Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
1.
J Biomed Sci ; 31(1): 39, 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38637878

ABSTRACT

BACKGROUND: High levels of neutrophil extracellular trap (NET) formation or NETosis and autoantibodies are related to poor prognosis and disease severity of COVID-19 patients. Human angiotensin-converting enzyme 2 (ACE2) cross-reactive anti-severe acute respiratory syndrome coronavirus 2 spike protein receptor-binding domain (SARS-CoV-2 RBD) antibodies (CR Abs) have been reported as one of the sources of anti-ACE2 autoantibodies. However, the pathological implications of CR Abs in NET formation remain unknown. METHODS: In this study, we first assessed the presence of CR Abs in the sera of COVID-19 patients with different severity by serological analysis. Sera and purified IgG from CR Abs positive COVID-19 patients as well as a mouse monoclonal Ab (mAb 127) that can recognize both ACE2 and the RBD were tested for their influence on NETosis and the possible mechanisms involved were studied. RESULTS: An association between CR Abs levels and the severity of COVID-19 in 120 patients was found. The CR Abs-positive sera and IgG from severe COVID-19 patients and mAb 127 significantly activated human leukocytes and triggered NETosis, in the presence of RBD. This NETosis, triggered by the coexistence of CR Abs and RBD, activated thrombus-related cells but was abolished when the interaction between CR Abs and ACE2 or Fc receptors was disrupted. We also revealed that CR Abs-induced NETosis was suppressed in the presence of recombinant ACE2 or the Src family kinase inhibitor, dasatinib. Furthermore, we found that COVID-19 vaccination not only reduced COVID-19 severity but also prevented the production of CR Abs after SARS-CoV-2 infection. CONCLUSIONS: Our findings provide possible pathogenic effects of CR Abs in exacerbating COVID-19 by enhancing NETosis, highlighting ACE2 and dasatinib as potential treatments, and supporting the benefit of vaccination in reducing disease severity and CR Abs production in COVID-19 patients.


Subject(s)
COVID-19 , Humans , Animals , Mice , SARS-CoV-2 , Angiotensin-Converting Enzyme 2 , COVID-19 Vaccines , Dasatinib , Immunoglobulin G/metabolism , Autoantibodies/metabolism , Spike Glycoprotein, Coronavirus , Protein Binding
2.
Am J Pathol ; 194(7): 1346-1373, 2024 07.
Article in English | MEDLINE | ID: mdl-38631549

ABSTRACT

Because the mechanotransduction by stromal stiffness stimulates the rupture and repair of the nuclear envelope in pancreatic progenitor cells, accumulated genomic aberrations are under selection in the tumor microenvironment. Analysis of cell growth, micronuclei, and phosphorylated Ser-139 residue of the histone variant H2AX (γH2AX) foci linked to mechanotransduction pressure in vivo during serial orthotopic passages of mouse KrasLSL-G12D/+;Trp53flox/flox;Pdx1-Cre (KPC) cancer cells in the tumor and in migrating through the size-restricted 3-µm micropores. To search for pancreatic cancer cell-of-origin, analysis of single-cell data sets revealed that the extracellular matrix shaped an alternate route of acinar-ductal transdifferentiation of acinar cells into topoisomerase II α (TOP2A)-overexpressing cancer cells and derived subclusters with copy number amplifications in MYC-PTK2 (protein tyrosine kinase 2) locus and PIK3CA. High-PTK2 expression is associated with 171 differentially methylated CpG loci, 319 differentially expressed genes, and poor overall survival in The Cancer Genome Atlas-Pancreatic Adenocarcinoma cohort. Abolished RGD-integrin signaling by disintegrin KG blocked the PTK2 phosphorylation, increased cancer apoptosis, decreased vav guanine nucleotide exchange factor 1 (VAV1) expression, and prolonged overall survival in the KPC mice. Reduction of α-smooth muscle actin deposition in the CD248 knockout KPC mice remodeled the tissue stroma and down-regulated TOP2A expression in the epithelium. In summary, stromal stiffness induced the onset of cancer cells-of-origin by ectopic TOP2A expression, and the genomic amplification of MYC-PTK2 locus via alternative transdifferentiation of pancreatic progenitor cells is the vulnerability useful for disintegrin KG treatment.


