Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
J Biol Chem ; 296: 100346, 2021.
Article in English | MEDLINE | ID: mdl-33524396

ABSTRACT

Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has developed into a global pandemic since its first outbreak in the winter of 2019. An extensive investigation of SARS-CoV-2 is critical for disease control. Various recombinant monoclonal antibodies of human origin that neutralize SARS-CoV-2 infection have been isolated from convalescent patients and will be applied as therapies and prophylaxis. However, the need for dedicated monoclonal antibodies suitable for molecular pathology research is not fully addressed. Here, we produced six mouse anti-SARS-CoV-2 spike monoclonal antibodies that not only exhibit robust performance in immunoassays including western blotting, ELISA, immunofluorescence, and immunoprecipitation, but also demonstrate neutralizing activity against SARS-CoV-2 infection to VeroE6/TMPRSS2 cells. Due to their mouse origin, our monoclonal antibodies are compatible with the experimental immunoassay setups commonly used in basic molecular biology research laboratories, providing a useful tool for future research. Furthermore, in the hope of applying the antibodies of clinical setting, we determined the variable regions of the antibodies and used them to produce recombinant human/mouse chimeric antibodies.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Antibodies, Neutralizing/biosynthesis , Antibodies, Viral/biosynthesis , COVID-19/prevention & control , SARS-CoV-2/genetics , Spike Glycoprotein, Coronavirus/genetics , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/isolation & purification , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/isolation & purification , Antibodies, Viral/chemistry , Antibodies, Viral/isolation & purification , Binding Sites , COVID-19/immunology , COVID-19/virology , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Mice , Neutralization Tests , Protein Binding , Protein Interaction Domains and Motifs , Protein Subunits/administration & dosage , Protein Subunits/genetics , Protein Subunits/immunology , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/administration & dosage , Spike Glycoprotein, Coronavirus/immunology , Vaccination
2.
Viruses ; 10(8)2018 08 14.
Article in English | MEDLINE | ID: mdl-30110893

ABSTRACT

Prevention of severe lower respiratory tract infections in infants caused by the human respiratory syncytial virus (hRSV) remains a major public health priority. Currently, the major focus of vaccine development relies on the RSV fusion (F) protein since it is the main target protein for neutralizing antibodies induced by natural infection. The protein conserves 5 N-glycosylation sites, two of which are located in the F2 subunit (N27 and N70), one in the F1 subunit (N500) and two in the p27 peptide (N116 and N126). To study the influence of the loss of one or more N-glycosylation sites on RSV F immunogenicity, BALB/c mice were immunized with plasmids encoding RSV F glycomutants. In comparison with F WT DNA immunized mice, higher neutralizing titres were observed following immunization with F N116Q. Moreover, RSV A2-K-line19F challenge of mice that had been immunized with mutant F N116Q DNA was associated with lower RSV RNA levels compared with those in challenged WT F DNA immunized animals. Since p27 is assumed to be post-translationally released after cleavage and thus not present on the mature RSV F protein, it remains to be elucidated how deletion of this glycan can contribute to enhanced antibody responses and protection upon challenge. These findings provide new insights to improve the immunogenicity of RSV F in potential vaccine candidates.


Subject(s)
Antibodies, Neutralizing/biosynthesis , Antibodies, Viral/biosynthesis , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/administration & dosage , Vaccines, DNA/administration & dosage , Viral Fusion Proteins/genetics , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Female , Glycosylation , Humans , Hydrolysis , Immunization , Mice , Mice, Inbred BALB C , Models, Molecular , Mutation , Plasmids/administration & dosage , Plasmids/genetics , Plasmids/immunology , Protein Engineering , Protein Subunits/administration & dosage , Protein Subunits/genetics , Protein Subunits/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus Vaccines/genetics , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus, Human/drug effects , Respiratory Syncytial Virus, Human/genetics , Respiratory Syncytial Virus, Human/immunology , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Viral Fusion Proteins/administration & dosage , Viral Fusion Proteins/immunology , Viral Load/drug effects
3.
J Biochem Mol Toxicol ; 31(11)2017 Nov.
Article in English | MEDLINE | ID: mdl-28700109

ABSTRACT

Light subunit of mushroom tyrosinase (LSMT) is a protein of unknown function from mushroom Agaricus bisporus that has been demonstrated to permeate through rat intestine ex vivo. Thus, it can be absorbed in the intestine, thereby holding a promise as a drug carrier for oral administration, similar to HA-33 protein from botulinum, one of the closest structural homologs of LSMT. However, the safety of LSMT should be ensured prior to its use. Here, we described biological response of LSMT upon weekly intraperitoneal administration of 50 µg/day to the Balb/c mice for 12 weeks. Motoric and behavior profiles, as well as the index of main organs (liver, spleen, lung, heart, and kidney), and body weight, were not significantly changed as compared with the control group. Also, no IgG was detected in the serum. The results suggest that LSMT is safe for further development.


Subject(s)
Agaricus/enzymology , Behavior, Animal/drug effects , Monophenol Monooxygenase/administration & dosage , Animals , Body Weight/drug effects , Female , Immune System/drug effects , Immunoglobulin G/blood , Infusions, Parenteral , Male , Mice, Inbred BALB C , Protein Subunits/administration & dosage
4.
J Immunol ; 193(10): 5044-55, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25288571

ABSTRACT

Myasthenia gravis (MG) and experimental autoimmune myasthenia gravis (EAMG) are caused by Ab-mediated autoimmune responses to muscle nicotinic acetylcholine receptors (AChRs) that impair neuromuscular transmission, thereby causing muscle weakness. Previously, we discovered that i.p. injection of a therapeutic vaccine consisting of bacterially expressed cytoplasmic domains of human AChR subunits reduced the development of chronic EAMG in rats. In this article, we show that immunization with the therapeutic vaccine in adjuvants does not induce EAMG and, thus, is safe. The potency and efficacy of the therapeutic vaccine were greatly increased by s.c. administration of repeated low doses in IFA. Onset of chronic EAMG could be prevented. Established chronic EAMG could be rapidly reversed, modeling therapy of chronic MG. Therapy reduced pathological Abs assayed by immune precipitation of a main immunogenic region chimera. Successfully treated rats exhibited long-term resistance to reinduction of EAMG, suggesting a lasting cure of MG. A long-term effect of therapy was to change the isotype of the pathogenic Ab response from IgG2b, which fixes complement, to IgG1, which does not. Prevention and reversal of chronic EAMG was not caused by the isotype switch, but the isotype switch may contribute to resistance to reinduction of EAMG. Immunization with AChR cytoplasmic domains in adjuvant is promising as a safe, Ag-specific, potent, effective, rapidly acting, and long-lasting therapeutic approach to MG.


Subject(s)
Myasthenia Gravis, Autoimmune, Experimental/drug therapy , Protein Subunits/immunology , Receptors, Cholinergic/immunology , Vaccines/immunology , Animals , Autoantibodies/biosynthesis , Female , Humans , Immunoglobulin Class Switching/drug effects , Immunoglobulin G/biosynthesis , Injections, Subcutaneous , Muscles/drug effects , Muscles/immunology , Muscles/pathology , Myasthenia Gravis, Autoimmune, Experimental/chemically induced , Myasthenia Gravis, Autoimmune, Experimental/immunology , Myasthenia Gravis, Autoimmune, Experimental/pathology , Protein Subunits/administration & dosage , Protein Subunits/chemistry , Rats , Rats, Inbred Lew , Receptors, Cholinergic/administration & dosage , Receptors, Cholinergic/chemistry , Recombinant Proteins/administration & dosage , Recombinant Proteins/chemistry , Recombinant Proteins/immunology , Torpedo , Vaccines/administration & dosage , Vaccines/chemistry
5.
J Immunol ; 191(1): 44-51, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23740950

ABSTRACT

Abs that bind the functional envelope glycoprotein (Env) spike are considered critical for a broadly effective prophylactic HIV-1 vaccine. The difficulty in eliciting such Abs by vaccination is partially attributed to the immunodominance of hydrophilic, surface-exposed variable protein regions of Env. However, little is known about the potential for competition between B cells that recognize distinct and distal epitopes on Env during protein subunit vaccination. In this study, we address this basic question at the level of Ab-secreting cells and serum IgG using a pair of isogenic soluble Env trimers, designated wildtype and gV3, which differ only in their potential to activate B cell responses against the highly immunogenic V3 region of Env. Immunization of mice with gV3 resulted in a markedly lower Ag-specific response compared with that induced by wildtype Env and could be explained by a loss of V3-directed reactivities. There was no redistribution of the response to other regions of Env in gV3-inoculated mice, suggesting that the epitope-specific Ab-secreting cell responses measured after boost are independently regulated rather than dictated by direct or indirect competition between B cells recognizing different structural elements of Env. This information is relevant for ongoing efforts in Env immunogen design to focus responses on conserved neutralizing determinants and for our general understanding of B cell responses to large-protein Ags that display numerous B cell epitopes.


Subject(s)
AIDS Vaccines/administration & dosage , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Cell Differentiation/immunology , Epitopes, B-Lymphocyte/biosynthesis , Epitopes, B-Lymphocyte/immunology , AIDS Vaccines/chemistry , AIDS Vaccines/immunology , Animals , B-Lymphocyte Subsets/cytology , Epitopes, B-Lymphocyte/metabolism , Female , HIV Envelope Protein gp120/administration & dosage , HIV Envelope Protein gp120/immunology , Humans , Immunization, Secondary , Immunoglobulin G/biosynthesis , Immunoglobulin M/biosynthesis , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred BALB C , Plasma Cells/cytology , Plasma Cells/immunology , Plasma Cells/metabolism , Protein Subunits/administration & dosage , Protein Subunits/chemistry , Protein Subunits/immunology , env Gene Products, Human Immunodeficiency Virus/administration & dosage , env Gene Products, Human Immunodeficiency Virus/immunology
6.
J Control Release ; 162(3): 477-84, 2012 Sep 28.
Article in English | MEDLINE | ID: mdl-22841796

ABSTRACT

The generation of both antibody and CD8⁺ T cell responses against pathogens is considered important for many advanced vaccines for diseases including tuberculosis, HIV and malaria. However, most current vaccines are delivered into muscle by the needle and syringe method and induce protection via humoral (antibody) immune responses. In this paper, we test the hypothesis that delivering a model subunit protein antigen (ovalbumin) to the skin's abundant immune cell population using a densely packed microprojection array (Nanopatch) enhances CD8⁺ T cell responses. We found that the Nanopatch significantly enhanced the CD8⁺ T cell responses when compared to intramuscular delivery of both antigen-only and adjuvanted cases (Quil-A and CpG; separately). To our knowledge, this is the first published study demonstrating significantly improved CD8⁺ T cell responses achieved by delivering subunit vaccines to the skin's abundant immune cell population. Successfully replicating these findings in humans could significantly advance the reach of vaccines.


