacology , Cell Adhesion/drug effects , Endothelial Cells/drug effects , Leukocytes, Mononuclear/drug effects , Cardiovascular Diseases/prevention & control , Chemokine CCL2/metabolism , Early Growth Response Protein 1/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/drug effects , Humans , Leukocytes, Mononuclear/metabolism , Signal Transduction/drug effects , Vascular Cell Adhesion Molecule-1/metabolism
16.
Toxicol Lett ; 347: 67-77, 2021 Sep 01.
Article in English | MEDLINE | ID: mdl-33865919

ABSTRACT

Ricin toxin (RT) is one of the most lethal toxins derived from the seed of castor beans. In addition to its main toxic mechanism of inhibiting the synthesis of cellular proteins, RT can induce the production of inflammatory cytokines. MicroRNAs (miRNAs) play a key role in regulating both innate and adaptive immunity. To elucidate the regulation of miRNAs in RT-induced inflammation injury, the RNA high-throughput sequencing (RNA-Seq) technology was used to analyze the expression profile of miRNAs and mRNAs in RT-treated RAW264.7 cells. Results showed that a total of 323 mRNAs and 19 miRNAs differentially expressed after RT treated. Meanwhile, 713 miRNA-mRNA interaction pairs were identified by bioinformatics analysis. KEGG (Kyoto Encyclopedia of Genes and Genomes) pathway analysis showed that those interaction pairs were mainly involved in JAK-STAT, T cell receptor, and MAPK signaling pathways. Moreover, we further predicted and determined the targeting relationship between miR-155-3p and GAB2 through TargetScan and dual-luciferase reporter assay. Mechanically, overexpression of miR-155-3p can reduce the secretion of TNF-α in RAW264.7 cells, revealing a possible mechanism of miR-155-3p regulating RT-induced inflammatory injury. This study provides a new perspective for clarifying the mechanism of RT-induced inflammatory injury and reveals the potential role of miRNAs in innate immune regulation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Inflammation Mediators/metabolism , Inflammation/chemically induced , Macrophages/drug effects , MicroRNAs/metabolism , RNA, Messenger/metabolism , Ricin/toxicity , Transcriptome , Adaptor Proteins, Signal Transducing/genetics , Animals , Gene Expression Profiling , Gene Expression Regulation , Gene Regulatory Networks , Inflammation/genetics , Inflammation/metabolism , Macrophages/metabolism , Mice , MicroRNAs/genetics , RAW 264.7 Cells , RNA, Messenger/genetics , Signal Transduction
17.
RSC Adv ; 11(26): 16096-16105, 2021 Apr 26.
Article in English | MEDLINE | ID: mdl-35481179

ABSTRACT

The recovery of boron from salt lake brines has become an effective way to meet the increasing demand, particularly in China. In this study, nine commercially available monohydric alcohols with different structures were selected for boron extraction from a salt lake brine with high magnesium content. Passing through the optimization, isodecanol was finally selected for the detailed study on boron recovery from brine due to its moderate viscosity, lower solubility water entrainment and high extraction efficiency. Parameter effects on boron extraction were systematically studied, including equilibrium pH, organic concentration, phase ratio, temperature, and salting-out effect. A McCabe-Thiele diagram was established to determine the boron extraction and stripping stages. An organic solution containing 2.5 mol L-1 isodecanol was used for a simulated three-stage counter-current extraction test under optimized extraction conditions, and the boron extraction reached 99.07%. A simulated four-stage counter-current stripping test was carried out with water with the stripping efficiency of 98.71%. In total the boron recovery reached 97.79%. Additionally, the mechanism of boron extraction by isodecanol was investigated using both slope analysis method and Fourier transform infrared spectroscopy (FTIR). The stoichiometric ratio of isodecanol to boron required to form boric acid ester was found to be 1.268. These results indicate that the extraction system has great potential for commercial application in boron recovery from salt lake brines with high magnesium content.