Subject(s)
Chromosomal Instability , Disease Progression , Pancreatic Neoplasms , Animals , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Mice , Humans , Carcinoma in Situ/pathology , Carcinoma in Situ/genetics , Carcinoma in Situ/metabolism , Stromal Cells/metabolism , Stromal Cells/pathology , Tumor Microenvironment , Mechanotransduction, Cellular , Focal Adhesion Kinase 1
3.
Front Immunol ; 13: 941923, 2022.
Article in English | MEDLINE | ID: mdl-36045680

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has spread globally since December 2019. Several studies reported that SARS-CoV-2 infections may produce false-positive reactions in dengue virus (DENV) serology tests and vice versa. However, it remains unclear whether SARS-CoV-2 and DENV cross-reactive antibodies provide cross-protection against each disease or promote disease severity. In this study, we confirmed that antibodies against the SARS-CoV-2 spike protein and its receptor-binding domain (S1-RBD) were significantly increased in dengue patients compared to normal controls. In addition, anti-S1-RBD IgG purified from S1-RBD hyperimmune rabbit sera could cross-react with both DENV envelope protein (E) and nonstructural protein 1 (NS1). The potential epitopes of DENV E and NS1 recognized by these antibodies were identified by a phage-displayed random peptide library. In addition, DENV infection and DENV NS1-induced endothelial hyperpermeability in vitro were inhibited in the presence of anti-S1-RBD IgG. Passive transfer anti-S1-RBD IgG into mice also reduced prolonged bleeding time and decreased NS1 seral level in DENV-infected mice. Lastly, COVID-19 patients' sera showed neutralizing ability against dengue infection in vitro. Thus, our results suggest that the antigenic cross-reactivity between the SARS-CoV-2 S1-RBD and DENV can induce the production of anti-SARS-CoV-2 S1-RBD antibodies that cross-react with DENV which may hinder dengue pathogenesis.


Subject(s)
COVID-19 , Dengue Virus , Dengue , Animals , Antibodies, Viral , Humans , Immunoglobulin G , Mice , Rabbits , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Viral Nonstructural Proteins
4.
Front Immunol ; 13: 868724, 2022.
Article in English | MEDLINE | ID: mdl-35603169

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is an emerging virus responsible for the ongoing COVID-19 pandemic. SARS-CoV-2 binds to the human cell receptor angiotensin-converting enzyme 2 (ACE2) through its receptor-binding domain in the S1 subunit of the spike protein (S1-RBD). The serum levels of autoantibodies against ACE2 are significantly higher in patients with COVID-19 than in controls and are associated with disease severity. However, the mechanisms through which these anti-ACE2 antibodies are induced during SARS-CoV-2 infection are unclear. In this study, we confirmed the increase in antibodies against ACE2 in patients with COVID-19 and found a positive correlation between the amounts of antibodies against ACE2 and S1-RBD. Moreover, antibody binding to ACE2 was significantly decreased in the sera of some COVID-19 patients after preadsorption of the sera with S1-RBD, which indicated that antibodies against S1-RBD can cross-react with ACE2. To confirm this possibility, two monoclonal antibodies (mAbs 127 and 150) which could bind to both S1-RBD and ACE2 were isolated from S1-RBD-immunized mice. Measurement of the binding affinities by Biacore showed these two mAbs bind to ACE2 much weaker than binding to S1-RBD. Epitope mapping using synthetic overlapping peptides and hydrogen deuterium exchange mass spectrometry (HDX-MS) revealed that the amino acid residues P463, F464, E465, R466, D467 and E471 of S1-RBD are critical for the recognition by mAbs 127 and 150. In addition, Western blotting analysis showed that these mAbs could recognize ACE2 only in native but not denatured form, indicating the ACE2 epitopes recognized by these mAbs were conformation-dependent. The protein-protein interaction between ACE2 and the higher affinity mAb 127 was analyzed by HDX-MS and visualized by negative-stain transmission electron microscopy imaging combined with antigen-antibody docking. Together, our results suggest that ACE2-cross-reactive anti-S1-RBD antibodies can be induced during SARS-CoV-2 infection due to potential antigenic cross-reactivity between S1-RBD and its receptor ACE2.


Subject(s)
Angiotensin-Converting Enzyme 2 , COVID-19 , Animals , Antibodies, Monoclonal , Antibodies, Viral , Humans , Mice , Pandemics , SARS-CoV-2 , Spike Glycoprotein, Coronavirus
5.
Toxins (Basel) ; 12(11)2020 11 09.
Article in English | MEDLINE | ID: mdl-33182321

ABSTRACT

Echistatin (Ech) is a short disintegrin with a long 42NPHKGPAT C-terminal tail. We determined the 3-D structure of Ech by X-ray crystallography. Superimposition of the structures of chains A and B showed conformational differences in their RGD loops and C-termini. The chain A structure is consistent with our NMR analysis that the GPAT residues of the C-terminus cannot be observed due to high flexibility. The hydrogen bond patterns of the RGD loop and between the RGD loop and C-terminus in Ech were the same as those of the corresponding residues in medium disintegrins. The mutant with C-terminal HKGPAT truncation caused 6.4-, 7.0-, 11.7-, and 18.6-fold decreases in inhibiting integrins αvß3, αIIbß3, αvß5, and α5ß1. Mutagenesis of the C-terminus showed that the H44A mutant caused 2.5- and 4.4-fold increases in inhibiting αIIbß3 and α5ß1, and the K45A mutant caused a 2.6-fold decrease in inhibiting αIIbß3. We found that Ech inhibited VEGF-induced HUVEC proliferation with an IC50 value of 103.2 nM and inhibited the migration of A375, U373MG, and Panc-1 tumor cells with IC50 values of 1.5, 5.7, and 154.5 nM. These findings suggest that Ech is a potential anticancer agent, and its C-terminal region can be optimized to improve its anticancer activity.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Integrins/chemistry , Intercellular Signaling Peptides and Proteins/chemistry , Intercellular Signaling Peptides and Proteins/pharmacology , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Molecular Docking Simulation , Protein Conformation , Structure-Activity Relationship , Vascular Endothelial Growth Factor A
6.
Biochem Mol Biol Educ ; 48(6): 631-634, 2020 11.
Article in English | MEDLINE | ID: mdl-33075847

