Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 354
Filter
1.
Cell Rep ; 43(5): 114235, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38748880

ABSTRACT

Nanoparticle vaccines displaying mosaic receptor-binding domains (RBDs) or spike (S) from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) or other sarbecoviruses are used in preparedness against potential zoonotic outbreaks. Here, we describe a self-assembling nanoparticle using lumazine synthase (LuS) as the scaffold to display RBDs from different sarbecoviruses. Mosaic nanoparticles induce sarbecovirus cross-neutralizing antibodies comparable to a nanoparticle cocktail. We find mosaic nanoparticles elicit a B cell receptor repertoire using an immunodominant germline gene pair of IGHV14-3:IGKV14-111. Most of the tested IGHV14-3:IGKV14-111 monoclonal antibodies (mAbs) are broadly cross-reactive to clade 1a, 1b, and 3 sarbecoviruses. Using mAb competition and cryo-electron microscopy, we determine that a representative IGHV14-3:IGKV14-111 mAb, M2-7, binds to a conserved epitope on the RBD, largely overlapping with the pan-sarbecovirus mAb S2H97. This suggests mosaic nanoparticles expand B cell recognition of the common epitopes shared by different clades of sarbecoviruses. These results provide immunological insights into the cross-reactive responses elicited by mosaic nanoparticles against sarbecoviruses.


Subject(s)
Nanoparticles , Nanoparticles/chemistry , Animals , Humans , SARS-CoV-2/immunology , Antibodies, Viral/immunology , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Mice , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/chemistry , Cross Reactions/immunology , Antibody Formation/immunology , COVID-19/immunology , COVID-19/virology , Protein Domains , Mice, Inbred BALB C , Multienzyme Complexes/immunology , Female , Immunodominant Epitopes/immunology
2.
Int J Mol Sci ; 23(3)2022 Jan 25.
Article in English | MEDLINE | ID: mdl-35163258

ABSTRACT

Antibody-enzyme complexes (AECs) are ideal molecular recognition elements for immunosensing applications. One molecule possesses both a binding ability to specific targets and catalytic activity to gain signals, particularly oxidoreductases, which can be integrated into rapid and sensitive electrochemical measurements. The development of AECs using fragment antibodies rather than intact antibodies, such as immunoglobulin G (IgG), has attracted attention for overcoming the ethical and cost issues associated with the production of intact antibodies. Conventionally, chemical conjugation has been used to fabricate AECs; however, controlling stoichiometric conjugation using this method is difficult. To prepare homogeneous AECs, methods based on direct fusion and enzymatic conjugation have been developed, and more convenient methods using Catcher/Tag systems as coupling modules have been reported. In this review, we summarize the methods for fabricating AECs using fragment antibodies developed for sensing applications and discuss the advantages and disadvantages of each method.


Subject(s)
Antibodies/immunology , Immunoassay/methods , Multienzyme Complexes/immunology , Animals , Humans , Immunoglobulin G/immunology
3.
Nature ; 601(7891): 110-117, 2022 01.
Article in English | MEDLINE | ID: mdl-34758478

ABSTRACT

Individuals with potential exposure to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) do not necessarily develop PCR or antibody positivity, suggesting that some individuals may clear subclinical infection before seroconversion. T cells can contribute to the rapid clearance of SARS-CoV-2 and other coronavirus infections1-3. Here we hypothesize that pre-existing memory T cell responses, with cross-protective potential against SARS-CoV-2 (refs. 4-11), would expand in vivo to support rapid viral control, aborting infection. We measured SARS-CoV-2-reactive T cells, including those against the early transcribed replication-transcription complex (RTC)12,13, in intensively monitored healthcare workers (HCWs) who tested repeatedly negative according to PCR, antibody binding and neutralization assays (seronegative HCWs (SN-HCWs)). SN-HCWs had stronger, more multispecific memory T cells compared with a cohort of unexposed individuals from before the pandemic (prepandemic cohort), and these cells were more frequently directed against the RTC than the structural-protein-dominated responses observed after detectable infection (matched concurrent cohort). SN-HCWs with the strongest RTC-specific T cells had an increase in IFI27, a robust early innate signature of SARS-CoV-2 (ref. 14), suggesting abortive infection. RNA polymerase within RTC was the largest region of high sequence conservation across human seasonal coronaviruses (HCoV) and SARS-CoV-2 clades. RNA polymerase was preferentially targeted (among the regions tested) by T cells from prepandemic cohorts and SN-HCWs. RTC-epitope-specific T cells that cross-recognized HCoV variants were identified in SN-HCWs. Enriched pre-existing RNA-polymerase-specific T cells expanded in vivo to preferentially accumulate in the memory response after putative abortive compared to overt SARS-CoV-2 infection. Our data highlight RTC-specific T cells as targets for vaccines against endemic and emerging Coronaviridae.


Subject(s)
Asymptomatic Infections , COVID-19/immunology , COVID-19/virology , DNA-Directed RNA Polymerases/immunology , Memory T Cells/immunology , SARS-CoV-2/immunology , Seroconversion , Cell Proliferation , Cohort Studies , DNA-Directed RNA Polymerases/metabolism , Evolution, Molecular , Female , Health Personnel , Humans , Male , Membrane Proteins/immunology , Memory T Cells/cytology , Multienzyme Complexes/immunology , SARS-CoV-2/enzymology , SARS-CoV-2/growth & development , Transcription, Genetic/immunology
4.
Int J Mol Sci ; 21(24)2020 Dec 19.
Article in English | MEDLINE | ID: mdl-33352757

ABSTRACT

Tumor-associated (TA) autoantibodies have been identified at the early tumor stage before developing clinical symptoms, which holds hope for early cancer diagnosis. We identified a TA autoantibody from HBx-transgenic (HBx-tg) hepatocellular carcinoma (HCC) model mouse, characterized its target antigen, and examined its relationship to human HCC. The mimotopes corresponding to the antigenic epitope of TA autoantibody were screened from a random cyclic peptide library and used for the detection of serum TA autoantibody. The target antigen of the TA autoantibody was identified as an oncogenic bi-functional purine biosynthesis protein, ATIC. It was upregulated in liver cancer tissues of HBx-tg mouse as well as human HCC tissues. Over-expressed ATIC was also secreted extracellularly via the cancer-derived exosomes, which might cause auto-immune responses. The cyclic peptide mimotope with a high affinity to anti-ATIC autoantibody, CLPSWFHRC, distinguishes between serum samples from HCC patients and healthy subjects with 70.83% sensitivity, 90.68% specificity (AUC = 0.87). However, the recombinant human ATIC protein showed a low affinity to anti-ATIC autoantibody, which may be incompatible as a capture antigen for serum TA autoantibody. This study indicates that anti-ATIC autoantibody can be a potential HCC-associated serum biomarker and suggests that autoantibody biomarker's efficiency can be improved by using antigenic mimicry to native antigens present in vivo.


Subject(s)
Autoantibodies/blood , Biomarkers, Tumor/blood , Carcinoma, Hepatocellular/diagnosis , Epitopes/immunology , Hydroxymethyl and Formyl Transferases/immunology , Liver Neoplasms/diagnosis , Multienzyme Complexes/immunology , Nucleotide Deaminases/immunology , Peptides, Cyclic/immunology , Adult , Aged , Aged, 80 and over , Animals , Autoantibodies/immunology , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/immunology , Female , Humans , Liver Neoplasms/blood , Liver Neoplasms/immunology , Male , Mice , Mice, Transgenic , Middle Aged , Peptide Library , Prognosis , Young Adult
5.
J Immunother Cancer ; 8(1)2020 06.
Article in English | MEDLINE | ID: mdl-32518090

ABSTRACT

BACKGROUND: Natural killer and cytotoxic CD8+ T cells are major players during antitumor immunity. They express NKG2D, an activating receptor that promotes tumor elimination through recognition of the MHC class I chain-related proteins A and B (MICA and MICB). Both molecules are overexpressed on a great variety of tumors from different tissues, making them attractive targets for immunotherapy. However, tumors shed MICA and MICB, and the soluble forms of both (sMICA and sMICB) mediate tumor-immune escape. Some reports indicate that anti-MICA antibodies (Ab) can promote the restoration of antitumor immunity through the induction of direct antitumor effects (antibody-dependent cell-mediated cytotoxicity, ADCC) and scavenging of sMICA. Therefore, we reasoned that an active induction of anti-MICA Ab with an immunogenic protein might represent a novel therapeutic and prophylactic alternative to restore antitumor immunity. METHODS: We generated a highly immunogenic chimeric protein (BLS-MICA) consisting of human MICA fused to the lumazine synthase from Brucella spp (BLS) and used it to generate anti-MICA polyclonal Ab (pAb) and to investigate if these anti-MICA Ab can reinstate antitumor immunity in mice using two different mouse tumors engineered to express MICA. We also explored the underlying mechanisms of this expected therapeutic effect. RESULTS: Immunization with BLS-MICA and administration of anti-MICA pAb elicited by BLS-MICA significantly delayed the growth of MICA-expressing mouse tumors but not of control tumors. The therapeutic effect of immunization with BLS-MICA included scavenging of sMICA and the anti-MICA Ab-mediated ADCC, promoting heightened intratumoral M1/proinflammatory macrophage and antigen-experienced CD8+ T cell recruitment. CONCLUSIONS: Immunization with the chimeric protein BLS-MICA constitutes a useful way to actively induce therapeutic anti-MICA pAb that resulted in a reprogramming of the antitumor immune response towards an antitumoral/proinflammatory phenotype. Hence, the BLS-MICA chimeric protein constitutes a novel antitumor vaccine of potential application in patients with MICA-expressing tumors.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibody-Dependent Cell Cytotoxicity/immunology , Histocompatibility Antigens Class I/immunology , Lymphoma/immunology , Recombinant Fusion Proteins/immunology , Urinary Bladder Neoplasms/immunology , Animals , Brucella/enzymology , Female , Histocompatibility Antigens Class I/genetics , Lymphoma/pathology , Lymphoma/therapy , Male , Mice , Mice, Inbred C57BL , Multienzyme Complexes/genetics , Multienzyme Complexes/immunology , NK Cell Lectin-Like Receptor Subfamily K/genetics , NK Cell Lectin-Like Receptor Subfamily K/immunology , Tumor Cells, Cultured , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/therapy
6.
Nature ; 570(7760): 194-199, 2019 06.
Article in English | MEDLINE | ID: mdl-31142841

ABSTRACT

Serine hydroxymethyltransferase 2 (SHMT2) regulates one-carbon transfer reactions that are essential for amino acid and nucleotide metabolism, and uses pyridoxal-5'-phosphate (PLP) as a cofactor. Apo SHMT2 exists as a dimer with unknown functions, whereas PLP binding stabilizes the active tetrameric state. SHMT2 also promotes inflammatory cytokine signalling by interacting with the deubiquitylating BRCC36 isopeptidase complex (BRISC), although it is unclear whether this function relates to metabolism. Here we present the cryo-electron microscopy structure of the human BRISC-SHMT2 complex at a resolution of 3.8 Å. BRISC is a U-shaped dimer of four subunits, and SHMT2 sterically blocks the BRCC36 active site and inhibits deubiquitylase activity. Only the inactive SHMT2 dimer-and not the active PLP-bound tetramer-binds and inhibits BRISC. Mutations in BRISC that disrupt SHMT2 binding impair type I interferon signalling in response to inflammatory stimuli. Intracellular levels of PLP regulate the interaction between BRISC and SHMT2, as well as inflammatory cytokine responses. These data reveal a mechanism in which metabolites regulate deubiquitylase activity and inflammatory signalling.


Subject(s)
Deubiquitinating Enzymes/metabolism , Glycine Hydroxymethyltransferase/metabolism , Interferon Type I/immunology , Multienzyme Complexes/immunology , Multienzyme Complexes/metabolism , Signal Transduction/immunology , Cryoelectron Microscopy , Deubiquitinating Enzymes/antagonists & inhibitors , Deubiquitinating Enzymes/chemistry , Deubiquitinating Enzymes/ultrastructure , Glycine Hydroxymethyltransferase/ultrastructure , HEK293 Cells , Humans , Inflammation/immunology , Models, Molecular , Multienzyme Complexes/chemistry , Multienzyme Complexes/genetics , Mutation , Protein Binding , Protein Multimerization , Protein Structure, Quaternary , Pyridoxal Phosphate/metabolism
7.
Arch Razi Inst ; 74(1): 1-6, 2019 03.
Article in English | MEDLINE | ID: mdl-31013002

ABSTRACT

Brucella bacterium causes Brucellosis, an infectious disease spreading from animals to human. Brucella lumazine synthase (BLS) is a highly immunogenic protein with adjuvant properties, which has been introduced as an effective protein carrier for vaccine development. This protein also plays a significant role in inducing immune system. This study aimed to clone, express, and purify the BLS gene from Brucella melitensis Rev1. The BLS gene was amplified by particular primers with the restriction enzyme sites as a linker and it was inserted into pTZ57R/T vector. Subsequently, it was ligated into pET32(a)+ expression vector. Recombinant expression vector containing coding sequence of BLS was transformed into E. coli BL21 (DE3) host gene expression and stimulated by 0.1mM IPTG. The results of sequencing showed that there were not any mutations in BLS encoding sequence. The expression results were set by sequencing and endorsed using sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) analyses and western blotting that showed 35 kDa protein band appropriately.


Subject(s)
Bacterial Proteins/immunology , Brucella Vaccine/immunology , Brucella melitensis/immunology , Brucellosis/veterinary , Genes, Bacterial , Multienzyme Complexes/immunology , Blotting, Western/veterinary , Brucellosis/prevention & control , Electrophoresis, Polyacrylamide Gel/veterinary , Escherichia coli/genetics , Microorganisms, Genetically-Modified/genetics , Recombinant Proteins/immunology , Vaccines, Subunit/immunology
8.
J Pharm Sci ; 107(9): 2283-2296, 2018 09.
Article in English | MEDLINE | ID: mdl-29763607

ABSTRACT

Lumazine synthase (LS) is a family of enzyme involved in the penultimate step in the biosynthesis of riboflavin. Its enzymatic mechanism has been well defined, and many LS structures have been solved using X-ray crystallography or cryoelectron microscopy. LS is composed of homooligomers, which vary in size and subunit number, including pentamers, decamers, and icosahedral sixty-mers, depending on its species of origin. Research on LS has expanded beyond the initial focus on its enzymatic function to properties related to its oligomeric structure and exceptional conformational stability. These attributes of LS systems have now been repurposed for use in various biomedical fields. This review primarily focuses on the applications of LS as a flexible vaccine presentation system. Presentation of antigens on the surface of LS results in a high local concentration of antigens displayed in an ordered array. Such repetitive structures enable the cross-linking of B-cell receptors and result in strong immune responses through an avidity effect. Potential issues with the use of this system and corresponding solutions are also discussed with the objective of improved utilization of the LS system in vaccine development.


Subject(s)
Drug Delivery Systems/trends , Multienzyme Complexes/administration & dosage , Multienzyme Complexes/immunology , Riboflavin Synthase/administration & dosage , Riboflavin Synthase/immunology , Animals , Drug Delivery Systems/methods , Humans , Immunogenicity, Vaccine/immunology , Multienzyme Complexes/chemistry , Protein Structure, Secondary , Riboflavin Synthase/chemistry
9.
Gene ; 662: 1-9, 2018 Jul 01.
Article in English | MEDLINE | ID: mdl-29627526

ABSTRACT

Proteinuria is a common adverse effect of renal injury in preeclampsia. To explore the effects of AKR1C3 in renal injury due to preeclampsia and to determine an optimal method to prevent proteinuria, glibenclamide treatment was used in unrestrained Wistar rats exposed to N-nitro-L-arginine methyl ester hydrochloride (L-NAME). Successful rat models for preeclampsia were confirmed based on mean arterial pressure, a 24-h protein urine test, and by observing the structure of the kidney by transmission electron microscopy (TEM). Forty Wistar rats were randomly divided into L-NAME-induced preeclampsia (pregnant and L-NAME), treatment (pregnant, L-NAME, and glybenclamide), non-pregnant (L-NAME), and control (pregnant and 0.9% saline) groups. Rats that were 19 days into their pregnancies were then euthanized and their kidneys were collected. After exposure to L-NAME, the mean arterial pressure increased by ~25 mmHg, which was largely prevented by the co-administration of glibenclamide. At 24 h, protein levels in the urine of the L-NAME-induced preeclampsia group were higher than those of the control and treatment groups. AKR1C3 was downregulated in the kidney and podocytes, whereas glibenclamide increased the expression of AKR1C3. The generation of reactive oxygen species (ROS) detected by ELISA was decreased by the glibenclamide co-administration. Compared with that in the L-NAME-induced preeclampsia group, the expression levels of AKR1C3 protein and mRNA significantly increased in the treatment group ([0.48 ±â€¯0.09] vs.[1.05 ±â€¯0.20];[0.05 ±â€¯0.02] vs.[0.22 ±â€¯0.06]; both P < 0.05]). Therefore, AKR1C3 expression was decreased in the kidneys of L-NAME-induced preeclampsia rats, and glibenclamide may be useful for the treatment of preeclampsia by regulating the generation of ROS and preventing proteinuria.


Subject(s)
Acute Kidney Injury/metabolism , Aldo-Keto Reductase Family 1 Member C3/metabolism , Glyburide/pharmacology , Multienzyme Complexes/metabolism , Pre-Eclampsia/drug therapy , Pre-Eclampsia/metabolism , Acute Kidney Injury/enzymology , Aldo-Keto Reductase Family 1 Member C3/immunology , Animals , Anti-Inflammatory Agents/pharmacology , Disease Models, Animal , Female , Microscopy, Electron, Transmission/methods , Multienzyme Complexes/immunology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/metabolism , Pre-Eclampsia/immunology , Pregnancy , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism
10.
Trop Anim Health Prod ; 50(5): 957-963, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29492808

ABSTRACT

Brucella poses a great threat to animal and human health. Vaccination is the most promising strategy in the effort to control Brucella abortus (B. abortus) infection, but the currently used live vaccines interfere with diagnostic tests and could potentially result in disease outbreak. Therefore, new subunit vaccines and combined immunization strategies are currently under investigation. In this study, immunogenicity and protection ability of a recombinant adenovirus and plasmid DNA vaccine co-expressing P39 and lumazine synthase proteins of B. abortus were evaluated based on the construction of the two molecular vaccines. Four immunization strategies (single adenovirus, single DNA, adenovirus/DNA, DNA/adenovirus) were investigated. The results showed that the immunization strategy of DNA priming followed by adenovirus boosting induced robust humoral and cellular immune responses, and it significantly reduced the numbers of B. abortus in a mouse model. These results suggest that it could be a potential antigen candidate for development of a new subunit vaccine against B. abortus infection.


Subject(s)
Brucella Vaccine/immunology , Brucellosis/immunology , Multienzyme Complexes/immunology , Vaccines, DNA/immunology , Adenoviridae , Animals , Antibodies, Bacterial/blood , Bacterial Proteins/immunology , Brucella abortus/immunology , Brucellosis/prevention & control , Brucellosis, Bovine/immunology , Cattle , Cell Proliferation , Cytokines/immunology , Female , Mice , Mice, Inbred BALB C
11.
Methods Mol Biol ; 1732: 203-213, 2018.
Article in English | MEDLINE | ID: mdl-29480477

ABSTRACT

The 5'-AMP-activated protein kinase is a complicated enzyme consisting of three different subunits, each of which is expressed as two or three isoforms. This gives the possibility of 12 different heterotrimeric complexes, which could have diverse functions within the cell. To map out which of these complexes are present and to what extent in skeletal muscle, we have used the immunoprecipitation technique and analyzed both the precipitates and the remaining supernatants for coprecipitation of complex partners. We have fine-tuned this method to give the best results on lysates from the skeletal muscle, liver, and heart muscle from mouse to man.


Subject(s)
AMP-Activated Protein Kinases/chemistry , Immunoprecipitation/methods , Multienzyme Complexes/chemistry , Muscle, Skeletal/metabolism , Protein Subunits/chemistry , AMP-Activated Protein Kinases/immunology , AMP-Activated Protein Kinases/metabolism , Animals , Humans , Immunoprecipitation/instrumentation , Liver/metabolism , Mice , Multienzyme Complexes/immunology , Multienzyme Complexes/metabolism , Myocardium/metabolism , Protein Subunits/immunology , Protein Subunits/metabolism
12.
Microb Pathog ; 112: 148-155, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28916316

ABSTRACT

Brucellosis is a globally distributed zoonotic disease that causes animal and human diseases. Although effective, the current Brucella vaccines (strain M5-90 or others) have several drawbacks. The first is their residual virulence for animals and humans and the second is their inability to differentiate natural infection from that caused by vaccination. In the present study, Brucella melitensis M5-90 manB mutant (M5-90ΔmanB) was generated to overcome these drawbacks. M5-90ΔmanB showed significantly reduced survival in macrophages and mice, and induced strong protective immunity in BALB/c mice. It elicited anti-Brucella-specific IgG1 and IgG2a subtype responses and induced the secretion of gamma interferon (IFN-γ) and interleukin-4(IL-4). Results of immune assays showed, M5-90ΔmanB immunization induced the secretion of IFN-γ in goats, and serum samples from goats inoculated with M5-90ΔmanB were negative by Bengal Plate Test (RBPT) and Standard Tube Agglutination Test (STAT). Further, the ManB antigen also allows serological assays differentiate infections caused by wild strains from infections by vaccination. These results show that M5-90ΔmanB is a suitable attenuated vaccine candidate against virulent Brucella melitensis 16 M (16 M) infection.


Subject(s)
Brucella Vaccine/immunology , Brucella melitensis/immunology , Brucellosis/immunology , Brucellosis/prevention & control , Immunization , Vaccines, Attenuated/immunology , Animals , Antibodies, Bacterial/blood , Bacterial Proteins/blood , Bacterial Proteins/immunology , Base Sequence , Brucella Vaccine/genetics , Brucella melitensis/enzymology , Brucella melitensis/genetics , Brucella melitensis/growth & development , Brucellosis/microbiology , DNA, Bacterial/genetics , Disease Models, Animal , Female , Gene Deletion , Immunoglobulin G/blood , Interferon-gamma/metabolism , Interleukin-4/metabolism , Macrophages/immunology , Macrophages/microbiology , Mannose-6-Phosphate Isomerase/blood , Mannose-6-Phosphate Isomerase/immunology , Mice, Inbred BALB C , Multienzyme Complexes/blood , Multienzyme Complexes/immunology , Nucleotidyltransferases/blood , Nucleotidyltransferases/immunology , Vaccination , Vaccines, Attenuated/genetics
13.
Steroids ; 127: 56-61, 2017 11.
Article in English | MEDLINE | ID: mdl-28863887

ABSTRACT

The human 3ß-hydroxysteroid dehydrogenase/isomerase (HSD3B) enzymes catalyze the conversion of 3ß-hydroxy Δ5-6 steroids into 3-keto Δ4-5 steroids, which is required for the synthesis of the mature steroid hormones secreted by the adrenal and gonads. The human has 2 isozymes, the HSD3B1 that is traditionally located in placenta and extra-adrenal tissues and the HSD3B2 that is expressed in the adrenal and gonads. Mice with both cryptochrome 1 and 2 genes deletion were recently found to have salt-sensitive hypertension and hyperaldosteronism. These deletions were also associated with overexpression of the Hsd3b6 enzyme, the homolog of the human HSD3B1, in the zona glomerulosa which was believed to explain the hyperaldosteronism. A report using antibodies against human HSD3B1 suggested that it was expressed in the zona glomerulosa of normal human adrenals and in patients with idiopathic hyperaldosteronism and the HSD3B2 expressed in both the zona fasciculata and glomerulosa. We have developed specific monoclonal antibodies against the human HSD3B1 and HSD3B2 isozymes and found that the main enzyme expressed in the zona glomerulosa was the HSD3B2. Faint staining of the adrenal was also obtained using the anti-HSD3B1antibody only at high concentrations of antibody. This study fails to confirm that HSD3B1 expression in the human zona glomerulosa and double immunofluorescence clearly shows that the HSD3B2 is expressed in the zona glomerulosa and fasciculata and in the zona glomerulosa HSD3B2 is co-expressed with aldosterone synthase (CYP11B2).


Subject(s)
Antibodies, Monoclonal/immunology , Multienzyme Complexes/immunology , Progesterone Reductase/immunology , Steroid Isomerases/immunology , Amino Acid Sequence , Animals , CHO Cells , Cell Line , Cricetulus , Gene Expression Regulation, Enzymologic , Humans , Multienzyme Complexes/chemistry , Multienzyme Complexes/metabolism , Progesterone Reductase/chemistry , Progesterone Reductase/metabolism , Steroid Isomerases/chemistry , Steroid Isomerases/metabolism , Zona Glomerulosa/metabolism
14.
Protein Sci ; 26(10): 2059-2072, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28736824