18.
Mikrochim Acta ; 187(12): 679, 2020 11 28.
Article in English | MEDLINE | ID: mdl-33247373

ABSTRACT

A point-of-care (POC) immunoassay was established for the sensitive and rapid detection of pathogenic Escherichia coli O157:H7, using magnetic Fe3O4 organic-inorganic composites (Ab@Fe3O4) for immunomagnetic separation, nanozyme platinum nanoparticle (PtNp) organic-inorganic composites (Ap@PtNp) for signal amplification, and thermometer readings. Antibodies and Fe3O4 were incubated in Cu2+ phosphate buffer to synthesize the magnetic composite Ab@Fe3O4 with antibodies, to specifically capture E. coli O157:H7. Antimicrobial peptides and PtNp were incubated in Cu2+ phosphate buffer to synthesize the signal composites Ap@PtNp with antimicrobial peptides (magainin I), recognizing and labeling E. coli O157:H7. In the presence of E. coli O157:H7, magnetic microcomposites targeted bacteria and signal microcomposites to form the sandwich structure: Ab@Fe3O4-bacteria-Ap@PtNp for magnetic separation. Ap@PtNp of signal composites catalyzed H2O2 to generate thermo-signals (temperature rise), which were determined by a thermometer. This point-of-care bioassay detected E. coli O157:H7 in the linear range of 101-107 CFU mL-1 and with a detection limit of 14 CFU mL-1. One-pot process magnetic Fe3O4 organic-inorganic composites (Ab@Fe3O4, magnetic microcomposites, MMC) for immunomagnetic separation and nanozyme platinum nanoparticle (PtNp) organic-inorganic composites (Ap@PtNp, signal microcomposites, SMC) were used as signal amplification and thermometer readings for E. coli O157:H7 detection.


Subject(s)
Antibodies, Bacterial/immunology , Escherichia coli O157/isolation & purification , Ferrosoferric Oxide/chemistry , Immunoassay/methods , Magnetics , Metal Nanoparticles/chemistry , Antibodies, Bacterial/chemistry , Escherichia coli O157/immunology , Food Microbiology , Immunoassay/instrumentation , Platinum/chemistry , Point-of-Care Systems , Thermometers
19.
Front Pharmacol ; 11: 526129, 2020.
Article in English | MEDLINE | ID: mdl-33013378

ABSTRACT

Ricin toxin binding subunit B (RTB) is a galactose-binding lectin protein derived from the beans of the castor oil plant (Ricinus communis). Our previous studies have reported a direct immunomodulatory effect of recombinant RTB, which stimulates RAW264.7 cells to produce cytokines including TNF-α. However, the role of RTB in innate immune response and its specific mechanism have not been reported in detail. In this work, the results showed that RTB treatment of macrophages significantly increased TLR4 protein levels. RTB also activated TLR4 downstream events, including MyD88, IRAK, and TRAF6, resulting in macrophage activation and TNF-α production. This process is reflected in the increase of IκB phosphorylation. TLR4 knockdown macrophages treated with RTB exhibited greatly reduced IκB phosphorylation and TNF-α secretion. Moreover, treatment with MyD88 inhibitor also suppressed TNF-α production. The docking of RT and TLR4 was simulated by computer, and the contact residues were concentrated on RTB. Our results suggest that recombinant RTB can activate mouse macrophages to secrete TNF-α through activation of NF-κB via the TLR4 signaling pathways.

20.
Mikrochim Acta ; 187(11): 600, 2020 10 09.
Article in English | MEDLINE | ID: mdl-33034762

ABSTRACT

An electrochemical immunosensor based on ferrocene (Fc)-functionalized nanocomposites was fabricated as an efficient electroactive signal probe to amplify electrochemical signals for Salmonella typhimurium detection. The electrochemical signal amplification probe was constructed by encapsulating ferrocene into S. typhimurium-specific antimicrobial peptides Magainin I (MI)-Cu3(PO4)2 organic-inorganic nanocomposites (Fc@MI) through a one-step process. Magnetic beads (MBs) coupled with antibody were used as capture ingredient for target magnetic separation, and Fc@MI nanoparticles were used as signal labels in the immunoassays. The sandwich of MBs-target-Fc@MI assay was performed using a screen-printed carbon electrode as transducer surface. The immunosensor platform presents a low limit of detection (LOD) of 3 CFU·mL-1 and a linear range from 10 to 107 CFU·mL-1, with good specificity and precision, and was successfully applied for S. typhimurium detection in milk. Graphical abstract One-pot process antimicrobial peptides Magainin I-Cu3(PO4)2 organic-inorganic nanocomposites (Fc@MI) were used as ideal electrochemical signal label, integrating both essential functions of biological recognition and signal amplification. Screen-printed carbon electrode (SPCE) was used as the electrochemical system for Salmonella typhimurium detection.


Subject(s)
Electrochemical Techniques/instrumentation , Ferrous Compounds/chemistry , Immunoassay/methods , Metallocenes/chemistry , Nanocomposites/chemistry , Salmonella typhimurium/isolation & purification , Electrochemical Techniques/methods , Sensitivity and Specificity , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL