Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 20
1.
Int J Mol Sci ; 25(6)2024 Mar 07.
Article En | MEDLINE | ID: mdl-38542074

Lethal toxin (LT) is the critical virulence factor of Bacillus anthracis, the causative agent of anthrax. One common symptom observed in patients with anthrax is thrombocytopenia, which has also been observed in mice injected with LT. Our previous study demonstrated that LT induces thrombocytopenia by suppressing megakaryopoiesis, but the precise molecular mechanisms behind this phenomenon remain unknown. In this study, we utilized 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced megakaryocytic differentiation in human erythroleukemia (HEL) cells to identify genes involved in LT-induced megakaryocytic suppression. Through cDNA microarray analysis, we identified Dachshund homolog 1 (DACH1) as a gene that was upregulated upon TPA treatment but downregulated in the presence of TPA and LT, purified from the culture supernatants of B. anthracis. To investigate the function of DACH1 in megakaryocytic differentiation, we employed short hairpin RNA technology to knock down DACH1 expression in HEL cells and assessed its effect on differentiation. Our data revealed that the knockdown of DACH1 expression suppressed megakaryocytic differentiation, particularly in polyploidization. We demonstrated that one mechanism by which B. anthracis LT induces suppression of polyploidization in HEL cells is through the cleavage of MEK1/2. This cleavage results in the downregulation of the ERK signaling pathway, thereby suppressing DACH1 gene expression and inhibiting polyploidization. Additionally, we found that known megakaryopoiesis-related genes, such as FOSB, ZFP36L1, RUNX1, FLI1, AHR, and GFI1B genes may be positively regulated by DACH1. Furthermore, we observed an upregulation of DACH1 during in vitro differentiation of CD34-megakaryocytes and downregulation of DACH1 in patients with thrombocytopenia. In summary, our findings shed light on one of the molecular mechanisms behind LT-induced thrombocytopenia and unveil a previously unknown role for DACH1 in megakaryopoiesis.


Anthrax , Bacillus anthracis , Leukemia, Erythroblastic, Acute , Thrombocytopenia , Animals , Humans , Mice , Antigens, Bacterial/metabolism , Bacillus anthracis/metabolism , Butyrate Response Factor 1/metabolism , Cell Differentiation , Thrombocytopenia/chemically induced , Thrombocytopenia/genetics
2.
Antimicrob Agents Chemother ; 68(4): e0095623, 2024 Apr 03.
Article En | MEDLINE | ID: mdl-38446062

Viral RNA-dependent RNA polymerase (RdRp), a highly conserved molecule in RNA viruses, has recently emerged as a promising drug target for broad-acting inhibitors. Through a Vero E6-based anti-cytopathic effect assay, we found that BPR3P0128, which incorporates a quinoline core similar to hydroxychloroquine, outperformed the adenosine analog remdesivir in inhibiting RdRp activity (EC50 = 0.66 µM and 3 µM, respectively). BPR3P0128 demonstrated broad-spectrum activity against various severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants of concern. When introduced after viral adsorption, BPR3P0128 significantly decreased SARS-CoV-2 replication; however, it did not affect the early entry stage, as evidenced by a time-of-drug-addition assay. This suggests that BPR3P0128's primary action takes place during viral replication. We also found that BPR3P0128 effectively reduced the expression of proinflammatory cytokines in human lung epithelial Calu-3 cells infected with SARS-CoV-2. Molecular docking analysis showed that BPR3P0128 targets the RdRp channel, inhibiting substrate entry, which implies it operates differently-but complementary-with remdesivir. Utilizing an optimized cell-based minigenome RdRp reporter assay, we confirmed that BPR3P0128 exhibited potent inhibitory activity. However, an enzyme-based RdRp assay employing purified recombinant nsp12/nsp7/nsp8 failed to corroborate this inhibitory activity. This suggests that BPR3P0128 may inhibit activity by targeting host-related RdRp-associated factors. Moreover, we discovered that a combination of BPR3P0128 and remdesivir had a synergistic effect-a result likely due to both drugs interacting with separate domains of the RdRp. This novel synergy between the two drugs reinforces the potential clinical value of the BPR3P0128-remdesivir combination in combating various SARS-CoV-2 variants of concern.