Subject(s)
Ovalbumin/administration & dosage , Protein Subunits/administration & dosage , Vaccination/methods , Vaccines, Subunit/administration & dosage , Adjuvants, Immunologic/administration & dosage , Animals , Antigen-Presenting Cells/immunology , CD8-Positive T-Lymphocytes/immunology , Female , Methylcellulose/chemistry , Mice , Mice, Inbred C57BL , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Oligodeoxyribonucleotides/administration & dosage , Quillaja Saponins , Saponins/administration & dosage , Skin/immunology , Spleen/cytology , Transdermal Patch
7.
Vaccine ; 30(32): 4719-27, 2012 Jul 06.
Article in English | MEDLINE | ID: mdl-22652401

ABSTRACT

We showed previously that calcineurin B subunit (CnB) protein activates innate immune cells including macrophages, monocytes and dendritic cells and acts as an adjuvant of a model antigen (ovalbumin) and a recombinant pneumolysin antigen, but the detailed mechanism is not clear and whether it can serve as an adjuvant of a commercial HBV vaccine is unknown. Here, we report that CnB promotes inflammatory cytokines production, splenocytes proliferation and NK lytic activity, and that CnB-induced inflammatory cytokines (IFN-γ, IL-6, TNF-α) production is dependent on integrin αM. Animal experiments demonstrate that CnB markedly increases the total anti-HBs antibodies in a dose and time dependent manner. Furthermore, CnB increases both anti-HBs IgM and anti-HBs IgG titers and changes the balance of IgG2a and IgG1. Combined use of CnB and CpG induces more cytokines production in splenocytes, as well as more anti-HBs antibodies production in vivo. These results reveal a probable mechanism of CnB-induced inflammatory cytokines production and further demonstrate that CnB is a novel and effective adjuvant of Engerix-B HBV vaccine.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Calcineurin/immunology , Hepatitis B Vaccines/immunology , Immunity, Innate , Animals , CD11b Antigen/immunology , Calcineurin/administration & dosage , Cells, Cultured , Cytokines/immunology , Dose-Response Relationship, Immunologic , Female , Hepatitis Antibodies/blood , Hepatitis B/immunology , Hepatitis B/prevention & control , Immunoglobulin G/blood , Immunoglobulin M/blood , Killer Cells, Natural/immunology , Mice , Mice, Inbred BALB C , Protein Subunits/administration & dosage , Protein Subunits/immunology , Spleen/cytology , Spleen/immunology , Vaccines, Synthetic/immunology
8.
Infect Immun ; 80(7): 2426-35, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22526674

ABSTRACT

Enterotoxigenic Escherichia coli (ETEC) produces both heat-labile (LT) and heat-stable (ST) enterotoxins and is a major cause of diarrhea in infants in developing countries and in travelers to those regions. In addition to inducing fluid secretion, LT is a powerful mucosal adjuvant capable of promoting immune responses to coadministered antigens. In this study, we examined purified A subunit to further understand the toxicity and adjuvanticity of LT. Purified A subunit was enzymatically active but sensitive to proteolytic degradation and unable to bind gangliosides, and even in the presence of admixed B subunit, it displayed low cyclic AMP (cAMP) induction and no enterotoxicity. Thus, the AB5 structure plays a key role in protecting the A subunit from proteolytic degradation and in delivering the enzymatic signals required for secretion. In contrast, the A subunit alone was capable of activating dendritic cells and enhanced immune responses to multiple antigens following intranasal immunization; therefore, unlike toxicity, LT adjuvanticity is not dependent on the AB5 holotoxin structure or the presence of the B subunit. However, immune responses were maximal when signals were received from both subunits either in an AB5 structure or with A and B admixed. Furthermore, the quality of the immune response (i.e., IgG1/IgG2 balance and mucosal IgA and IL-17 secretion) was determined by the presence of an A subunit, revealing for the first time induction of Th17 responses with the A subunit alone. These results have important implications for understanding ETEC pathogenesis, unraveling immunologic responses induced by LT-based adjuvants, and developing new mucosal vaccines.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Bacterial Toxins/administration & dosage , Enterotoxins/administration & dosage , Escherichia coli Proteins/administration & dosage , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Th17 Cells/immunology , Vaccines/immunology , Adjuvants, Immunologic/adverse effects , Administration, Intranasal , Animals , Bacterial Toxins/adverse effects , Dendritic Cells/immunology , Enterotoxins/adverse effects , Escherichia coli Proteins/adverse effects , Mice , Mice, Inbred BALB C , Protein Subunits/administration & dosage , Vaccines/administration & dosage , Vaccines/adverse effects
9.
Toxicol Sci ; 126(1): 72-83, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21987460

ABSTRACT

Ricin, isolated from the castor bean plant Ricinus communis, is included on the Centers for Disease Control and Prevention Category B list of bioterrorism agents, indicating that the toxin is moderately easy to disseminate and could result in moderate morbidity rates. This study evaluated two promising recombinant ricin subunit vaccines, one made using an Escherichia coli codon-optimized gene and the other using a yeast codon-optimized gene in E. coli-based fermentations. Rabbits were vaccinated four times over a period of 6 months and challenged with ∼10 to 30 times the median lethal dose of aerosolized ricin. All unvaccinated control rabbits were either found dead or humanely euthanized within 30 h postchallenge, while the rabbits vaccinated with either vaccine survived the exposure without adverse clinical signs. When the protective antibody responses were analyzed, no significant difference was seen between the two vaccines. However, there was a significant difference in the immune response over time for both vaccines tested. Although clinical pathology was unremarkable, significant histological lesions in the control animals included fibrinonecrotic pneumonia, acute necrotizing lesions in the upper respiratory tract, and necrotizing lymphadenitis in the lymph nodes draining the upper and lower respiratory tract. Vaccine-treated rabbits exhibited resolving lesions associated with ricin exposure, namely chronic inflammation in the upper respiratory tract and lungs, fibrosis, type II pneumocyte hyperplasia, and bronchiolitis obliterans. This study confirmed the safety and efficacy of two recombinant ricin subunit vaccines in rabbits, offering potential protection to warfighters and select populations.


Subject(s)
Protein Subunits/antagonists & inhibitors , Ricin/antagonists & inhibitors , Toxins, Biological/antagonists & inhibitors , Vaccines, Subunit/therapeutic use , Administration, Inhalation , Animals , Biological Warfare Agents , Codon , Escherichia coli/genetics , Escherichia coli/metabolism , Female , Lung/drug effects , Lung/immunology , Lung/pathology , Male , Protein Subunits/administration & dosage , Protein Subunits/genetics , Rabbits , Random Allocation , Recombinant Proteins/administration & dosage , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/genetics , Ricin/administration & dosage , Ricin/genetics , Ricin/toxicity , Specific Pathogen-Free Organisms , Toxins, Biological/administration & dosage , Toxins, Biological/genetics , Toxins, Biological/toxicity , Vaccines, Subunit/adverse effects , Vaccines, Subunit/immunology , Vaccines, Synthetic/adverse effects , Vaccines, Synthetic/immunology , Vaccines, Synthetic/therapeutic use , Yeasts/genetics , Yeasts/metabolism
10.
Vaccine ; 29(45): 8094-100, 2011 Oct 19.
Article in English | MEDLINE | ID: mdl-21856355

ABSTRACT

Shigella dysenteriae is the causative agent of the third commonest bacterial disease for childhood diarrhoea and responsible for millions of deaths per year. It produces potent toxin termed Shiga toxin which is listed in category B biological warfare agent of CDC, USA. Earlier we have reported production of recombinant Shiga toxin B subunit that produced antibodies which neutralized Shiga toxin toxicity in HeLa cells. In the present study, we have evaluated the immunomodulatory potential of rStxB protein in Balb/c mice using Freunds adjuvants. Animal protection with recombinant StxB was conferred through both humoral and cellular immune responses as indicated by an increased antibody titre with predominance of IgG2a and IgG2b isotypes along with elevated levels of IgG1 subtype. Cytokine profile of the mice antiserum and splenocyte also indicates Th2 and Th1 type of immune responses where former dominates in early stage of immunization. Histopathology study of kidneys of vaccinated mice showed remarkable differences when compared to the animals infected with Shigella dysenteriae type1. The present study indicates that recombinant StxB is a promising vaccine candidate and can be used for production of therapeutic antibodies to counter Shiga intoxication.


Subject(s)
Shiga Toxin/immunology , Shigella Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Bacterial/blood , Antitoxins/blood , Cytokines/blood , Cytokines/metabolism , Dysentery, Bacillary/pathology , Dysentery, Bacillary/prevention & control , Female , Freund's Adjuvant/administration & dosage , Histocytochemistry , Immunoglobulin G/blood , Kidney/pathology , Leukocytes, Mononuclear/immunology , Mice , Mice, Inbred BALB C , Protein Subunits/administration & dosage , Protein Subunits/immunology , Shiga Toxin/administration & dosage , Shigella Vaccines/administration & dosage , Shigella dysenteriae/immunology , Shigella dysenteriae/pathogenicity , Spleen/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology
11.
J Immunol ; 181(12): 8278-87, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-19050244

