Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Sci Rep ; 10(1): 3764, 2020 02 28.
Article in English | MEDLINE | ID: mdl-32111878

ABSTRACT

A critical limitation of Salmonella typhimurium (S. typhimurium) as an anti-cancer agent is the loss of their invasive or replicative activities, which results in no or less delivery of anti-cancer agents inside cancer cells in cancer therapy. Here we developed an oxytolerant attenuated Salmonella strain (KST0650) from the parental KST0649 (ΔptsIΔcrr) strain using radiation mutation technology (RMT). The oxytolerant KST0650 strain possessed 20-times higher replication activity in CT26 cancer cells and was less virulent than KST0649. Furthermore, KST0650 migrated effectively into tumor tissues in mice. KST0650 was further equipped with a plasmid harboring a spliced form of the intracellular pro-apoptotic protein sATF6, and the expression of sATF6 was controlled by the radiation-inducible recN promoter. The new strain was named as KST0652, in which sATF6 protein expression was induced in response to radiation in a dose-dependent manner. This strain was effectively delivered inside cancer cells and tumor tissues via the Salmonella type III secretion system (T3SS). In addition, combination treatment with KST0652 and radiation showed a synergistic anti-tumor effect in murine tumor model with complete inhibition of tumor growth and protection against death. In conclusion, we showed that RMT can be used to effectively develop an anti-tumor Salmonella strain for delivering anti-cancer agents inside tumors.


Subject(s)
Activating Transcription Factor 6 , Cancer Vaccines , Mutation , Neoplasm Proteins , Neoplasms, Experimental , Salmonella typhimurium , Type III Secretion Systems , Activating Transcription Factor 6/biosynthesis , Activating Transcription Factor 6/genetics , Animals , Cancer Vaccines/genetics , Cancer Vaccines/metabolism , Cell Line, Tumor , Male , Mice , Mice, Inbred BALB C , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/microbiology , Neoplasms, Experimental/therapy , Salmonella typhimurium/genetics , Salmonella typhimurium/metabolism , Type III Secretion Systems/genetics , Type III Secretion Systems/metabolism
2.
J Immunol ; 203(2): 429-440, 2019 07 15.
Article in English | MEDLINE | ID: mdl-31167774

ABSTRACT

Foot-and-mouth disease virus (FMDV) is highly infectious and causes a major plague in animal farming. Unfolded protein response is one of the major cellular responses to pathogenic infections, which performs a crucial role in cell survival, apoptosis, and antiviral innate immune response. In this study, we showed that FMDV infection activated two unfolded protein response branches (PERK-eIF2α and ATF6 signaling) in both baby hamster kidney cells (BHK-21) and porcine kidney (PK-15) cells, whereas it suppressed the IRE1α-XBP1 signaling by decreasing IRE1α level. Further study revealed IRE1α signaling as an important antiviral innate immune mechanism against FMDV. Sec62, the transport protein, was greatly decreased at the late stages of FMDV infection. By overexpression and knockdown study, we also found that the expression of Sec62 was positively involved in the levels of IRE1α and RIG-I and subsequent activation of downstream antiviral signaling pathways in FMDV-infected PK-15 cells. Taken together, our study demonstrates that Sec62 is an important antiviral factor that upregulates IRE1α-RIG-I-dependent antiviral innate immune responses, and FMDV evades antiviral host defense mechanism by downregulating Sec62-IRE1α/RIG-I.


Subject(s)
Antiviral Agents/immunology , Cell Proliferation/physiology , Foot-and-Mouth Disease Virus/immunology , Signal Transduction/immunology , Viral Proteins/immunology , Virus Replication/immunology , Animals , Cell Line , Cricetinae , Endoribonucleases , Immunity, Innate/immunology , Membrane Transport Proteins/immunology , Protein Serine-Threonine Kinases/immunology , Receptors, Cell Surface/immunology , Swine , Unfolded Protein Response/immunology
3.
Hum Vaccin Immunother ; 14(1): 67-73, 2018 01 02.
Article in English | MEDLINE | ID: mdl-28933634

ABSTRACT

Group B Streptococcus (GBS) is a leading cause of sepsis in infants as well as chorioamnionitis in pregnant women. Opsonophagocytic killing assays (OPAs) are an essential technique in vaccine studies of encapsulated bacteria for estimating serotype-specific functional antibody levels in vitro. Here, we developed a three-fold multiplexed OPA (MOPA) to enable practical, large-scale assessment of GBS vaccine immunogenicity, including against serotypes Ia, III, and V. First, three target bacteria strains resistant to streptomycin, spectinomycin, or kanamycin were generated by natural selection through exposure to increasing antibiotic concentrations. Since a high level of nonspecific killing (NSK) of serotype V was observed in a 12.5% baby rabbit complement (BRC) solution, the BRC concentration was optimized. The final GBS-MOPA BRC concentration was 9%, which resulted in less than 20% NSK. The specificity was measured by preabsorbing serum with inactivated GBS. The opsonic index (OI) of preabsorbed serum with the homologous serotype GBS was significantly reduced in all three serotypes tested. The accuracy of the MOPA was compared with that of a single OPA (SOPA) with 35 serum samples. The OIs of the MOPA correlated well with those of the SOPA, and the r2 values were higher than 0.950 for all three serotypes. The precision of the MOPA assay was assessed in five independent experiments with five serum samples. The inter-assay precision of the GBS-MOPA was 12.5% of the average coefficient of variation. This is the first report to develop and standardize a GBS-MOPA, which will be useful for GBS vaccine development and evaluation.


Subject(s)
Antibodies, Bacterial/blood , Phagocytosis/immunology , Streptococcal Infections/prevention & control , Streptococcal Vaccines/immunology , Streptococcus agalactiae/immunology , Adult , Antibodies, Bacterial/immunology , Complement System Proteins/immunology , Drug Development/methods , Drug Development/standards , Healthy Volunteers , Humans , Immunoassay/methods , Immunoassay/standards , Immunogenicity, Vaccine , Opsonin Proteins/blood , Opsonin Proteins/immunology , Streptococcus agalactiae/pathogenicity , Young Adult
4.
J Infect Dis ; 217(1): 93-102, 2017 12 27.
Article in English | MEDLINE | ID: mdl-29106586

ABSTRACT

Streptococcus agalactiae (group B streptococcus [GBS]) is a leading cause of invasive diseases in neonates and severe infections in elderly individuals. GBS serine-rich repeat glycoprotein 1 (Srr1) acts as a critical virulence factor by facilitating GBS invasion into the central nervous system through interaction with the fibrinogen Aα chain. This study revealed that srr1 is highly conserved, with 86.7% of GBS clinical isolates expressing the protein. Vaccination of mice with different Srr1 truncated peptides revealed that only Srr1 truncates containing the latch domain protected against GBS meningitis. Furthermore, the latch peptide alone was immunogenic and elicited protective antibodies, which efficiently enhanced antibody-mediated opsonophagocytic killing of GBS by HL60 cells and provided heterogeneous protection against 4 different GBS serogroups. Taken together, these findings indicated that the latch domain of Srr1 may constitute an effective peptide vaccine candidate for GBS.


Subject(s)
Cross Protection , Immunity, Heterologous , Meningitis, Bacterial/prevention & control , Streptococcal Infections/prevention & control , Streptococcal Vaccines/immunology , Streptococcus agalactiae/immunology , Animals , Antibodies, Bacterial/blood , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Blood Bactericidal Activity , Disease Models, Animal , Male , Meningitis, Bacterial/immunology , Meningitis, Bacterial/microbiology , Mice , Opsonin Proteins/blood , Phagocytosis , Streptococcal Infections/immunology , Streptococcal Infections/microbiology , Streptococcal Vaccines/administration & dosage , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology
5.
Virulence ; 8(6): 875-890, 2017 08 18.
Article in English | MEDLINE | ID: mdl-28059611

ABSTRACT

The pneumococcal capsule is indispensable for pathogenesis in systemic infections; however, many pneumococcal diseases, including conjunctivitis, otitis media, and some systemic infections in immunocompromised patients, are caused by nonencapsulated Streptococcus pneumoniae (NESp). Null capsule clade 1 (NCC1), found in group 2 NESp, expresses pneumococcal surface protein K (PspK) and is becoming prevalent among pneumococcal organisms owing to the widespread use of pneumococcal conjugate vaccines. Despite its clinical importance, the molecular mechanisms underlying the prevalence of NCC1 have not been fully elucidated. Here, we investigated the role of the R3 domain of PspK in the epithelial cell adherence of NCC1. We found that the R3 domain of PspK mediated NCC1 adherence via its direct interaction with the epithelial surface protein annexin A2. Additionally, neutralization with purified recombinant PspK-R3 or rabbit anti-UD:R3 IgG inhibited binding of NESp to lung epithelial cells in vitro. Immunization with the 'repeat' domain of PspK-R3 or PspK-UD:R3 effectively elicited mucosal and systemic immune responses against PspK-R3 and provided protection against nasopharyngeal, lung, and middle ear colonization of NESp in mice. Additionally, we found that rabbit anti-UD:R3 IgG bound to PspC-R1 of the encapsulated TIGR4 strain and that UD:R3 immunization provided protection against nasopharyngeal and lung colonization of TIGR4 and deaths by TIGR4 and D39 in mice. Further studies using 68 pneumococcal clinical isolates showed that 79% of clinical isolates showed cross-reactivity to rabbit anti-UD:R3 IgG. About 87% of serotypes in the 13-valent pneumococcal conjugate vaccine (PCV) and 68% of non-vaccine serotypes were positive for cross-reactivity with rabbit anti-UD:R3 IgG. Thus, the R3 domain of PspK may be an effective vaccine candidate for both NESp and encapsulated Sp.


Subject(s)
Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/immunology , Epithelial Cells/microbiology , Pneumococcal Vaccines , Streptococcus pneumoniae/immunology , A549 Cells , Animals , Annexin A2/genetics , Annexin A2/metabolism , Antigens, Bacterial , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Epithelial Cells/physiology , Humans , Immunization , Immunoglobulin G/metabolism , Mice , Nasopharynx/microbiology , Pneumococcal Infections/immunology , Pneumococcal Infections/microbiology , Pneumococcal Vaccines/genetics , Pneumococcal Vaccines/immunology , Protein Domains , Rabbits , Serogroup , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/pathogenicity , Vaccines, Conjugate
6.
Mol Med Rep ; 12(5): 6483-90, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26330195

ABSTRACT

Methicillin­resistant Staphylococcus aureus (MRSA) is difficult to treat using available antibiotic agents. Honeybee venom has been widely used as an oriental treatment for several inflammatory diseases and bacterial infections. The venom contains predominantly biologically active compounds, however, the therapeutic effects of such materials when used to treat MRSA infections have not been investigated extensively. The present study evaluated bee venom and its principal active component, melittin, in terms of their antibacterial activities and in vivo protection against MRSA infections. In vitro, bee venom and melittin exhibited comparable levels of antibacterial activity, which was more marked against MRSA strains, compared with other Gram­positive bacteria. When MRSA­infected mice were treated with bee venom or melittin, only the latter animals were successfully rescued from MRSA­ induced bacteraemia or exhibited recovery from MRSA­infected skin wounds. Together, the data of the present study demonstrated for the first time, to the best of our knowledge, that melittin may be used as a promising antimicrobial agent to enhance the healing of MRSA­induced wounds.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Bee Venoms/chemistry , Melitten/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Staphylococcal Infections/drug therapy , Animals , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/isolation & purification , Antimicrobial Cationic Peptides/chemical synthesis , Antimicrobial Cationic Peptides/isolation & purification , Bees/chemistry , Bees/physiology , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans , MCF-7 Cells , Male , Melitten/chemical synthesis , Melitten/isolation & purification , Methicillin-Resistant Staphylococcus aureus/growth & development , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Mice , Microbial Sensitivity Tests , Staphylococcal Infections/microbiology , Staphylococcal Infections/mortality , Streptococcal Infections/microbiology , Streptococcus/drug effects , Streptococcus/growth & development , Streptococcus/isolation & purification , Survival Analysis
7.
Sci Rep ; 3: 3398, 2013 Dec 02.
Article in English | MEDLINE | ID: mdl-24292328