Adenosine Monophosphate/analogs & derivatives , Alanine/analogs & derivatives , COVID-19 , Pyrazoles , Quinolines , Humans , SARS-CoV-2/metabolism , RNA-Dependent RNA Polymerase/metabolism , Molecular Docking Simulation , COVID-19 Drug Treatment , Antiviral Agents/chemistry
3.
Mol Ther ; 31(11): 3322-3336, 2023 11 01.
Article En | MEDLINE | ID: mdl-37689971

The ongoing evolution of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), resulting in the emergence of new variants that are resistant to existing vaccines and therapeutic antibodies, has raised the need for novel strategies to combat the persistent global COVID-19 epidemic. In this study, a monoclonal anti-human angiotensin-converting enzyme 2 (hACE2) antibody, ch2H2, was isolated and humanized to block the viral receptor-binding domain (RBD) binding to hACE2, the major entry receptor of SARS-CoV-2. This antibody targets the RBD-binding site on the N terminus of hACE2 and has a high binding affinity to outcompete the RBD. In vitro, ch2H2 antibody showed potent inhibitory activity against multiple SARS-CoV-2 variants, including the most antigenically drifted and immune-evading variant Omicron. In vivo, adeno-associated virus (AAV)-mediated delivery enabled a sustained expression of monoclonal antibody (mAb) ch2H2, generating a high concentration of antibodies in mice. A single administration of AAV-delivered mAb ch2H2 significantly reduced viral RNA load and infectious virions and mitigated pulmonary pathological changes in mice challenged with SARS-CoV-2 Omicron BA.5 subvariant. Collectively, the results suggest that AAV-delivered hACE2-blocking antibody provides a promising approach for developing broad-spectrum antivirals against SARS-CoV-2 and potentially other hACE2-dependent pathogens that may emerge in the future.


Antibodies, Monoclonal , Broadly Neutralizing Antibodies , COVID-19 , Animals , Humans , Mice , Angiotensin-Converting Enzyme 2/genetics , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antibodies, Viral , COVID-19/therapy , Dependovirus/genetics , RNA, Viral , SARS-CoV-2/genetics , Broadly Neutralizing Antibodies/pharmacology , Broadly Neutralizing Antibodies/therapeutic use
4.
Biomed J ; 44(3): 293-303, 2021 06.
Article En | MEDLINE | ID: mdl-34119448

BACKGROUND: While severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection presents with mild or no symptoms in most cases, a significant number of patients become critically ill. Remdesivir has been approved for the treatment of coronavirus disease 2019 (COVID-19) in several countries, but its use as monotherapy has not substantially lowered mortality rates. Because agents from traditional Chinese medicine (TCM) have been successfully utilized to treat pandemic and endemic diseases, we designed the current study to identify novel anti-SARS-CoV-2 agents from TCM. METHODS: We initially used an antivirus-induced cell death assay to screen a panel of herbal extracts. The inhibition of the viral infection step was investigated through a time-of-drug-addition assay, whereas a plaque reduction assay was carried out to validate the antiviral activity. Direct interaction of the candidate TCM compound with viral particles was assessed using a viral inactivation assay. Finally, the potential synergistic efficacy of remdesivir and the TCM compound was examined with a combination assay. RESULTS: The herbal medicine Perilla leaf extract (PLE, approval number 022427 issued by the Ministry of Health and Welfare, Taiwan) had EC50 of 0.12 ± 0.06 mg/mL against SARS-CoV-2 in Vero E6 cells - with a selectivity index of 40.65. Non-cytotoxic PLE concentrations were capable of blocking viral RNA and protein synthesis. In addition, they significantly decreased virus-induced cytokine release and viral protein/RNA levels in the human lung epithelial cell line Calu-3. PLE inhibited viral replication by inactivating the virion and showed additive-to-synergistic efficacy against SARS-CoV-2 when used in combination with remdesivir. CONCLUSION: Our results demonstrate for the first time that PLE is capable of inhibiting SARS-CoV-2 replication by inactivating the virion. Our data may prompt additional investigation on the clinical usefulness of PLE for preventing or treating COVID-19.


Drugs, Chinese Herbal/pharmacology , Perilla frutescens , Plant Extracts/pharmacology , SARS-CoV-2/drug effects , Virus Inactivation , Animals , COVID-19 , Chlorocebus aethiops , Humans , Perilla frutescens/chemistry
5.
Carbohydr Polym ; 236: 116041, 2020 May 15.
Article En | MEDLINE | ID: mdl-32172855

Fucoidan/trimethylchitosan nanoparticles (FUC-TMC-NPs) have the potential to improve the immunostimulating efficiency of anthrax vaccine adsorbed (AVA). FUC-TMC-NPs with positive (+) or negative (-) surface charges were prepared via polyelectrolyte complexation, both charged NP types permitted high viability and presented no cytotoxicity on L929, A549 and JAWS II dendritic cells. Flow cytometry measurements indicated lower (+)-FUC-TMC-NPs internalization levels than (-)-FUC-TMC-NPs, yet produced high levels of pro-inflammatory cytokines IFN-γ, IL12p40, and IL-4. Moreover, fluorescence microscope images proved that both charged NP could deliver drugs into the nucleus. In vivo studies on A/J mice showed that (+)-FUC-TMC-NPs carrying AVA triggered an efficient response with a higher IgG anti-PA antibody titer than AVA with CpG oligodeoxynucleotides, and yielded 100 % protection when challenged with the anthracis spores. Furthermore, PA-specific IgG1 and IgG2a analysis confirmed that (+)-FUC-TMC-NPs strongly stimulated humoral immunity. In conclusion, (+)-FUC-TMC-NP is promising anthrax vaccine adjuvant as an alternative to CpG.