ABSTRACT

Polyvalent antigen display is an effective strategy to enhance the immunogenicity of subunit vaccines by clustering them in an array-like manner on a scaffold system. This strategy results in a higher local density of antigens, increased high avidity interactions with B cells and other antigen presenting cells, and therefore a more effective presentation of vaccine antigens. In this study, we used lumazine synthase (LS), an icosahedral symmetry capsid derived from Bacillus anthracis, as a scaffold to present 60 copies of a linear B cell epitope (PB10) from the ricin toxin fused to the C terminus of LS via four different linkers. We then investigated the effects of linker length, linker rigidity and formaldehyde crosslinking on the protein assembly, conformational integrity, thermal stability, in vitro antibody binding, and immunogenicity in mice. Fusion of the PB10 peptide onto LS, with varying linker lengths, did not affect protein assembly, thermal stability or exposure of the epitope, but had a minor impact on protein conformation. Formaldehyde crosslinking considerably improved protein thermal stability with only minor impact on protein conformation. All LS_PB10 constructs, when administered to mice by injection without adjuvant, elicited measurable anti-ricin serum IgG titers, although the titers were not sufficient to confer protection against a 10× lethal dose ricin challenge. This work sheds light on the biophysical properties, immunogenicity and potential feasibility of LS from B. anthracis as a scaffold system for polyvalent antigen display.


Subject(s)
Anthrax Vaccines , Antigens, Bacterial , Bacillus anthracis , Epitopes, B-Lymphocyte , Multienzyme Complexes , Vaccines, Subunit , Animals , Anthrax Vaccines/chemistry , Anthrax Vaccines/genetics , Anthrax Vaccines/immunology , Anthrax Vaccines/metabolism , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Antigens, Bacterial/chemistry , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Antigens, Bacterial/metabolism , Bacillus anthracis/enzymology , Bacillus anthracis/immunology , Epitopes, B-Lymphocyte/chemistry , Epitopes, B-Lymphocyte/genetics , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/metabolism , Female , Immunoglobulin G/blood , Immunoglobulin G/immunology , Mice , Models, Molecular , Multienzyme Complexes/chemistry , Multienzyme Complexes/genetics , Multienzyme Complexes/immunology , Multienzyme Complexes/metabolism , Protein Stability , Ricin/chemistry , Ricin/genetics , Ricin/immunology , Ricin/metabolism , Vaccines, Subunit/chemistry , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology , Vaccines, Subunit/metabolism
15.
Protein Sci ; 26(5): 1049-1059, 2017 05.
Article in English | MEDLINE | ID: mdl-28257593

ABSTRACT

Aiming to combine the flexibility of Brucella lumazine synthase (BLS) to adapt different protein domains in a decameric structure and the capacity of BLS and flagellin to enhance the immunogenicity of peptides that are linked to their structure, we generated a chimeric protein (BLS-FliC131) by fusing flagellin from Salmonella in the N-termini of BLS. The obtained protein was recognized by anti-flagellin and anti-BLS antibodies, keeping the oligomerization capacity of BLS, without affecting the folding of the monomeric protein components determined by circular dichroism. Furthermore, the thermal stability of each fusion partner is conserved, indicating that the interactions that participate in its folding are not affected by the genetic fusion. Besides, either in vitro or in vivo using TLR5-deficient animals we could determine that BLS-FliC131 retains the capacity of triggering TLR5. The humoral response against BLS elicited by BLS-FliC131 was stronger than the one elicited by equimolar amounts of BLS + FliC. Since BLS scaffold allows the generation of hetero-decameric structures, we expect that flagellin oligomerization on this protein scaffold will generate a new vaccine platform with enhanced capacity to activate immune responses.


Subject(s)
Brucella , Flagellin , Multienzyme Complexes , Recombinant Fusion Proteins , Salmonella typhimurium , Animals , Brucella/enzymology , Brucella/genetics , Brucella/immunology , Caco-2 Cells , Female , Flagellin/biosynthesis , Flagellin/genetics , Flagellin/immunology , Humans , Immunity, Humoral , Mice , Mice, Knockout , Multienzyme Complexes/biosynthesis , Multienzyme Complexes/genetics , Multienzyme Complexes/immunology , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Salmonella typhimurium/genetics , Salmonella typhimurium/immunology , Salmonella typhimurium/metabolism , Toll-Like Receptor 5/genetics , Toll-Like Receptor 5/immunology
16.
Vaccine ; 34(39): 4732-4737, 2016 09 07.
Article in English | MEDLINE | ID: mdl-27527816

ABSTRACT

Shiga toxin producing Escherichia coli (STEC) are bacterial pathogens involved in food-borne diseases. Shiga toxin (Stx) is the main virulence factor of STEC and is responsible for systemic complications including Hemolytic Uremic Syndrome (HUS). It has been previously demonstrated that Shiga toxin type 2 (Stx2) induces pregnancy loss in rats in early stage of pregnancy. The main purpose of this study was to determine if an active immunization prevents Stx2 mediated pregnancy loss and confers passive protective immunity to the offspring. For that purpose Sprague Dawley female rats were immunized with the chimera based on the enzyme lumazine synthase from Brucella spp. (BLS) and the B subunit of Shiga toxin 2 (Stx2B) named BLS-Stx2B. After immunization females were mated with males. At day 8 of gestation, dams were challenged intraperitoneally with a sublethal and abortifacient dose of Stx2. The immunization induced high anti-Stx2B-specific antibody titers in sera and most important, prevented pregnancy loss. Pups born and breastfeed by immunized dams had high anti-Stx2B-specific antibody titers in sera. Cross-fostering experiments indicated that passive protective immunity against Stx2 was transmitted through lactation. These results indicate that immunization of adult female rats with BLS-Stx2B prevents Stx2-induced pregnancy loss and confers anti Stx2 protective immunity to the offspring.