ABSTRACT

The anti-apoptotic protein Bax inhibitor-1 (BI-1) is a regulator of apoptosis linked to endoplasmic reticulum (ER) stress. It has been hypothesized that BI-1 protects against neuron degenerative diseases. In this study, BI-1⁻/⁻ mice showed increased vulnerability to chronic mild stress accompanied by alterations in the size and morphology of the hippocampi, enhanced ROS accumulation and an ER stress response compared with BI-1⁺/⁺ mice. BI-1⁻/⁻ mice exposed to chronic mild stress showed significant activation of monoamine oxidase A (MAO-A), but not MAO-B, compared with BI-1⁺/⁺ mice. To examine the involvement of BI-1 in the Ca²âº-sensitive MAO activity, thapsigargin-induced Ca²âº release and MAO activity were analyzed in neuronal cells overexpressing BI-1. The in vitro study showed that BI-1 regulates Ca²âº release and related MAO-A activity. This study indicates an endogenous protective role of BI-1 under conditions of chronic mild stress that is primarily mediated through Ca²âº-associated MAO-A regulation.


Subject(s)
Endoplasmic Reticulum Stress/drug effects , Membrane Proteins/metabolism , Monoamine Oxidase Inhibitors/pharmacology , Monoamine Oxidase/metabolism , Animals , Calcium/metabolism , Cell Line , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Female , Hippocampus/drug effects , Hippocampus/metabolism , Mice , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Thapsigargin/pharmacology
8.
Int J Endocrinol ; 2013: 751854, 2013.
Article in English | MEDLINE | ID: mdl-24027582

ABSTRACT

Eucommia ulmoides Oliv. (EU) has been used for treatment of liver diseases. The protective effects of Eucommia Ulmoides Oliv. cortex extracts (EUCE) on the carbon tetrachloride- (CCl4-) induced hepatic lipid accumulation were examined in this study. Rats were orally treated with EUCE in different doses prior to an intraperitoneal injection of 1 mg/kg CCl4. Acute injection of CCl4 decreased plasma triglyceride but increased hepatic triglyceride and cholesterol as compared to control rats. On the other hand, the pretreatment with EUCE diminished these effects at a dose-dependent manner. CCl4 treatment decreased glutathione (GSH) and increased malondialdehyde (MDA) accompanied by activated P450 2E1. The pretreatment with EUCE significantly improved these deleterious effects of CCl4. CCl4 treatment increased P450 2E1 activation and ApoB accumulation. Pretreatment with EUCE reversed these effects. ER stress response was significantly increased by CCl4, which was inhibited by EUCE. One of the possible ER stress regulatory mechanisms, lysosomal activity, was examined. CCl4 reduced lysosomal enzymes that were reversed with the EUCE. The results indicate that oral pretreatment with EUCE may protect liver against CCl4-induced hepatic lipid accumulation. ER stress and its related ROS regulation are suggested as a possible mechanism in the antidyslipidemic effect of EUCE.

9.
Contrast Media Mol Imaging ; 8(1): 20-6, 2013.
Article in English | MEDLINE | ID: mdl-23109389

ABSTRACT

This study was undertaken to investigate the effect of a magnetic resonance imaging (MRI) contrast agent, superparamagnetic iron oxide nanoparticle (SPIO), on signal transduction by glucose and insulin in pancreatic ß-cells. INS-1 cells were labeled in culture medium containing clinically approved SPIO for 24 h. Labeled and unlabeled cells were stimulated with glucose (25 mM) or insulin (0.1-1 µM) for 12 h. The phosphorylation of extracellular signal-regulated kinase1/2 (ERK1/2) and protein kinase B (AKT) and intracellular insulin protein levels were assessed by Western blotting. After labeling with increasing amounts of SPIO, cytotoxicity was not observed, yet the intracellular iron concentration increased in a dose-dependent manner. SPIO labeling (200 µg Fe ml(-1)) induced a significant increase in ERK1/2 and AKT phosphorylation (labeled vs unlabeled, p < 0.05), but significantly reduced the glucose-stimulated phosphorylation of ERK1/2 and AKT and insulin-stimulated phosphorylation of AKT (labeled vs unlabeled, p < 0.05). The level of intracellular insulin protein was found to be lower in labeled cells than unlabeled cells (labeled vs unlabeled, p < 0.05). This study demonstrates that SPIO labeling alters some fundamental functional variables, at least in INS-1 cells, through modulation of the glucose- or insulin-induced activation of ERK1/2 and AKT, which leads to insulin biosynthesis.