ABSTRACT

Science requires that we are always current with research, techniques, and tools but what are the best approaches for continuing education? The presenters in this session described a range of approaches used in universities, government bodies, and industry.


Subject(s)
Education, Continuing , Interdisciplinary Studies , Molecular Biology/education , Congresses as Topic , Humans
7.
Toxins (Basel) ; 12(7)2020 06 28.
Article in English | MEDLINE | ID: mdl-32605221

ABSTRACT

Polymer polyethylene glycol (PEG), or PEGylation of polypeptides improves protein drug stability by decrease degradation and reduces renal clearance. To produce a pharmaceutical disintegrin derivative, the N-terminal PEGylation technique was used to modify the disintegrin derivative [KGDRR]trimucrin for favorable safety and pharmacokinetic profiles and antithrombotic efficacy. We compared intact [KGDRR]trimucrin (RR) and PEGylated KGDRR (PEG-RR) by in vitro and in vivo systems for their antithrombotic activities. The activity of platelet aggregation inhibition and the bleeding tendency side effect were also investigated. PEG-RR exhibited optimal potency in inhibiting platelet aggregation of human/mouse platelet-rich plasma activated by collagen or ADP with a lower IC50 than the intact derivative RR. In the illumination-induced mesenteric venous thrombosis model, RR and PEG-RR efficaciously prevented occlusive thrombosis in a dose-dependent manner. In rotational thromboelastometry assay, there was no effect of PEG-RR in human whole blood coagulation even given at a higher concentration (30 µg/mL), while RR slightly prolonged clotting time. However, RR and PEG-RR were not associated with severe thrombocytopenia or bleeding in FcγRIIa-transgenic mice at equally efficacious antithrombotic dosages. We also found the in vivo half-life of PEGylation was longer than RR (RR: 15.65 h vs. PEG-RR: 20.45 h). In conclusion, injectable PEG-RR with prolonged half-life and decreased bleeding risk is a safer anti-thrombotic agent for long-acting treatment of thrombus diseases.


Subject(s)
Blood Coagulation/drug effects , Disintegrins/pharmacology , Fibrinolytic Agents/pharmacology , Peptides/pharmacology , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Polyethylene Glycols/chemistry , Thrombosis/prevention & control , Animals , Disease Models, Animal , Disintegrins/chemistry , Disintegrins/toxicity , Drug Compounding , Fibrinolytic Agents/chemistry , Fibrinolytic Agents/toxicity , Hemorrhage/chemically induced , Humans , Male , Mice , Mice, Inbred ICR , Mice, Transgenic , Peptides/chemistry , Peptides/toxicity , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Receptors, IgG/genetics , Receptors, IgG/metabolism , Thrombocytopenia/chemically induced , Thrombosis/blood , Thrombosis/etiology
8.
Clin Epigenetics ; 12(1): 87, 2020 06 17.
Article in English | MEDLINE | ID: mdl-32552862

ABSTRACT

BACKGROUND: Cancer subtype switching, which involves unclear cancer cell origin, cell fate decision, and transdifferentiation of cells within a confined tumor microenvironment, remains a major problem in pancreatic cancer (PDA). RESULTS: By analyzing PDA subtypes in The Cancer Genome Atlas, we identified that epigenetic silencing of apoptosis-associated tyrosine kinase (AATK) inversely was correlated with mRNA expression and was enriched in the quasi-mesenchymal cancer subtype. By comparing early mouse pancreatic lesions, the non-invasive regions showed AATK co-expression in cells with acinar-to-ductal metaplasia, nuclear VAV1 localization, and cell cycle suppression; but the invasive lesions conversely revealed diminished AATK expression in those with poorly differentiated histology, cytosolic VAV1 localization, and co-expression of p63 and HNF1α. Transiently activated AATK initiates acinar differentiation into a ductal cell fate to establish apical-basal polarization in acinar-to-ductal metaplasia. Silenced AATK and ectopically expressed p63 and HNF1α allow the proliferation of ductal PanINs in mice. CONCLUSION: Epigenetic silencing of AATK regulates the cellular transdifferentiation, proliferation, and cell cycle progression in converting PDA-subtypes.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Epigenesis, Genetic/genetics , Metaplasia/genetics , Pancreatic Neoplasms/genetics , Protein-Tyrosine Kinases/genetics , Aged , Animals , Cell Differentiation , DNA Methylation/genetics , Disease Models, Animal , Female , Gene Silencing , Hepatocyte Nuclear Factor 1-alpha/genetics , Humans , Metaplasia/diagnosis , Mice , Middle Aged , Pancreatic Neoplasms/pathology , Pregnancy , Proto-Oncogene Proteins c-vav/genetics , RNA, Messenger/genetics , Trans-Activators/genetics , Tumor Microenvironment/genetics
9.
Toxins (Basel) ; 12(1)2019 12 21.
Article in English | MEDLINE | ID: mdl-31877725