ABSTRACT

Mucosal administration of Ag conjugated to cholera toxin B subunit (CTB) can efficiently induce peripheral immunologic tolerance, so-called oral tolerance, associated with development of Foxp3(+)CD25(+)CD4(+) regulatory T (Treg) cells. Using an established sublingual tolerization regimen with Ag(OVA)/CTB conjugate, wherein CTB mediates Ag uptake and presentation by most B lymphocytes irrespective of their Ag specificity, we have assessed the importance of B cells for induction of Ag-specific Treg cells and oral tolerance. We found that Treg cells are reduced in microMT(-/-) B cell-deficient mice compared with wild-type (WT) mice. After sublingual Ag/CTB treatment, Treg cells increased much more in WT than in microMT(-/-) mice; however, adoptive transfer of B cells before treatment normalized Treg cell development and functional oral tolerance. B cells from OVA/CTB-treated mice expressed more IL-10 and less CD86 than control B cells. Adoptive transfer of these cells before parenteral immunization with OVA led to efficient suppression of proliferation and to induction of apoptotic depletion of Ag-specific CD25(-)CD4(+) effector T cells associated with the expansion of Treg cells. However, also OVA/CTB-treated microMT(-/-) mice could suppress the immune response to parenteral immunization with OVA, which was associated with a strong increase in Foxp3(-)CD4(+) T cells expressing LAP/TGF-beta. Our results indicate that mucosal tolerance comprises at least two separate pathways: one being B cell dependent and associated with expansion of Treg cells and Treg-mediated suppression and depletion of effector T cells, and one being B cell independent and associated with development of Foxp3(-)LAP(+)TGF-beta(+) regulatory T cells.


Subject(s)
B-Lymphocyte Subsets/immunology , Cell Proliferation , Cholera Toxin/immunology , Immune Tolerance , Mouth Mucosa/immunology , Ovalbumin/immunology , Peptide Fragments/immunology , Protein Subunits/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Amino Acid Sequence , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/transplantation , B-Lymphocyte Subsets/transplantation , Cholera Toxin/administration & dosage , Cholera Toxin/metabolism , Cross-Linking Reagents/metabolism , Female , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Mouth Mucosa/cytology , Ovalbumin/administration & dosage , Ovalbumin/metabolism , Peptide Fragments/administration & dosage , Peptide Fragments/metabolism , Protein Subunits/administration & dosage , Protein Subunits/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/transplantation
12.
Yao Xue Xue Bao ; 43(10): 1060-5, 2008 Oct.
Article in Chinese | MEDLINE | ID: mdl-19127872

ABSTRACT

Phycocyanin subunits liposomes (PCS-lip) were prepared and its cellular uptake and photodynamic therapy (PDT) effect on cancer cells were studied. In the experiment, film dispersion method was used to prepare phycocyanin subunits liposomes; particle size and distribution were detected by zetasizer and transmission electric microscope; the effects of liposome as carrier on cell uptake in vitro were evaluated in S180 by using fluorescence microscope; and photodynamic therapy effect was assessed with MTT method. As shown in the results, the particle size mainly ranged from 80 nm to 160 nm, and average encapsulation rate was 42.3%. In the concentration of 100 microg x mL(-1), transfection rate reached (18.5 +/- 0.8)% at 2 h, (23.1 +/- 0.9)% at 4 h, keeping a balance in 5-6 h, and its photodynamic therapy effect in vitro improved with the increasing of concentration of phycocyanin subunits liposomes. In the concentration of 200 micro x mL(-1) cell survival rate of BGC-823 and S180 reached (45 +/- 5.2)% and (36 +/- 5.5)%, respectively, and the cell survival rate differentiation between PCS-PDT group and PCS-lip-PDT group reached 7%-11% (P < 0.05). In this study film dispersion method could keep the biological activity of phycocyanin subunits very well. Phycocyanin subunits liposomes will transfect cells more quickly than phycocyanin subunits in the same concentration, and in the same conditions, phycocyanin subunits liposomes have the better PDT effect on cancer cells as they were incubated with cells for 4 h.


Subject(s)
Liposomes/chemistry , Photochemotherapy/methods , Phycocyanin , Sarcoma 180/pathology , Stomach Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Survival/drug effects , Drug Carriers , Drug Stability , Humans , Mice , Particle Size , Phycocyanin/administration & dosage , Phycocyanin/pharmacology , Protein Subunits/administration & dosage , Protein Subunits/pharmacology
13.
Clin Vaccine Immunol ; 15(2): 359-66, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18003816

ABSTRACT

Enterohemorrhagic Escherichia coli (EHEC) strains are important human food-borne pathogens. EHEC strains elaborate potent Shiga toxins (Stx1, and/or Stx2) implicated in the development of hemorrhagic colitis (HC) or hemolytic-uremic syndrome (HUS). In this report, we evaluated the immunogenicity and protective efficacy of Stx1 subunit B (StxB1) administered by transcutaneous immunization (TCI). Three groups of Dutch Belted rabbits received patches containing StxB1, StxB1 in combination with Escherichia coli heat-labile enterotoxin (LT), or LT alone. An additional group of naïve rabbits served as controls. The protective efficacy following TCI with StxB1 was assessed by challenging rabbits with a virulent Stx1-producing strain, RDEC-H19A, capable of inducing HC and HUS in rabbits. Antibodies specific to StxB1 from serum and bile samples were determined by enzyme-linked immunosorbent assay and toxin neutralization test. Rabbits immunized with StxB1 demonstrated improved weight gain and reduced Stx-induced histopathology. Rabbits receiving StxB or StxB1/LT showed a significant increase in serum immunoglobulin G titers specific to StxB1 as well as toxin neutralization titers. These data demonstrated that the StxB delivered by TCI could induce significant systemic immune responses. Thus, Stx subunit B vaccine delivered by a patch for a high-risk population may be a practical approach to prevent (and/or reduce) Stx-induced pathology.