Subject(s)
Contrast Media/adverse effects , Ferric Compounds/adverse effects , Glucose/metabolism , Insulin-Secreting Cells/metabolism , Insulin/biosynthesis , MAP Kinase Signaling System/drug effects , Magnetite Nanoparticles/adverse effects , Mitogen-Activated Protein Kinase 3/metabolism , Animals , Cell Line, Tumor , Contrast Media/pharmacology , Ferric Compounds/pharmacology , Insulin-Secreting Cells/pathology , Magnetic Resonance Imaging/methods , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt , Rats
10.
Proc Natl Acad Sci U S A ; 108(7): 2662-7, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21282616

ABSTRACT

For ultrasensitive magnetic resonance imaging (MRI), magnetic nanoparticles with extremely high r2 relaxivity are strongly desired. Magnetosome-like nanoparticles were prepared by coating polyethylene glycol-phospholipid (PEG-phospholipid) onto ferrimagnetic iron oxide nanocubes (FIONs). FIONs exhibited a very high relaxivity (r2) of 324 mM(-1) s(-1), allowing efficient labeling of various kinds of cells. The magnetic resonance (MR) imaging of single cells labeled with FIONs is demonstrated not only in vitro but also in vivo. Pancreatic islet grafts and their rejection could be imaged using FIONs on a 1.5 T clinical MRI scanner. The strong contrast effect of FIONs enabled MR imaging of transplanted islets in small rodents as well as in large animals. Therefore, we expect that MR imaging of pancreatic islet grafts using FIONs has the potentials for clinical applications. Furthermore, FIONs will enable highly sensitive noninvasive assessment after cell transplantation.


Subject(s)
Islets of Langerhans Transplantation/pathology , Islets of Langerhans/ultrastructure , Magnetic Resonance Imaging/methods , Magnetite Nanoparticles/chemistry , Monitoring, Physiologic/methods , Polyethylene Glycols/chemistry , Ferric Compounds/chemistry , Phospholipids/chemistry , Staining and Labeling/methods
11.
Korean J Radiol ; 10(6): 613-22, 2009.
Article in English | MEDLINE | ID: mdl-19885318

ABSTRACT

OBJECTIVE: To determine the feasibility of labeling human mesenchymal stem cells (hMSCs) with bifunctional nanoparticles and assessing their potential as imaging probes in the monitoring of hMSC transplantation. MATERIALS AND METHODS: The T1 and T2 relaxivities of the nanoparticles (MNP@SiO(2)[RITC]-PEG) were measured at 1.5T and 3T magnetic resonance scanner. Using hMSCs and the nanoparticles, labeling efficiency, toxicity, and proliferation were assessed. Confocal laser scanning microscopy and transmission electron microscopy were used to specify the intracellular localization of the endocytosed iron nanoparticles. We also observed in vitro and in vivo visualization of the labeled hMSCs with a 3T MR scanner and optical imaging. RESULTS: MNP@SiO(2)(RITC)-PEG showed both superparamagnetic and fluorescent properties. The r(1) and r(2) relaxivity values of the MNP@SiO(2)(RITC)-PEG were 0.33 and 398 mM(-1) s(-1) at 1.5T, respectively, and 0.29 and 453 mM(-1) s(-1) at 3T, respectively. The effective internalization of MNP@SiO(2)(RITC)-PEG into hMSCs was observed by confocal laser scanning fluorescence microscopy. The transmission electron microscopy images showed that MNP@SiO(2)(RITC)-PEG was internalized into the cells and mainly resided in the cytoplasm. The viability and proliferation of MNP@SiO(2)(RITC)-PEG-labeled hMSCs were not significantly different from the control cells. MNP@SiO(2)(RITC)-PEG-labeled hMSCs were observed in vitro and in vivo with optical and MR imaging. CONCLUSION: MNP@SiO(2)(RITC)-PEG can be a useful contrast agent for stem cell imaging, which is suitable for a bimodal detection by MRI and optical imaging.


Subject(s)
Magnetic Resonance Imaging/methods , Mesenchymal Stem Cells , Nanoparticles/chemistry , Animals , Biocompatible Materials , Cells, Cultured , Cobalt , Feasibility Studies , Ferric Compounds , Humans , Mice , Mice, Nude , Microscopy, Confocal , Microscopy, Electron , Phantoms, Imaging , Polyethylene Glycols , Rats , Rhodamines , Silicon Dioxide , Staining and Labeling/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...