Subject(s)
Abortion, Spontaneous/prevention & control , Escherichia coli Infections/prevention & control , Escherichia coli Vaccines/immunology , Immunity, Maternally-Acquired , Shiga Toxin 2/immunology , Abortion, Spontaneous/microbiology , Animals , Antibodies, Bacterial/blood , Antibodies, Neutralizing/blood , Brucella/enzymology , Female , Foodborne Diseases/prevention & control , Hemolytic-Uremic Syndrome/prevention & control , Male , Multienzyme Complexes/immunology , Pregnancy , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/immunology , Shiga-Toxigenic Escherichia coli
17.
Clin Exp Immunol ; 186(2): 164-176, 2016 11.
Article in English | MEDLINE | ID: mdl-27414259

ABSTRACT

Autoimmune hepatitis (AIH) is a chronic liver disease characterized by progressive inflammation, female preponderance and seropositivity for autoantibodies such as anti-smooth muscle actin and/or anti-nuclear, anti-liver kidney microsomal type 1 (anti-LKM1) and anti-liver cytosol type 1 (anti-LC1) in more than 80% of cases. AIH is linked strongly to several major histocompatibility complex (MHC) alleles, including human leucocyte antigen (HLA)-DR3, -DR7 and -DR13. HLA-DR4 has the second strongest association with adult AIH, after HLA-DR3. We investigated the role of HLA-DR4 in the development of AIH by immunization of HLA-DR4 (DR4) transgenic non-obese diabetic (NOD) mice with DNA coding for human CYP2D6/FTCD fusion autoantigen. Immunization of DR4 mice leads to sustained mild liver injury, as assessed biochemically by elevated alanine aminotransferase, histologically by interface hepatitis, plasma cell infiltration and mild fibrosis and immunologically by the development of anti-LKM1/anti-LC1 antibodies. In addition, livers from DR4 mice had fewer regulatory T cells (Tregs ), which had decreased programmed death (PD)-1 expression. Splenic Tregs from these mice also showed impaired inhibitory capacity. Furthermore, DR4 expression enhanced the activation status of CD8+ T cells, macrophages and dendritic cells in naive DR4 mice compared to naive wild-type (WT) NOD mice. Our results demonstrate that HLA-DR4 is a susceptibility factor for the development of AIH. Impaired suppressive function of Tregs and reduced PD-1 expression may result in spontaneous activation of key immune cell subsets, such as antigen-presenting cells and CD8+ T effectors, facilitating the induction of AIH and persistent liver damage.


Subject(s)
HLA-DR4 Antigen/genetics , HLA-DR4 Antigen/immunology , Hepatitis, Autoimmune/etiology , Hepatitis, Autoimmune/pathology , Ammonia-Lyases , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Autoantibodies/immunology , Autoantigens/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cytochrome P-450 CYP2D6/genetics , Cytochrome P-450 CYP2D6/immunology , Cytokines/metabolism , Disease Models, Animal , Glutamate Formimidoyltransferase , Humans , Hypergammaglobulinemia/immunology , Immunization , Immunoglobulin G/immunology , Inflammation Mediators/metabolism , Mice , Mice, Inbred NOD , Mice, Transgenic , Multienzyme Complexes/genetics , Multienzyme Complexes/immunology , Multifunctional Enzymes , Plasma Cells/immunology , Plasma Cells/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
18.
Vet Parasitol ; 224: 13-19, 2016 Jul 15.
Article in English | MEDLINE | ID: mdl-27270384

ABSTRACT

The liver fluke Fasciola hepatica remains an important agent of food-borne trematode disease producing great economic losses due to its negative effect on productivity of livestock grazing in temperate areas. The prevailing control strategy based on anthelmintic drugs is unsustainable due to widespread resistance hence vaccination appears as an attractive option to pursue. In this study we evaluate the effect of vaccination in calves with a functional recombinant thioredoxin glutathione reductase (rFhTGR) from liver fluke, a critical antioxidant enzyme at the crossroads of the thioredoxin and glutathione metabolism in flatworms. The recombinant enzyme produced in Escherichia coli was tested in two vaccination experiments; in the first trial rFhTGR was administered in combination with Freund́s Incomplete Adjuvant (FIA) in a three-inoculation scheme on weeks 0, 4 and 8; in the second trial rFhTGR was given mixed with Adyuvac 50 or Alum as adjuvants on weeks 0 and 4. In both cases calves were challenged with metacercariae (400 in the first and 500 in the second trial) 2 weeks after the last inoculation. Our results demonstrate that two or three doses of the vaccine induced a non-significant reduction in worm counts of 8.2% (FIA), 3.8% (Adyuvac 50) and 23.0% (Alum) compared to adjuvant controls indicating that rFhTGR failed to induce a protective immunity in challenged calves. All vaccine formulations induced a mixed IgG1/IgG2 response but no booster was observed after challenge. No correlations between antibody titres and worm burdens were found.