ABSTRACT

Life-threatening thrombocytopenia and bleeding, common side effects of clinically available αIIbß3 antagonists, are associated with the induction of ligand-induced integrin conformational changes and exposure of ligand-induced binding sites (LIBSs). To address this issue, we examined intrinsic mechanisms and structure-activity relationships of purified disintegrins, from Protobothrops flavoviridis venom (i.e., Trimeresurus flavoviridis), TFV-1 and TFV-3 with distinctly different pro-hemorrhagic tendencies. TFV-1 with a different αIIbß3 binding epitope from that of TFV-3 and chimeric 7 × 103 Fab, i.e., Abciximab, decelerates αIIbß3 ligation without causing a conformational change in αIIbß3, as determined with the LIBS antibody, AP5, and the mimetic, drug-dependent antibody (DDAb), AP2, an inhibitory monoclonal antibody raised against αIIbß3. Consistent with their different binding epitopes, a combination of TFV-1 and AP2 did not induce FcγRIIa-mediated activation of the ITAM-Syk-PLCγ2 pathway and platelet aggregation, in contrast to the clinical antithrombotics, abciximab, eptifibatide, and disintegrin TFV-3. Furthermore, TFV-1 selectively inhibits Gα13-mediated platelet aggregation without affecting talin-driven clot firmness, which is responsible for physiological hemostatic processes. At equally efficacious antithrombotic dosages, TFV-1 caused neither severe thrombocytopenia nor bleeding in FcγRIIa-transgenic mice. Likewise, it did not induce hypocoagulation in human whole blood in the rotational thromboelastometry (ROTEM) assay used in perioperative situations. In contrast, TFV-3 and eptifibatide exhibited all of these hemostatic effects. Thus, the αIIbß3 antagonist, TFV-1, efficaciously prevents arterial thrombosis without adversely affecting hemostasis.


Subject(s)
Disintegrins/pharmacology , Fibrinolytic Agents/adverse effects , Fibrinolytic Agents/pharmacology , Hemorrhage/chemically induced , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Snake Venoms/pharmacology , Abciximab/pharmacology , Animals , Binding Sites , Bleeding Time , Epitopes , Humans , Male , Mice , Mice, Inbred ICR , Platelet Aggregation , Platelet Aggregation Inhibitors/pharmacology , Structure-Activity Relationship , Trimeresurus
10.
PLoS One ; 12(4): e0175321, 2017.
Article in English | MEDLINE | ID: mdl-28399159

ABSTRACT

Rhodostomin (Rho) is a medium disintegrin containing a 48PRGDMP motif. We here showed that Rho proteins with P48A, M52W, and P53N mutations can selectively inhibit integrin αIIbß3. To study the roles of the RGD loop and C-terminal region in disintegrins, we expressed Rho 48PRGDMP and 48ARGDWN mutants in Pichia pastoris containing 65P, 65PR, 65PRYH, 65PRNGLYG, and 65PRNPWNG C-terminal sequences. The effect of C-terminal region on their integrin binding affinities was αIIbß3 > αvß3 ≥ α5ß1, and the 48ARGDWN-65PRNPWNG protein was the most selective integrin αIIbß3 mutant. The 48ARGDWN-65PRYH, 48ARGDWN-65PRNGLYG, and 48ARGDWN-65PRNPWNG mutants had similar activities in inhibiting platelet aggregation and the binding of fibrinogen to platelet. In contrast, 48ARGDWN-65PRYH and 48ARGDWN-65PRNGLYG exhibited 2.9- and 3.0-fold decreases in inhibiting cell adhesion in comparison with that of 48ARGDWN-65PRNPWNG. Based on the results of cell adhesion, platelet aggregation and the binding of fibrinogen to platelet inhibited by ARGDWN mutants, integrin αIIbß3 bound differently to immobilized and soluble fibrinogen. NMR structural analyses of 48ARGDWN-65PRYH, 48ARGDWN-65PRNGLYG, and 48ARGDWN-65PRNPWNG mutants demonstrated that their C-terminal regions interacted with the RGD loop. In particular, the W52 sidechain of 48ARGDWN interacted with H68 of 65PRYH, L69 of 65PRNGLYG, and N70 of 65PRNPWNG, respectively. The docking of the 48ARGDWN-65PRNPWNG mutant into integrin αIIbß3 showed that the N70 residue formed hydrogen bonds with the αIIb D159 residue, and the W69 residue formed cation-π interaction with the ß3 K125 residue. These results provide the first structural evidence that the interactions between the RGD loop and C-terminus of medium disintegrins depend on their amino acid sequences, resulting in their functional differences in the binding and selectivity of integrins.


Subject(s)
Integrins/physiology , Oligopeptides/pharmacology , Peptides/pharmacology , Blood Platelets/drug effects , Cells, Cultured , Humans , Mass Spectrometry , Mutation , Oligopeptides/chemistry , Peptides/chemistry , Peptides/genetics , Platelet Aggregation/drug effects , Protein Conformation , Sequence Homology, Amino Acid
11.
J Biomol NMR ; 66(1): 55-68, 2016 09.
Article in English | MEDLINE | ID: mdl-27613298

ABSTRACT

A tool for predicting the redox state and secondary structure of cysteine residues using multi-dimensional analyses of different combinations of nuclear magnetic resonance (NMR) chemical shifts has been developed. A data set of cysteine [Formula: see text], (13)C(α), (13)C(ß), (1)H(α), (1)H(N), and (15)N(H) chemical shifts was created, classified according to redox state and secondary structure, using a library of 540 re-referenced BioMagResBank (BMRB) entries. Multi-dimensional analyses of three, four, five, and six chemical shifts were used to derive rules for predicting the structural states of cysteine residues. The results from 60 BMRB entries containing 122 cysteines showed that four-dimensional analysis of the C(α), C(ß), H(α), and N(H) chemical shifts had the highest prediction accuracy of 100 and 95.9 % for the redox state and secondary structure, respectively. The prediction of secondary structure using 3D, 5D, and 6D analyses had the accuracy of ~90 %, suggesting that H(N) and [Formula: see text] chemical shifts may be noisy and made the discrimination worse. A web server (6DCSi) was established to enable users to submit NMR chemical shifts, either in BMRB or key-in formats, for prediction. 6DCSi displays predictions using sets of 3, 4, 5, and 6 chemical shifts, which shows their consistency and allows users to draw their own conclusions. This web-based tool can be used to rapidly obtain structural information regarding cysteine residues directly from experimental NMR data.


Subject(s)
Cysteine/chemistry , Nuclear Magnetic Resonance, Biomolecular , Oxidation-Reduction , Protein Structure, Secondary , Proteins/chemistry , Algorithms , Cluster Analysis , Models, Chemical , Nuclear Magnetic Resonance, Biomolecular/methods , Software , Web Browser
12.
Thromb Res ; 143: 3-10, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27161326

ABSTRACT

INTRODUCTION: The applicability of protein drugs is confined by protein degradation and rapid elimination. PEGylation of polypeptides improves protein stability by sterically obstructing the degradation by serum proteases and reduces renal clearance by the increased mass. EXPERIMENTAL APPROACH: We compared the antithrombotic activities of intact rhodostomin (Rn) and PEGylated rhodostomin (PRn) both in vitro and in vivo systems. In addition, the functional half-life in inhibiting platelet aggregation and the tendency in causing bleeding side effect were investigated. RESULTS: Rn and PRn both potently inhibited human and mouse platelet aggregation induced by collagen, thrombin or ADP in vitro with a similar IC50 around 60-100nM. Rotational thromboelastometry assay indicated that PRn was more effective than Rn in preventing clot formation in human whole blood. In platelet-rich plasma from mice injected with Rn or PRn, the inhibitory effects on collagen-induced platelet aggregation were also comparable, but Rn caused higher bleeding tendency. In ferric chloride-induced arterial thrombosis, Rn and PRn significantly prolonged occlusion time at high dosage (0.2µg/g). However, PRn obviously prolonged the occlusion time even given at a lower dosage (0.06µg/g). The functional half-life assay revealed that PEGylation prolonged the in vivo half-life of Rn. CONCLUSIONS: PRn exhibits higher antithrombotic potency and longer half-life in vivo as compared with native Rn on a molar basis. In addition, PRn exhibits a better safety profile at an efficacious antithrombotic dose in vivo. Therefore, PEGylation may be one of the ideal options in modifying disintegrin derivatives as the safe therapeutic agents for integrin-related diseases.