Subject(s)
Administration, Cutaneous , Escherichia coli Infections/prevention & control , Shiga Toxin/immunology , Shiga-Toxigenic Escherichia coli/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Bacterial/analysis , Antibodies, Bacterial/blood , Antitoxins/analysis , Antitoxins/blood , Bacterial Toxins/administration & dosage , Bile/immunology , Body Weight/immunology , Cecum/pathology , Colitis/prevention & control , Enterotoxins/administration & dosage , Escherichia coli Infections/immunology , Escherichia coli Proteins/administration & dosage , Feces/microbiology , Hemolytic-Uremic Syndrome/prevention & control , Intestinal Mucosa/pathology , Kidney/pathology , Protein Subunits/administration & dosage , Protein Subunits/immunology , Rabbits , Serum/immunology , Shiga Toxin/administration & dosage , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology
14.
J Infect Dis ; 196(7): 1093-101, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17763334

ABSTRACT

Subtilase cytotoxin (SubAB) is the prototype of a recently discovered AB(5) cytotoxin family produced by certain strains of Shiga toxigenic Escherichia coli (STEC). The catalytic A subunit is a highly specific subtilase-like serine protease that cleaves the endoplasmic reticulum chaperone BiP. The toxin is lethal for mice, but the pathology it induces is poorly understood. Here, we show that intraperitoneal injection of SubAB causes microangiopathic hemolytic anemia, thrombocytopenia, and renal impairment in mice--characteristics typical of Shiga toxin-induced hemolytic uremic syndrome. SubAB caused extensive microvascular thrombosis and other histologic damage in the brain, kidneys, and liver, as well as dramatic splenic atrophy. Peripheral blood leukocyte levels were increased at 24 h; there was also significant neutrophil infiltration in the liver, kidneys, and spleen and toxin-induced apoptosis at these sites. These findings raise the possibility that SubAB directly contributes to pathology in humans infected with strains of STEC that produce both Shiga toxin and SubAB.


Subject(s)
Cytotoxins/toxicity , Endoplasmic Reticulum/pathology , Escherichia coli/pathogenicity , Protein Subunits/toxicity , Shiga Toxins/toxicity , Anemia, Hemolytic/etiology , Anemia, Hemolytic/pathology , Animals , Apoptosis , Brain/pathology , Cytotoxins/administration & dosage , Escherichia coli/metabolism , Escherichia coli Proteins/administration & dosage , Escherichia coli Proteins/toxicity , Humans , Kidney/cytology , Kidney/pathology , Liver/cytology , Liver/pathology , Male , Mice , Mice, Inbred BALB C , Protein Subunits/administration & dosage , Shiga Toxins/administration & dosage , Spleen/cytology , Spleen/pathology , Thrombocytopenia/etiology , Thrombocytopenia/pathology
15.
Nano Lett ; 7(9): 2718-23, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17655369

ABSTRACT

Silica nanowires (NWs) were used to introduce the Shiga toxin type 1 A subunit (StxA1) into cultured bovine and human epithelial cells. We extended technology developed in our laboratories that employs fibronectin (Fn) to induce integrin-mediated uptake of NWs by coating NWs with StxA1 and Fn. The bonding strengths of Fn and StxA1 to the surface of NWs were measured by X-ray photoelectron spectroscopy. This technique demonstrated complex interactions between Fn, StxA1, and the NWs. Neutral red cytotoxicity assays and field emission scanning electron microscopy confirmed that the NW-StxA1-Fn complexes were effectively internalized and caused cell death. This indicates that NWs can carry StxA1 and potentially other toxic or therapeutic agents into eukaryotic cells. Ongoing studies include improved functionalizing of NWs aimed at increasing internalization efficiency and substituting ligands for specific cell targeting.


Subject(s)
Cell Survival/drug effects , Drug Carriers/administration & dosage , Escherichia coli O157/metabolism , Nanostructures/chemistry , Nanotubes/chemistry , Protein Subunits/administration & dosage , Protein Subunits/chemistry , Shiga Toxin/administration & dosage , Shiga Toxin/chemistry , Animals , Cattle , Crystallization/methods , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Compounding/methods , Humans , Materials Testing , Nanostructures/ultrastructure , Nanotechnology/methods , Nanotubes/ultrastructure , Particle Size , Protein Subunits/pharmacokinetics , Shiga Toxin/pharmacokinetics
16.
Microbiol Immunol ; 51(4): 445-55, 2007.
Article in English | MEDLINE | ID: mdl-17446685

ABSTRACT

Recombinant whole heavy chains (H, 100 kDa) and their N-terminal (Hn, 50 kDa) and C-terminal (Hc, 50 kDa) half fragments of Clostridium botulinum type C and D neurotoxins were expressed as glutathione S-transferase (GST) fusion proteins in Escherichia coli. GST eliminated-preparations of H (10 microg), Hn (5 microg), Hc (5 microg), or a mixture of Hn (5 microg) and Hc (5 microg) of types C and D were mixed with an equal volume of adjuvant, and then were twice injected into mice subcutaneously. After immunization, the mice were challenged with up to 10(6) the minimum lethal doses (MLD)/0.5 ml of C or D toxin, the type of which was same as that of the immunogens. All of the mice immunized with antigens except for Hn survived against 10(5) to 10(6) MLD/0.5 ml of the toxins, but the mice immunized with Hn were killed by 100 MLD/0.5 ml. The mice immunized with a mixture of C-Hc and D-Hc, each 5 microg, also showed a high level of resistance against both C and D toxins. Antibody levels immunized with GST fused-or GST eliminatedpreparation were quite similar. These results indicate that recombinant GST-fused Hc can be used as a safe and effective vaccine for type C and D botulism in animals. It also became clear that one time inoculation with a large amount of C-Hc or D-Hc, 100 microg, is useful for vaccine trials in mice.