Adjuvants, Immunologic/therapeutic use , Anthrax Vaccines/therapeutic use , Chitosan/analogs & derivatives , Chitosan/therapeutic use , Nanoparticles/therapeutic use , Polysaccharides/therapeutic use , A549 Cells , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/toxicity , Animals , Anthrax/therapy , Anthrax Vaccines/immunology , Bacillus anthracis/immunology , Chitosan/toxicity , Cytokines/metabolism , Female , Humans , Mice , Nanoparticles/toxicity , Oligodeoxyribonucleotides/therapeutic use , Polysaccharides/chemistry , Polysaccharides/toxicity
6.
Carbohydr Polym ; 229: 115403, 2020 Feb 01.
Article En | MEDLINE | ID: mdl-31826481

We examined the efficacy of fucoidan-N-(2-hydroxy-3-trimethylammonium)propylchitosan nanoparticles (FUC-HTCC NPs) as adjuvants for anthrax vaccine adsorbed (AVA). Positively and negatively surface-charged FUC-HTCC NPs were prepared via polyelectrolyte complexation by varying the mass ratio of FUC and HTCC. When cultured with L929 cells or JAWS II dendritic cells, both charged NPs showed high cell viability and low cytotoxicity, observed via MTT assay and lactate dehydrogenase release assay, respectively. In addition, we have monitored excellent NPs uptake efficacy by dendritic cells and observed that combining FUC-HTCC NPs with AVA significantly increases the magnitude of IgG-anti-protective antigen titers in A/J mice compared to that by CpG oligodeoxynucleotides plus AVA or AVA alone, and PA-specific IgG1 and IgG2a analysis confirmed that FUC-HTCC NPs strongly stimulated humoral immunity. Furthermore, FUC-HTCC NPs plus AVA provided a superior survival rate (100%) of A/J mice compared to CpG oligodeoxynucleotides plus AVA (75%) or AVA alone (50%) following anthrax lethal toxin challenge. The findings support FUC-HTCC NPs as a potential adjuvant of AVA for rapid induction of protective immunity.


Adjuvants, Immunologic/administration & dosage , Anthrax Vaccines/administration & dosage , Chitosan/administration & dosage , Nanoparticles/administration & dosage , Polysaccharides/administration & dosage , Animals , Cell Line , Cell Survival/drug effects , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Female , Mice , Oligodeoxyribonucleotides
7.
Virulence ; 8(7): 1216-1228, 2017 10 03.
Article En | MEDLINE | ID: mdl-28102766

As one of the virulence factors of Bacillus anthracis, lethal toxin (LT) induces various pathogenic responses including the suppression of the coagulation system. In this study, we observed that LT markedly increased the circulating soluble P-selectin (sP-sel) levels and microparticle (MP) count in wild-type but not P-selectin (P-sel, Selp-/-) or P-sel ligand-1 (PSGL-1, Selplg-/-) knockout mice. Because sP-sel induces a hypercoagulable state through PSGL-1 pathway to generate tissue factor-positive MPs, we hypothesized that the increase in plasma sP-sel levels can be a self-rescue response in hosts against the LT-mediated suppression of the coagulation system. In agreement with our hypothesis, our results indicated that compared with wild-type mice, Selp-/- and Selplg-/- mice were more sensitive to LT. In addition, the recombinant sP-sel treatment markedly ameliorated LT-mediated pathogenesis and reduced mortality. As a result, elicitation of circulating sP-sel is potentially a self-rescue response, which is beneficial to host recovery from an LT-induced hypocoagulation state. These results suggest that the administration of sP-sel is likely to be useful in the development of a new strategy to treat anthrax.