Subject(s)
Hemorrhage/chemically induced , Peptides/adverse effects , Peptides/therapeutic use , Platelet Aggregation Inhibitors/adverse effects , Platelet Aggregation Inhibitors/therapeutic use , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Thrombosis/drug therapy , Animals , Blood Platelets/drug effects , Disintegrins/adverse effects , Disintegrins/chemistry , Disintegrins/pharmacokinetics , Disintegrins/therapeutic use , Drug Stability , Half-Life , Hemorrhage/blood , Humans , Male , Mice , Mice, Inbred ICR , Peptides/chemistry , Peptides/pharmacokinetics , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/chemistry , Platelet Aggregation Inhibitors/pharmacokinetics , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Polyethylene Glycols/chemistry , Protein Stability , Thrombosis/blood
13.
Sci Rep ; 6: 26026, 2016 05 16.
Article in English | MEDLINE | ID: mdl-27181595

ABSTRACT

Clearance of apoptotic cells by macrophages plays an important role in maintaining tissue homeostasis. Previous study indicated that streptococcal pyrogenic exotoxin B (SPE B) reduces phagocytic activity in group A streptococcus (GAS) infection. Here, we demonstrate that SPE B causes an inhibitory effect on protein S-mediated phagocytosis. In the presence of SPE B, serum- and purified protein S-mediated phagocytosis of apoptotic cells were significantly inhibited. The binding abilities of protein S to apoptotic cells were decreased by treatment with SPE B. Bacterial culture supernatants from GAS NZ131 strain also caused a reduction of protein S binding to apoptotic cells, but speB mutant strain did not. SPE B directly cleaved protein S in vitro and in vivo, whereas a lower level of cleavage occurred in mice infected with a speB isogenic mutant strain. SPE B-mediated initial cleavage of protein S caused a disruption of phagocytosis, and also resulted in a loss of binding ability of protein S-associated C4b-binding protein to apoptotic cells. Taken together, these results suggest a novel pathogenic role of SPE B that initiates protein S degradation followed by the inhibition of apoptotic cell clearance by macrophages.


Subject(s)
Cysteine Endopeptidases/metabolism , Macrophages, Peritoneal/immunology , Protein S/metabolism , Streptococcal Infections/immunology , Streptococcus pyogenes/physiology , Animals , Apoptosis , Complement C4b-Binding Protein/metabolism , Cysteine Endopeptidases/genetics , Host-Pathogen Interactions , Humans , Macrophages, Peritoneal/microbiology , Mice , Mice, Inbred BALB C , Mutation/genetics , Phagocytosis , Protein Binding , Proteolysis , Streptococcal Infections/microbiology , THP-1 Cells
14.
Sci Rep ; 6: 23387, 2016 Mar 18.
Article in English | MEDLINE | ID: mdl-26987407

ABSTRACT

Bacterial infection-induced sepsis is the leading cause of septic inflammatory disease. Rhodostomin (Rn), a snake venom disintegrin, was previously reported to interact with the αVß3 integrin and the TLR4 on phagocyte in attenuating LPS-induced endotoxemia. In this report, we further evaluated the effects of Rn on TLR2-activated monocytes and its in vivo efficacy. Rn effectively suppressed the adhesion, migration, and cytokine release of Pam3CSK4-activated THP-1 cells. Rn specifically bound to integrin αVß3 of TLR2-activated THP-1. Integrin αV and Akt siRNA transfection both restrained Pam3CSK4-elicited cytokine release. Rn decreased the Pam3CSK4-induced phosporylation of MAPKs, degradation of IκB and activation of FAK, Akt, c-Src and Syk. The Pam3CSK4-induced translocation of MyD88, a central adaptor of TLR2, to the cell membrane was also inhibited by Rn treatment. In the polymicrobial inflammatory caecal ligation and puncture model, Rn significantly reduced pro-inflammatory cytokine and chemokine release, alleviated tissue injury and elevated survival rate in vivo. Taken together, in addition to inhibiting the activation of TLR4, Rn exhibits anti-inflammatory activity through antagonizing the activation of phagocytes and interrupting the crosstalk between αVß3 and TLR2-dependent signaling pathways.


Subject(s)
Integrin alphaVbeta3/metabolism , Monocytes/drug effects , Peptides/administration & dosage , Sepsis/drug therapy , Toll-Like Receptor 2/metabolism , Animals , Cell Adhesion/drug effects , Cell Line , Cell Movement/drug effects , Chemokines/metabolism , Cytokines/metabolism , Disease Models, Animal , Gene Expression Regulation/drug effects , Humans , Male , Mice , Monocytes/cytology , Peptides/pharmacology , Sepsis/metabolism , Signal Transduction/drug effects
15.
J Clin Microbiol ; 54(1): 157-62, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26560544

ABSTRACT

emm typing is the most widely used molecular typing method for the human pathogen Streptococcus pyogenes (group A streptococcus [GAS]). emm typing is based on a small variable region of the emm gene; however, the emm cluster typing system defines GAS types according to the nearly complete sequence of the emm gene. Therefore, emm cluster typing is considered to provide more information regarding the functional and structural properties of M proteins in different emm types of GAS. In the present study, 677 isolates collected between 1994 and 2008 in a hospital in southern Taiwan were analyzed by the emm cluster typing system. emm clusters A-C4, E1, E6, and A-C3 were the most prevalent emm cluster types and accounted for 67.4% of total isolates. emm clusters A-C4 and E1 were associated with noninvasive diseases, whereas E6 was significantly associated with both invasive and noninvasive manifestations. In addition, emm clusters D4, E2, and E3 were significantly associated with invasive manifestations. Furthermore, we found that the functional properties of M protein, including low fibrinogen-binding and high IgG-binding activities, were correlated significantly with invasive manifestations. In summary, the present study provides updated epidemiological information on GAS emm cluster types in southern Taiwan.