Subject(s)
Antigens, Helminth/immunology , Cattle Diseases/prevention & control , Fascioliasis/veterinary , Immunization/veterinary , Multienzyme Complexes/immunology , NADH, NADPH Oxidoreductases/immunology , Adjuvants, Immunologic , Animals , Cattle , Fasciola hepatica/enzymology , Fasciola hepatica/immunology , Fascioliasis/prevention & control , Immunization/standards , Parasite Load , Recombinant Proteins/immunology
19.
Nat Commun ; 6: 8819, 2015 Nov 09.
Article in English | MEDLINE | ID: mdl-26549640

ABSTRACT

Interferon gamma (IFNγ)-producing CD8(+) T cells (Tc1) play important roles in immunological disease. We now report that CD3/CD28-mediated stimulation of CD8(+) T cells to generate Tc1 cells, not only increases IFNγ production but also boosts the generation of reactive oxygen species (ROS) and augments expression of CD39. Inhibition of NADPH oxidases or knockdown of gp91phox in CD8(+) T cells abrogates ROS generation, which in turn modulates JNK and NFκB signalling with decreases in both IFNγ levels and CD39 expression. CD39(+)CD8(+) T cells substantially inhibit IFNγ production by CD39(-)CD8(+) T cells via the paracrine generation of adenosine, which is operational via adenosine type 2A receptors. Increases in numbers of CD39(+)CD8(+) T cells and associated enhancements in ROS signal transduction are noted in cells from patients with Crohn's disease. Our findings provide insights into Tc1-mediated IFNγ responses and ROS generation and link these pathways to CD39/adenosine-mediated effects in immunological disease.


Subject(s)
Adenosine/metabolism , Antigens, CD/immunology , Apyrase/immunology , Colon/immunology , Crohn Disease/immunology , Interferon-gamma/immunology , Leukocytes, Mononuclear/immunology , Multienzyme Complexes/immunology , NADH, NADPH Oxidoreductases/immunology , NADPH Oxidases/immunology , T-Lymphocytes, Cytotoxic/immunology , Adult , Aged , Antigens, CD/metabolism , Apyrase/metabolism , Blotting, Western , CD28 Antigens , CD3 Complex , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Chromatin Immunoprecipitation , Colon/cytology , Female , Flow Cytometry , Humans , Immunosuppressive Agents , Interferon-gamma/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , MAP Kinase Signaling System/immunology , Male , Middle Aged , Multienzyme Complexes/metabolism , NADH, NADPH Oxidoreductases/metabolism , NADPH Oxidases/metabolism , NF-kappa B/immunology , NF-kappa B/metabolism , Reactive Oxygen Species , Receptors, Purinergic P1/metabolism , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes, Cytotoxic/metabolism , Young Adult
20.
PLoS One ; 10(5): e0126827, 2015.
Article in English | MEDLINE | ID: mdl-25973756

ABSTRACT

Brucella Lumazine Synthase (BLS) is a highly immunogenic decameric protein which can accept the fusion of foreign proteins at its ten N-termini. These chimeras are very efficient to elicit systemic and oral immunity without adjuvants. BLS signaling via Toll-Like Receptor 4 (TLR4) regulates innate and adaptive immune responses, inducing dendritic cell maturation and CD8(+) T-cell cytotoxicity. In this work we study the effect induced by BLS in TLR4-expressing B16 melanoma. In order to evaluate the effectiveness of BLS as a preventive vaccine, C57BL/6J mice were immunized with BLS or BLS-OVA, and 35 days later were subcutaneously inoculated with B16-OVA melanoma. BLS or BLS-OVA induced a significant inhibition of tumor growth, and 50% of mice immunized with the highest dose of BLS did not develop visible tumors. This effect was not observed in TLR4-deficient mice. For treatment experiments, mice were injected with BLS or BLS-OVA 2 days after the inoculation of B16 cells. Both treatments induced significant and equal tumor growth delay and increased survival. Moreover, BLS and BLS-OVA stimulation were also effective in TLR4-deficient mice. In order to study whether BLS has a direct effect on tumor cells, B16 cells were preincubated with BLS, and after 48h, cells were inoculated. Tumors induced by BLS-stimulated cells had inhibited growth and survival was increased. In the BLS group, 40% of mice did not develop tumors. This effect was abolished by the addition of TLR4/MD2 blocking antibody to cells before BLS stimulation. Our work demonstrates that BLS immunization induces a preventive antitumor response that depends on mice TLR4. We also show that BLS generates a therapeutic effect in mice inoculated with B16 cells. Our results show that BLS acts directly in cultured tumor cells via TLR4, highly suggesting that BLS elicits its therapeutic effects acting on the TLR4 from B16 melanoma cells.


Subject(s)
Brucella/enzymology , Multienzyme Complexes/metabolism , Toll-Like Receptor 4/genetics , Animals , Apoptosis , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Melanoma, Experimental/drug therapy , Melanoma, Experimental/mortality , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Multienzyme Complexes/genetics , Multienzyme Complexes/immunology , Ovalbumin/genetics , Ovalbumin/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/therapeutic use , Recombinant Proteins/biosynthesis , Recombinant Proteins/immunology , Recombinant Proteins/therapeutic use , Survival Rate , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/metabolism , Transplantation, Homologous
SELECTION OF CITATIONS
SEARCH DETAIL
...