Anthrax/drug therapy , Antigens, Bacterial/toxicity , Bacterial Toxins/toxicity , Membrane Glycoproteins/metabolism , P-Selectin/administration & dosage , Animals , Anthrax/metabolism , Anthrax/mortality , Anthrax/physiopathology , Antigens, Bacterial/metabolism , Bacillus anthracis/metabolism , Bacterial Toxins/metabolism , Blood Coagulation/drug effects , Hemostasis , Humans , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL
8.
Nanomaterials (Basel) ; 6(12)2016 Dec 09.
Article En | MEDLINE | ID: mdl-28335365

The bactericidal activity of conventional titanium dioxide (TiO2) photocatalyst is effective only on irradiation by ultraviolet light, which restricts the applications of TiO2 for use in living environments. Recently, carbon-containing TiO2 nanoparticles [TiO2(C) NP] were found to be a visible-light-responsive photocatalyst (VLRP), which displayed significantly enhanced antibacterial properties under visible light illumination. However, whether TiO2(C) NPs exert antibacterial properties against Bacillus anthracis remains elusive. Here, we evaluated these VLRP NPs in the reduction of anthrax-induced pathogenesis. Bacteria-killing experiments indicated that a significantly higher proportion (40%-60%) of all tested Bacillus species, including B. subtilis, B. cereus, B. thuringiensis, and B. anthracis, were considerably eliminated by TiO2(C) NPs. Toxin inactivation analysis further suggested that the TiO2(C) NPs efficiently detoxify approximately 90% of tested anthrax lethal toxin, a major virulence factor of anthrax. Notably, macrophage clearance experiments further suggested that, even under suboptimal conditions without considerable bacterial killing, the TiO2(C) NP-mediated photocatalysis still exhibited antibacterial properties through the reduction of bacterial resistance against macrophage killing. Our results collectively suggested that TiO2(C) NP is a conceptually feasible anti-anthrax material, and the relevant technologies described herein may be useful in the development of new strategies against anthrax.

9.
Virulence ; 6(5): 466-75, 2015.
Article En | MEDLINE | ID: mdl-25906166

Mice treated with anthrax lethal toxin (LT) exhibit hemorrhage caused by unknown mechanisms. Moreover, LT treatment in mice induced liver damage. In this study, we hypothesized that a suppressed coagulation function may be associated with liver damage, because the liver is the major producing source of coagulation factors. The hepatic expression of coagulant factors and the survival rates were analyzed after cultured cells or mice were exposed to LT. In agreement with our hypothesis, LT induces cytotoxicity against hepatic cells in vitro. In addition, suppressed expression of coagulation factor VIII (FVIII) in the liver is associated with a prolonged plasma clotting time in LT-treated mice, suggesting a suppressive role of LT in coagulation. Accordingly, we further hypothesized that a loss-of-function approach involving treatments of an anticoagulant should exacerbate LT-induced abnormalities, whereas a gain-of-function approach involving injections of recombinant FVIII to complement the coagulation deficiency should ameliorate the pathogenesis. As expected, a sublethal dose of LT caused mortality in the mice that were non-lethally pretreated with an anticoagulant (warfarin). By contrast, treatments of recombinant FVIII reduced the mortality from a lethal dose of LT in mice. Our results indicated that LT-induced deficiency of FVIII is involved in LT-mediated pathogenesis. Using recombinant FVIII to correct the coagulant defect may enable developing a new strategy to treat anthrax.


Antigens, Bacterial/toxicity , Bacterial Toxins/toxicity , Hemophilia A/chemically induced , Liver/cytology , Liver/metabolism , Animals , Anticoagulants/pharmacology , Bacillus anthracis/pathogenicity , Blood Coagulation , Blood Coagulation Factors/genetics , Blood Coagulation Factors/metabolism , Cell Death , Cell Line, Tumor , Cells, Cultured , Factor VIII/administration & dosage , Factor VIII/metabolism , Hemophilia A/metabolism , Hep G2 Cells , Humans , Male , Mice , Recombinant Proteins/administration & dosage , Warfarin/pharmacology
10.
PLoS One ; 9(11): e111149, 2014.
Article En | MEDLINE | ID: mdl-25384016

Anthrax lethal toxin (LT), one of the primary virulence factors of Bacillus anthracis, causes anthrax-like symptoms and death in animals. Experiments have indicated that levels of erythrocytopenia and hypoxic stress are associated with disease severity after administering LT. In this study, the granulocyte colony-stimulating factor (G-CSF) was used as a therapeutic agent to ameliorate anthrax-LT- and spore-induced mortality in C57BL/6J mice. We demonstrated that G-CSF promoted the mobilization of mature erythrocytes to peripheral blood, resulting in a significantly faster recovery from erythrocytopenia. In addition, combined treatment using G-CSF and erythropoietin tended to ameliorate B. anthracis-spore-elicited mortality in mice. Although specific treatments against LT-mediated pathogenesis remain elusive, these results may be useful in developing feasible strategies to treat anthrax.


Antigens, Bacterial/toxicity , Bacterial Toxins/toxicity , Erythrocytes/drug effects , Erythropoietin/pharmacology , Granulocyte Colony-Stimulating Factor/pharmacology , Analysis of Variance , Animals , Antigens, Bacterial/poisoning , Bacterial Toxins/poisoning , Erythrocytes/physiology , Erythroid Precursor Cells , Erythropoiesis/drug effects , Erythropoiesis/physiology , Flow Cytometry , Granulocyte Colony-Stimulating Factor/therapeutic use , Kaplan-Meier Estimate , Mice , Mice, Inbred C57BL
11.
PLoS One ; 8(8): e71718, 2013.
Article En | MEDLINE | ID: mdl-23977125

Anthrax is a disease caused by the bacterium Bacillus anthracis, which results in high mortality in animals and humans. Although some of the mechanisms are already known such as asphyxia, extensive knowledge of molecular pathogenesis of this disease is deficient and remains to be further investigated. Lethal toxin (LT) is a major virulence factor of B. anthracis and a specific inhibitor/protease of mitogen-activated protein kinase kinases (MAPKKs). Anthrax LT causes lethality and induces certain anthrax-like symptoms, such as anemia and hypoxia, in experimental mice. Mitogen-activated protein kinases (MAPKs) are the downstream pathways of MAPKKs, and are important for erythropoiesis. This prompted us to hypothesize that anemia and hypoxia may in part be exacerbated by erythropoietic dysfunction. As revealed by colony-forming cell assays in this study, LT challenges significantly reduced mouse erythroid progenitor cells. In addition, in a proteolytic activity-dependent manner, LT suppressed cell survival and differentiation of cord blood CD34(+)-derived erythroblasts in vitro. Suppression of cell numbers and the percentage of erythroblasts in the bone marrow were detected in LT-challenged C57BL/6J mice. In contrast, erythropoiesis was provoked through treatments of erythropoietin, significantly ameliorating the anemia and reducing the mortality of LT-treated mice. These data suggested that suppressed erythropoiesis is part of the pathophysiology of LT-mediated intoxication. Because specific treatments to overcome LT-mediated pathogenesis are still lacking, these efforts may help the development of effective treatments against anthrax.


Anthrax/microbiology , Anthrax/pathology , Antigens, Bacterial/toxicity , Bacterial Toxins/toxicity , Disease Progression , Erythropoiesis/drug effects , Anemia/complications , Anemia/pathology , Animals , Anthrax/complications , Apoptosis/drug effects , Biocatalysis/drug effects , Cell Differentiation/drug effects , Colony-Forming Units Assay , Erythroid Cells/drug effects , Erythroid Cells/metabolism , Erythroid Cells/pathology , Erythropoietin/pharmacology , Hemolysis/drug effects , Humans , Male , Mice , Mice, Inbred C57BL , Proteolysis/drug effects , Survival Analysis
12.
Anal Chem ; 85(11): 5562-8, 2013 Jun 04.
Article En | MEDLINE | ID: mdl-23627570

Rapid identification of single and multiple infectious agents is vital in clinical settings and during biothreat attack. This study assesses the assay of single-stranded multiplex polymerase chain reaction (PCR) amplicons by suspension bead array (SSMP-SBA) for multiple pathogens identification in a single-tube reaction. A 15-plex assay for identification of 11 highly infectious pathogens was developed to evaluate the performance of SSMP-SBA. Pathogen-specific amplicons were obtained by sequential amplification of genomic DNAs using gene-specific primers tagged with artificial unique sequences and unique primers of which the reverse primer was modified by biotin and phosphorothioate. The SSMP products generated by T7 exonuclease-mediated DNA hydrolysis were hybridized to 15 sets of beads coupled with gene-specific and control oligonucleotide probes for pathogen identification and quantification by flow cytometry. This method was validated via assessment of 57 reference strains and one clinical bacterial isolate. All 11 pathogens can be detected by the 15-plex SSMP-SBA assay, and this design significantly enhanced the signal-to-noise ratio and improved the assay performance. This assay achieves similar sensitivity to our in-house real-time PCR system with the limit of detection equivalent to 5-100 genome copies and a linear dynamic range crossing three to five logs. In the validation assay, a 100% accuracy rate was achieved when the pathogens were among the target species. Notably, the species of pathogens were accurately identified from the samples with multiple infections. SSMP-SBA presents superior performance with multiplexing capability in a single-tube reaction and provides a new approach for detection and species identification of multiple pathogen infections.


Blood-Borne Pathogens/isolation & purification , Brucella/genetics , DNA, Bacterial/analysis , Multiplex Polymerase Chain Reaction/methods , Blood-Borne Pathogens/classification , Brucella/isolation & purification , Brucellosis/diagnosis , Brucellosis/genetics , DNA, Bacterial/genetics , Female , Humans , Middle Aged , Real-Time Polymerase Chain Reaction , Signal-To-Noise Ratio
13.
PLoS One ; 8(3): e59512, 2013.
Article En | MEDLINE | ID: mdl-23555687

Anthrax lethal toxin (LT) is a major virulence factor of Bacillus anthracis. LT challenge suppresses platelet counts and platelet function in mice, however, the mechanism responsible for thrombocytopenia remains unclear. LT inhibits cellular mitogen-activated protein kinases (MAPKs), which are vital pathways responsible for cell survival, differentiation, and maturation. One of the MAPKs, the MEK1/2-extracellular signal-regulated kinase pathway, is particularly important in megakaryopoiesis. This study evaluates the hypothesis that LT may suppress the progenitor cells of platelets, thereby inducing thrombocytopenic responses. Using cord blood-derived CD34(+) cells and mouse bone marrow mononuclear cells to perform in vitro differentiation, this work shows that LT suppresses megakaryopoiesis by reducing the survival of megakaryocytes. Thrombopoietin treatments can reduce thrombocytopenia, megakaryocytic suppression, and the quick onset of lethality in LT-challenged mice. These results suggest that megakaryocytic suppression is one of the mechanisms by which LT induces thrombocytopenia. These findings may provide new insights for developing feasible approaches against anthrax.


Antigens, Bacterial/toxicity , Bacterial Toxins/toxicity , Megakaryocytes/cytology , Megakaryocytes/drug effects , Animals , Antigens, CD34/metabolism , Cell Death/drug effects , Fetal Blood/cytology , Humans , Male , Megakaryocytes/metabolism , Mice , Stem Cells/cytology , Stem Cells/drug effects , Thrombocytopenia/chemically induced , Thrombocytopenia/drug therapy , Thrombopoietin/pharmacology , Thrombopoietin/therapeutic use
14.
J Biomed Sci ; 19: 98, 2012 Nov 21.
Article En | MEDLINE | ID: mdl-23170801

BACKGROUND: Lethal toxin (LT) is a major virulence factor of Bacillus anthracis. Sprague Dawley rats manifest pronounced lung edema and shock after LT treatments, resulting in high mortality. The heart failure that is induced by LT has been suggested to be a principal mechanism of lung edema and mortality in rodents. Since LT-induced death occurs more rapidly in rats than in mice, suggesting that other mechanisms in addition to the heart dysfunction may be contributed to the fast progression of LT-induced pathogenesis in rats. Coagulopathy may contribute to circulatory failure and lung injury. However, the effect of LT on coagulation-induced lung dysfunction is unclear. METHODS: To investigate the involvement of coagulopathy in LT-mediated pathogenesis, the mortality, lung histology and coagulant levels of LT-treated rats were examined. The effects of activated protein C (aPC) on LT-mediated pathogenesis were also evaluated. RESULTS: Fibrin depositions were detected in the lungs of LT-treated rats, indicating that coagulation was activated. Increased levels of plasma D-dimer and thrombomodulin, and the ameliorative effect of aPC further suggested that the activation of coagulation-fibrinolysis pathways plays a role in LT-mediated pathogenesis in rats. Reduced mortality was associated with decreased plasma levels of D-dimer and thrombomodulin following aPC treatments in rats with LT-mediated pathogenesis. CONCLUSIONS: These findings suggest that the activation of coagulation in lung tissue contributes to mortality in LT-mediated pathogenesis in rats. In addition, anticoagulant aPC may help to develop a feasible therapeutic strategy.


Anthrax , Bacillus anthracis , Heart Failure/chemically induced , Protein C , Animals , Anthrax/drug therapy , Anthrax/microbiology , Anthrax/physiopathology , Antigens, Bacterial/drug effects , Antigens, Bacterial/toxicity , Bacillus anthracis/drug effects , Bacillus anthracis/pathogenicity , Bacterial Toxins/antagonists & inhibitors , Bacterial Toxins/toxicity , Blood Coagulation/drug effects , Heart Failure/drug therapy , Heart Failure/metabolism , Heart Failure/physiopathology , Humans , Injections, Intraventricular , Lung/blood supply , Lung/drug effects , Lung/metabolism , Lung/physiopathology , Protein C/administration & dosage , Protein C/metabolism , Rats , Rats, Sprague-Dawley
15.
PLoS One ; 5(12): e14289, 2010 Dec 10.
Article En | MEDLINE | ID: mdl-21170330

BACKGROUND: Lethal toxin (LT), the major virulence factor produced by Bacillus anthracis, has been shown to suppress the immune system, which is beneficial to the establishment of B. anthracis infections. It has been suggested that the suppression of MEK/MAPK signaling pathways of leukocytes contributes to LT-mediated immunosuppressive effects. However, the involvement of MAPK independent pathways has not been clearly elucidated; nor has the crucial role played by LT in the early stages of infection. Determining whether LT exerts any pathological effects before being enriched to an MEK inhibitory level is an important next step in the furtherance of this field. METHODOLOGY/PRINCIPAL FINDINGS: Using a cell culture model, we determined that low doses of LT inhibited phagocytosis of macrophages, without influencing MAPK pathways. Consistent low doses of LT significantly suppressed bacterial clearance and enhanced the mortality of mice with bacteremia, without suppressing the MEK1 of splenic and peripheral blood mononuclear cells. CONCLUSION/SIGNIFICANCE: These results suggest that LT suppresses the phagocytes in a dose range lower than that required to suppress MEK1 in the early stages of infection.


Antigens, Bacterial/metabolism , Bacterial Toxins/metabolism , Macrophages/immunology , Macrophages/metabolism , Animals , Bacillus anthracis/immunology , Flow Cytometry/methods , Immunosuppressive Agents/metabolism , Leukocytes/cytology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/microbiology , MAP Kinase Kinase 1/metabolism , Macrophages/microbiology , Male , Mice , Mice, Inbred C57BL , Models, Biological , Phagocytosis , Sepsis/microbiology
16.
PLoS One ; 4(1): e4167, 2009.
Article En | MEDLINE | ID: mdl-19132100

BACKGROUND: Photocatalysis of titanium dioxide (TiO(2)) substrates is primarily induced by ultraviolet light irradiation. Anion-doped TiO(2) substrates were shown to exhibit photocatalytic activities under visible-light illumination, relative environmentally-friendly materials. Their anti-spore activity against Bacillus anthracis, however, remains to be investigated. We evaluated these visible-light activated photocatalysts on the reduction of anthrax spore-induced pathogenesis. METHODOLOGY/PRINCIPAL FINDINGS: Standard plating method was used to determine the inactivation of anthrax spore by visible light-induced photocatalysis. Mouse models were further employed to investigate the suppressive effects of the photocatalysis on anthrax toxin- and spore-mediated mortality. We found that anti-spore activities of visible light illuminated nitrogen- or carbon-doped titania thin films significantly reduced viability of anthrax spores. Even though the spore-killing efficiency is only approximately 25%, our data indicate that spores from photocatalyzed groups but not untreated groups have a less survival rate after macrophage clearance. In addition, the photocatalysis could directly inactivate lethal toxin, the major virulence factor of B. anthracis. In agreement with these results, we found that the photocatalyzed spores have tenfold less potency to induce mortality in mice. These data suggest that the photocatalysis might injury the spores through inactivating spore components. CONCLUSION/SIGNIFICANCE: Photocatalysis induced injuries of the spores might be more important than direct killing of spores to reduce pathogenicity in the host.


Bacillus anthracis/radiation effects , Bacterial Toxins/antagonists & inhibitors , Light , Photosensitizing Agents/pharmacology , Titanium/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Antigens, Bacterial/metabolism , Bacillus anthracis/pathogenicity , Bacillus subtilis/metabolism , Bacterial Toxins/metabolism , Mice , Mice, Inbred C57BL , Spores, Bacterial/pathogenicity , Spores, Bacterial/radiation effects
17.
J Biotechnol ; 126(3): 277-85, 2006 Nov 10.
Article En | MEDLINE | ID: mdl-16797097

Lethal toxin (LT) that composed by protective antigen and lethal factor (LF) is the major virulence factor of Bacillus anthracis. The treatments of LT in animals could reproduce most manifestations of B. anthracis infections that greatly improves our knowledge in LT-mediated pathogenesis and facilitates anthrax-related researches without having to directly contact the hazardous bacterium B. anthracis. The recombinant protein of LF (rLF), however, still lacks a simple purification method. Herein, we developed single-step nickel affinity purification of rLF with yield up to 3mg/l. By fusion to the leader sequence of outer membrane protein OmpA, rLF could easily be purified from the periplasm of Escherichia coli. To investigate whether the rLT is functional in our system, both wild type rLF and the catalytic mutant rLF that contains a single amino acid substitution at zinc-binding site (LF(E687A)), were subjected to macrophage cytotoxicity analysis. Our data showed that the rLT is fully functional, while the LF(E687A) fail to induce cell death of tested macrophage cells. These findings suggested that the purification protocol herein is a user-friendly method that allows researchers to obtain the functional rLF by single-step purification.


Antigens, Bacterial/isolation & purification , Bacillus anthracis/metabolism , Bacterial Toxins/isolation & purification , Escherichia coli/metabolism , Periplasm/chemistry , Protein Engineering/methods , Solid Phase Extraction/methods , Animals , Antigens, Bacterial/pharmacology , Bacillus anthracis/genetics , Bacterial Toxins/pharmacology , Cell Line , Cell Survival/drug effects , Escherichia coli/genetics , Macrophages/cytology , Macrophages/drug effects , Mice , Recombinant Proteins/isolation & purification
18.
J Infect Dis ; 192(8): 1465-74, 2005 Oct 15.
Article En | MEDLINE | ID: mdl-16170766

Anthrax lethal toxin (LT) is the major virulence factor produced by Bacillus anthracis, but the mechanism by which it induces high mortality remains unclear. We found that LT treatment could induce severe hemorrhage in mice and significantly suppress human whole-blood clotting and platelet aggregation in vitro. In addition, LT could inhibit agonist-induced platelet surface P-selectin expression, resulting in the inhibition of platelet-endothelial cell engagements. Data from Western blot analysis indicated that LT treatment resulted in the suppression of p42/44 and p38 mitogen-activated protein kinase pathways in platelets. Combined treatments with LT and antiplatelet agents such as aspirin and the RGD-containing disintegrin rhodostomin significantly increased mortality in mice. Our data suggest that platelets are a pathogenic target for anthrax LT.


Antigens, Bacterial/toxicity , Bacillus anthracis/chemistry , Bacterial Toxins/toxicity , Blood Platelets/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Blood Platelets/enzymology , Blood Platelets/metabolism , Mice , Virulence
19.
Cell Biol Int ; 27(2): 123-33, 2003.
Article En | MEDLINE | ID: mdl-12662969

Cell-adhesion and spread are important for cell survival. Although extensive studies have suggested several potential mechanisms of action, it is not yet clear how important cell-morphological change per se contributes to the cell-surviving signal. We employed a non-integrin-mediated cell-adhesion system to explore this question. BHK-Japanese encephalitis virus (JEV) cells (BHK21 cells that are persistently infected with JEV) express a large amount of JEV-envelope protein (JEV E) on their surfaces, and can attach and form pseudopodia on the anti-JEV E antibody-coated substrates. However, cells that adhered on the antibody substrate underwent a caspase-3-mediated apoptosis together with a down-regulation of mitogen-activated protein kinase activity within 20 h after adhesion, which indicates that viral-protein-mediated cell-adhesion and cell-spread are not sufficient for supporting cell survival. This provides a different perspective for the study of the relationships between the cell-morphological change and the cell-survival signal.


Antibodies, Monoclonal/pharmacology , Encephalitis Virus, Japanese/growth & development , Animals , Apoptosis/drug effects , Blotting, Western , Caspase 3 , Caspases/metabolism , Cell Adhesion/drug effects , Cell Division/drug effects , Cell Line , Cell Movement/drug effects , Cell Survival/drug effects , Clone Cells/cytology , Clone Cells/drug effects , Clone Cells/virology , Encephalitis Virus, Japanese/metabolism , Fibronectins/pharmacology , Flow Cytometry , Mitogen-Activated Protein Kinases/metabolism , Viral Envelope Proteins/immunology , Viral Envelope Proteins/metabolism , Viral Nonstructural Proteins/immunology , Viral Nonstructural Proteins/metabolism
20.
Curr Microbiol ; 44(2): 106-11, 2002 Feb.
Article En | MEDLINE | ID: mdl-11815854

Previous studies have shown that the Bacillus anthracis lethal toxin can induce both necrosis and apoptosis in mouse macrophage-like J774A.1 cells depending on both the toxin concentration and the phosphatase activity. In this study several protein kinase or phosphatase inhibitors were employed to evaluate the hypothesis that the lethal toxin induces cell death via protein phosphorylation processes. Pretreatment with a serine/threonine phosphatase inhibitor Calyculin A (300 nM) could inhibit about 78% of cell death induced by the lethal toxin, whereas inhibitors of kinases, such as H7, HA, Sphingosine, and Genestein, but other inhibitors of phosphatases, such as Okadaic acid, Tautomycin, and Cyclosporin A, did not. In addition, recent reports have demonstrated that the MEK1 protein may serve as a proteolytic target within its N-terminus for lethal factor cleavage. In this study, Calyculin A is shown to enhance the phosphorylation of the MEK1 protein. This prevents the cleavage of the MEK1 by lethal factor. These results suggest that a putative Calyculin A-sensitive protein phosphatase is involved in anthrax toxin induced cytotoxicity and that the blocking effect of Calyculin A on lethal factor cytotoxicity may be mediated through the MEK signaling pathway.


Antigens, Bacterial , Bacillus anthracis/enzymology , Bacterial Toxins/metabolism , Enzyme Inhibitors/pharmacology , MAP Kinase Kinase Kinase 1 , Oxazoles/pharmacology , Phosphoprotein Phosphatases/metabolism , Pyrans , Spiro Compounds , Animals , Antifungal Agents/pharmacology , Bacillus anthracis/metabolism , Bacillus anthracis/pathogenicity , Bacterial Toxins/toxicity , Blotting, Western , Cells, Cultured , Cyclosporine/pharmacology , Genistein/pharmacology , Marine Toxins , Mice , Okadaic Acid/pharmacology , Phosphoprotein Phosphatases/antagonists & inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Sphingosine/pharmacology
...