Subject(s)
Antigens, Bacterial/genetics , Bacterial Outer Membrane Proteins/genetics , Carrier Proteins/genetics , Genetic Variation , Molecular Typing/methods , Streptococcal Infections/epidemiology , Streptococcal Infections/microbiology , Streptococcus pyogenes/classification , Streptococcus pyogenes/isolation & purification , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Female , Fibrinogen/metabolism , Genotype , Hospitals , Humans , Immunoglobulin G/metabolism , Infant , Infant, Newborn , Male , Middle Aged , Molecular Epidemiology/methods , Prevalence , Protein Binding , Streptococcus pyogenes/genetics , Streptococcus pyogenes/pathogenicity , Taiwan/epidemiology , Virulence , Young Adult
16.
PLoS One ; 10(12): e0145223, 2015.
Article in English | MEDLINE | ID: mdl-26710228

ABSTRACT

Clustered regularly interspaced short palindromic repeats (CRISPR) are the bacterial adaptive immune system against foreign nucleic acids. Given the variable nature of CRISPR, it could be a good marker for molecular epidemiology. Group A streptococcus is one of the major human pathogens. It has two CRISPR loci, including CRISPR01 and CRISPR02. The aim of this study was to analyze the distribution of CRISPR-associated gene cassettes (cas) and CRISPR arrays in highly prevalent emm types. The cas cassette and CRISPR array in two CRISPR loci were analyzed in a total of 332 strains, including emm1, emm3, emm4, emm12, and emm28 strains. The CRISPR type was defined by the spacer content of each CRISPR array. All strains had at least one cas cassette or CRISPR array. More than 90% of the spacers were found in one emm type, specifically. Comparing the consistency between emm and CRISPR types by Simpson's index of diversity and the adjusted Wallace coefficient, CRISPR01 type was concordant to emm type, and CRISPR02 showed unidirectional congruence to emm type, suggesting that at least for the majority of isolates causing infection in high income countries, the emm type can be inferred from CRISPR analysis, which can further discriminate isolates sharing the same emm type.


Subject(s)
Antigens, Bacterial/genetics , Bacterial Outer Membrane Proteins/genetics , Carrier Proteins/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , DNA, Intergenic/genetics , Molecular Typing/methods , Streptococcus pyogenes/genetics , Humans , Sequence Analysis, DNA
17.
Anesthesiology ; 122(3): 666-76, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25383568

ABSTRACT

BACKGROUND: The pivotal role of glial activation and up-regulated inflammatory mediators in the opioid tolerance has been confirmed in rodents but not yet in humans. Here, the authors investigated the intraspinal cytokine and chemokine profiles of opioid-tolerant cancer patients; and to determine if up-regulated chemokines could modify opioid tolerance in rats. METHODS: Cerebrospinal fluid samples from opioid-tolerant cancer patients and opioid-naive subjects were compared. The cerebrospinal fluid levels of tumor necrosis factor-alpha, CXCL1, CXCL10, CCL2, and CX3CL1 were assayed. The rat tail flick test was utilized to assess the effects of intrathecal CXCL1 on morphine-induced acute antinociception and analgesic tolerance. RESULTS: CXCL1 level in cerebrospinal fluid was significantly up-regulated in the opioid-tolerant group (n = 30, 18.8 pg/ml vs. 13.2 pg/ml, P = 0.02) and was positively correlated (r = 0.49, P < 0.01) with opioid dosage. In rat experiment, after induction of tolerance by morphine infusion, the spinal cord CXCL1 messenger RNA was up-regulated to 32.5 ± 11.9-fold. Although CXCL1 infusion alone did not affect baseline tail-flick latency, the analgesic efficacy of a single intraperitoneal injection of morphine dropped significantly on day 1 to day 3 after intrathecal infusion of CXCL1. After establishing tolerance by intrathecal continuous infusion of morphine, its development was accelerated by coadministration of CXCL1 and attenuated by coadministration of CXCL1-neutralizing antibody or CXCR2 antagonist. CONCLUSIONS: CXCL1 is up-regulated in both opioid-tolerant patients and rodents. The onset and extent of opioid tolerance was affected by antagonizing intrathecal CXCL1/CXCR2 signaling. Therefore, the CXCL1/CXCR2 signal pathway may be a novel target for the treatment of opioid tolerance.