Subject(s)
Bacterial Vaccines/immunology , Botulinum Toxins/immunology , Clostridium botulinum type C/chemistry , Clostridium botulinum type D/chemistry , Protein Subunits/immunology , Recombinant Fusion Proteins/immunology , Animals , Botulinum Toxins/genetics , Botulinum Toxins/pharmacology , Botulism/prevention & control , Escherichia coli/genetics , Glutathione Transferase/metabolism , Mice , Molecular Weight , Neurotoxins , Protein Subunits/administration & dosage , Protein Subunits/genetics , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/isolation & purification , Toxicity Tests , Vaccination , Vaccines, Synthetic/immunology
17.
J Pharm Pharmacol ; 58(6): 787-99, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16734980

ABSTRACT

Vesicular adjuvant systems composing dimethyldioctadecylammonium (DDA) can promote both cell-mediated and humoral immune responses to the tuberculosis vaccine fusion protein in mice. However, these DDA preparations were found to be physically unstable, forming aggregates under ambient storage conditions. Therefore there is a need to improve the stability of such systems without undermining their potent adjuvanticity. To this end, the effect of incorporating non-ionic surfactants, such as 1-monopalmitoyl glycerol (MP), in addition to cholesterol (Chol) and trehalose 6,6'-dibehenate (TDB), on the stability and efficacy of these vaccine delivery systems was investigated. Differential scanning calorimetry revealed a reduction in the phase transition temperature (T(c)) of DDA-based vesicles by approximately 12 degrees C when MP and cholesterol (1:1 molar ratio) were incorporated into the DDA system. Transmission electron microscopy (TEM) revealed the addition of MP to DDA vesicles resulted in the formation of multi-lamellar vesicles. Environmental scanning electron microscopy (ESEM) of MP-Chol-DDA-TDB (16:16:4:0.5 micromol) indicated that incorporation of antigen led to increased stability of the vesicles, perhaps as a result of the antigen embedding within the vesicle bilayers. At 4 degrees C DDA liposomes showed significant vesicle aggregation after 28 days, although addition of MP-Chol or TDB was shown to inhibit this instability. Alternatively, at 25 degrees C only the MP-based systems retained their original size. The presence of MP within the vesicle formulation was also shown to promote a sustained release of antigen in-vitro. The adjuvant activity of various systems was tested in mice against three subunit antigens, including mycobacterial fusion protein Ag85B-ESAT-6, and two malarial antigens (Merozoite surface protein 1, MSP1, and the glutamate rich protein, GLURP). The MP- and DDA-based systems induced antibody responses at comparable levels whereas the DDA-based systems induced more powerful cell-mediated immune responses.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Liposomes , Protein Subunits/administration & dosage , Quaternary Ammonium Compounds/administration & dosage , Vaccines, Subunit/administration & dosage , Animals , Calorimetry, Differential Scanning , Female , Immunization , Immunoglobulin G/blood , Mice , Mice, Inbred C57BL , Microscopy, Electron, Scanning , Particle Size
18.
J Immunol ; 176(5): 2749-57, 2006 Mar 01.
Article in English | MEDLINE | ID: mdl-16493030

ABSTRACT

The use of the nontoxic B subunit of cholera toxin (CTB) as mucosal adjuvant and carrier-delivery system for inducing secretory Ab responses has been documented previously with different soluble Ags. In this study, we have evaluated this approach for inducing CTL responses against a prototype Ag, OVA, in the female genital mucosa. We report here the ability of an immunogen comprised of CTB conjugated to OVA (CTB-OVA) given by intravaginal (ivag) route to induce genital OVA-specific CTLs in mice. Using adoptive transfer models, we demonstrate that ivag application of CTB-OVA activates OVA-specific IFN-gamma-producing CD4 and CD8 T cells in draining lymph nodes (DLN). Moreover, ivag CTB induces an expansion of IFN-gamma-secreting CD8+ T cells in DLN and genital mucosa and promotes Ab responses to OVA. In contrast, ivag administration of OVA alone or coadministered with CTB failed to induce such responses. Importantly, we demonstrate that ivag CTB-OVA generates OVA-specific CTLs in DLN and the genital mucosa. Furthermore, genital CD11b+ CD11c+ dendritic cells (DCs), but not CD8+ CD11c+ or CD11c- APCs, present MHC class I epitopes acquired after ivag CTB-OVA, suggesting a critical role of this DC subset in the priming of genital CTLs. Inhibition studies indicate that the presentation of OVA MHC class I epitopes by DCs conditioned with CTB-OVA involves a proteasome-dependent and chloroquine-sensitive mechanism. These results demonstrate that CTB is an efficient adjuvant-delivery system for DC-mediated induction of genital CTL responses and may have implications for the design of vaccines against sexually transmitted infections.


Subject(s)
Cholera Toxin/immunology , Cholera Vaccines/immunology , Cytotoxicity, Immunologic , Dendritic Cells/immunology , Protein Subunits/immunology , Administration, Intravaginal , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Cells, Cultured , Cholera Toxin/administration & dosage , Cholera Vaccines/administration & dosage , Epitopes, T-Lymphocyte/immunology , Female , Immunity, Mucosal , Interferon-gamma/metabolism , Interleukin-10/metabolism , Lymph Nodes/cytology , Lymph Nodes/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Ovalbumin/administration & dosage , Ovalbumin/immunology , Protein Subunits/administration & dosage
19.
Exp Neurol ; 197(2): 347-52, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16289170

ABSTRACT

We investigated whether administration of neurotrophin-3 (NT-3) and NMDA-2D-expressing units, found previously to enhance transmission in neonatal rat spinal cord, strengthens synaptic connections in the injured neonatal cord. We employed electrophysiological methods to evaluate the strength of synaptic transmission to individual motoneurons in the contusion and staggered double hemisection spinal cord injury (SCI) models. SCI at caudal thoracic levels (T11-T12) was carried out at postnatal day 2 (P2). Plugs containing NT-3- secreting fibroblasts and NR2D-expressing HSV-1 amplicons (HSVnr2d) were implanted above the lesion. Control animals were treated with an amplicon-expressing beta-galactosidase (HSVlac). After 8-10 days of treatment, the rats were sacrificed and spinal cords were removed for intracellular recording. Untreated contused cords preserved a fraction of white matter and weak monosynaptic responses were observed through the injury region. However, no synaptic connections were observed in control cords receiving double hemisection injury. Combined treatment with NT-3 and HSVnr2d strengthened monosynaptic connections in contused cords and induced the appearance of weak but functional multisynaptic connections in double hemisected cords. In contrast, treatment with either NT-3 or HSVnr2d alone failed to induce appearance of synaptic responses through the hemisected region. These results suggest that chronic treatment with NT-3 secreting fibroblasts combined with facilitated function of NMDA receptors by HSVnr2d treatment strengthens connections that survive incomplete SCI and therefore that such combined treatment might facilitate recovery of function following SCI.


Subject(s)
Neurotrophin 3/administration & dosage , Protein Subunits/administration & dosage , Receptors, N-Methyl-D-Aspartate/chemistry , Spinal Cord Injuries/therapy , Synaptic Transmission/physiology , Animals , Animals, Newborn , Disease Models, Animal , Genetic Engineering/methods , Genetic Vectors/physiology , Herpesvirus 1, Human/physiology , Motor Neurons/metabolism , Motor Neurons/pathology , Neural Conduction/physiology , Neural Conduction/radiation effects , Neurotrophin 3/biosynthesis , Protein Subunits/biosynthesis , Rats , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology
20.
J Immunol ; 175(4): 2219-26, 2005 Aug 15.
Article in English | MEDLINE | ID: mdl-16081789

ABSTRACT

Immunological rejection is the major cause of human corneal allograft failure. We hypothesized that local production of IL-4 or the p40 subunit of IL-12 (p40 IL-12) by the grafted cornea might prolong allograft survival. Replication-deficient adenoviral vectors encoding ovine IL-4 or p40 IL-12 and GFP were generated and used to infect ovine corneas ex vivo. mRNA for each cytokine was detected in infected corneas, and the presence of secreted protein in corneal supernatants was confirmed by bioassay (for IL-4) or immunoprecipitation (for p40 IL-12). Sheep received uninfected or gene-modified orthotopic corneal allografts. Postoperatively, untreated corneas (n = 13) and corneas expressing GFP (n = 6) were rejected at a median of 21 and 20 days, respectively. Corneas expressing IL-4 (n = 6) underwent rejection at 18.5 days (p > 0.05 compared with controls) and histology demonstrated the presence of eosinophils. In contrast, corneas expressing p40 IL-12 (n = 9) showed prolonged allograft survival (median day to rejection = 45 days, p = 0.003). Local intraocular production of p40 IL-12 thus prolonged corneal graft survival significantly, but local production of the prototypic immunomodulatory cytokine IL-4 induced eosinophilia, inflammation, and rejection. These findings have important implications for the development of novel strategies to improve human corneal graft survival.


Subject(s)
Corneal Transplantation/immunology , Endothelium, Corneal/immunology , Gene Transfer Techniques , Graft Enhancement, Immunologic , Graft Survival/genetics , Graft Survival/immunology , Interleukin-12/genetics , Interleukin-4/genetics , Protein Subunits/genetics , Adenoviridae/genetics , Animals , Cell Movement/immunology , Cell-Free System/immunology , Cell-Free System/metabolism , Corneal Transplantation/pathology , Endothelium, Corneal/metabolism , Endothelium, Corneal/pathology , Genetic Vectors , Graft Enhancement, Immunologic/methods , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Interleukin-12/administration & dosage , Interleukin-12/biosynthesis , Interleukin-12 Subunit p40 , Interleukin-4/biosynthesis , Iris/immunology , Iris/pathology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/pathology , Organ Culture Techniques , Protein Subunits/administration & dosage , Protein Subunits/biosynthesis , Sheep , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...