Subject(s)
Analgesics, Opioid/administration & dosage , Chemokine CXCL1/cerebrospinal fluid , Drug Tolerance/physiology , Pain Measurement/drug effects , Translational Research, Biomedical/methods , Adult , Aged , Animals , Female , Humans , Injections, Spinal , Male , Middle Aged , Pain Measurement/methods , Rats , Rats, Sprague-Dawley
18.
J Biomol NMR ; 59(3): 175-84, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24838372

ABSTRACT

A method for predicting type I and II ß-turns using nuclear magnetic resonance (NMR) chemical shifts is proposed. Isolated ß-turn chemical-shift data were collected from 1,798 protein chains. One-dimensional statistical analyses on chemical-shift data of three classes ß-turn (type I, II, and VIII) showed different distributions at four positions, (i) to (i + 3). Considering the central two residues of type I ß-turns, the mean values of Cο, Cα, H(N), and N(H) chemical shifts were generally (i + 1) > (i + 2). The mean values of Cß and Hα chemical shifts were (i + 1) < (i + 2). The distributions of the central two residues in type II and VIII ß-turns were also distinguishable by trends of chemical shift values. Two-dimensional cluster analyses on chemical-shift data show positional distributions more clearly. Based on these propensities of chemical shift classified as a function of position, rules were derived using scoring matrices for four consecutive residues to predict type I and II ß-turns. The proposed method achieves an overall prediction accuracy of 83.2 and 84.2% with the Matthews correlation coefficient values of 0.317 and 0.632 for type I and II ß-turns, indicating that its higher accuracy for type II turn prediction. The results show that it is feasible to use NMR chemical shifts to predict the ß-turn types in proteins. The proposed method can be incorporated into other chemical-shift based protein secondary structure prediction methods.


Subject(s)
Magnetic Resonance Spectroscopy/methods , Protein Structure, Secondary , Proteins/chemistry , Amino Acid Sequence , Amino Acids/chemistry , Cluster Analysis , Models, Molecular , Molecular Sequence Data
19.
PLoS One ; 9(2): e89027, 2014.
Article in English | MEDLINE | ID: mdl-24586487

ABSTRACT

Group A streptococcus (GAS, Streptococcus pyogenes) is a strict human pathogen that causes severe, invasive diseases. GAS does not produce catalase, but has an ability to resist killing by reactive oxygen species (ROS) through novel mechanisms. The peroxide response regulator (PerR), a member of ferric uptake regulator (Fur) family, plays a key role for GAS to cope with oxidative stress by regulating the expression of multiple genes. Our previous studies have found that expression of an iron-binding protein, Dpr, is under the direct control of PerR. To elucidate the molecular interactions of PerR with its cognate promoter, we have carried out structural studies on PerR and PerR-DNA complex. By combining crystallography and small-angle X-ray scattering (SAXS), we confirmed that the determined PerR crystal structure reflects its conformation in solution. Through mutagenesis and biochemical analysis, we have identified DNA-binding residues suggesting that PerR binds to the dpr promoter at the per box through a winged-helix motif. Furthermore, we have performed SAXS analysis and resolved the molecular architecture of PerR-DNA complex, in which two 30 bp DNA fragments wrap around two PerR homodimers by interacting with the adjacent positively-charged winged-helix motifs. Overall, we provide structural insights into molecular recognition of DNA by PerR and define the hollow structural arrangement of PerR-30bpDNA complex, which displays a unique topology distinct from currently proposed DNA-binding models for Fur family regulators.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , DNA, Bacterial/metabolism , Gene Expression Regulation, Bacterial/physiology , Models, Molecular , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Streptococcus pyogenes/metabolism , Crystallography , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Bacterial/genetics , Mutagenesis , Protein Conformation , Reactive Oxygen Species/metabolism , Scattering, Small Angle
20.
Pathog Dis ; 70(2): 194-201, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24339221

ABSTRACT

Hyaluronic acid capsule is one of the most important virulence factors of group A Streptococcus (GAS). Over-production of capsule has been thought to enhance GAS virulence during infections. However, although the increased of capsule expression associates with increased bacterial virulence and invasive ability, over-production of capsule has not often been observed among clinical isolates. In the present study, we identified two mucoid emm12 type isolates that can convert to the hypermucoid morphology under both in vitro and in vivo conditions. Consistent with previous studies, hypermucoid variants are more invasive in the mouse air-pouch infection model. However, one of the hypermucoid variants showed a growth-defective phenotype in regular broth culture conditions and is significantly more susceptible to various DNA-damaging treatments when compared with the mucoid variant. These properties of the hypermucoid variant may be adverse factors inhibiting its adaptation to the host environment during infections.


Subject(s)
Mutagens/toxicity , Polysaccharides, Bacterial/metabolism , Streptococcus pyogenes/drug effects , Streptococcus pyogenes/growth & development , Virulence Factors/metabolism , Animals , DNA Damage , DNA, Bacterial/drug effects , Disease Models, Animal , Mice, Inbred BALB C , Microbial Viability , Soft Tissue Infections/microbiology , Soft Tissue Infections/pathology , Streptococcal Infections/microbiology , Streptococcal Infections/pathology , Streptococcus pyogenes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL