Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Int Immunopharmacol ; 133: 112120, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38657497

ABSTRACT

Despite the efforts of global programme to eliminate lymphatic filariasis (GPELF), the threat of lymphatic filariasis (LF) still looms over humanity in terms of long-term disabilities, and morbidities across the globe. In light of this situation, investigators have chosen to focus on the development of immunotherapeutics targeting the physiologically important filarial-specific proteins. Glutaredoxin (16.43 kDa) plays a pivotal role in filarial redox biology, serving as a vital contributor. In the context of the intra-host survival of filarial parasites, this antioxidant helps in mitigating the oxidative stress imposed by the host immune system. Given its significant contribution, the development of a vaccine targeting glutaredoxin holds promise as a new avenue for achieving a filaria-free world. Herein, multi-epitope-based vaccine was designed using advanced immunoinformatics approach. Initially, 4B-cell epitopes and 6 T-cell epitopes (4 MHC I and 2 MHC II) were identified from the 146 amino acid long sequence of glutaredoxin of the human filarid, Wuchereria bancrofti. Subsequent clustering of these epitopes with linker peptides finalized the vaccine structure. To boost TLR-mediated innate immunity, TLR-specific adjuvants were incorporated into the designed vaccine. After that, experimental analyses confirm the designed vaccine, Vac4 as anefficient ligand of human TLR5 to elicit protective innate immunity against filarial glutaredoxin. Immune simulation further demonstrated abundant levels of IgG and IgM as crucial contributors in triggering vaccine-induced adaptive responses in the recipients. Hence, to facilitate the validation of immunogenicity of the designed vaccine, Vac4 was cloned in silico in pET28a(+) expression vector for recombinant production. Taken together, our findings suggest that vaccine-mediated targeting of filarial glutaredoxin could be a future option for intervening LF on a global scale.


Subject(s)
Elephantiasis, Filarial , Glutaredoxins , Wuchereria bancrofti , Glutaredoxins/immunology , Glutaredoxins/metabolism , Animals , Elephantiasis, Filarial/prevention & control , Elephantiasis, Filarial/immunology , Humans , Wuchereria bancrofti/immunology , Epitopes, T-Lymphocyte/immunology , Vaccinology/methods , Epitopes, B-Lymphocyte/immunology , Vaccines, Subunit/immunology , Mice , Antigens, Helminth/immunology , Female , Mice, Inbred BALB C
2.
Fish Shellfish Immunol ; 107(Pt A): 385-394, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33141077

ABSTRACT

Glutaredoxins (Grxs) are well-known oxidoreductases involved in a wide range of redox activities in organisms. In this study, two invertebrate Grxs (AbGrx1-like and AbGrx2) from disk abalone were identified and characterized in an effort to gain a deeper understanding into their immune and redox regulatory roles. Both AbGrxs share typical thioredoxin/Grx structures. AbGrx1-like and AbGrx2 were identified as monothiol and diothiol Grxs, respectively. AbGrxs were significantly expressed at the egg and 16-cell stage of early abalone development. Although the expression of both AbGrxs demonstrated similar patterns, the expression of AbGrx1-like was higher than AbGrx2 during development stages. In contrast, AbGrx2 expression was significantly higher than that of AbGrx1-like in adult tissues. Highest AbGrx1-like expression was observed in the hepatopancreas and digestive tract, while highest AbGrx2 expression was found in the gills, followed by the mantle, in healthy adult abalone tissues. The highest expression of AbGrx1-like was observed in the gills at 12 h and 6 h post injection (p.i) of Vibrio parahemolyticus and other stimulants, respectively. The highest expression of AbGrx2 in the gills were observed at 120 h, 6 h, 24 h, and 12 h post injection of V. parahaemolyticus, Listeria monocytogenes, Viral hemorrhagic septicemia virus, and Polyinosinic:polycytidylic acid, respectively. AbGrxs possessed significant 2-hydroxyethyl disulfide (HED) and dehydroascorbate (DHA) reduction activity, but AbGrx2 exhibited higher redox activity than AbGrx1-like. Altogether, our results suggest an important role of AbGrx1-like and AbGrx2 in redox homeostasis, as well as in the invertebrate immune defense system. Our findings will aid the development of new disease management strategies for this economically valuable species.


Subject(s)
Gastropoda/genetics , Gastropoda/immunology , Glutaredoxins/genetics , Glutaredoxins/immunology , Amino Acid Sequence , Animals , Base Sequence , Glutaredoxins/chemistry , Immunity, Innate , Oxidation-Reduction , Protein Structure, Tertiary , Sequence Alignment
3.
Fish Shellfish Immunol ; 95: 411-421, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31586678

ABSTRACT

Glutaredoxins are oxidoreductases present in almost all living organisms. They belong to the thioredoxin superfamily and share the thioredoxin structure and catalytic motif. Glutaredoxin 2 has been identified as a mitochondrial protein in vertebrates. In this study, the sequence of Glutaredoxin 2 from Hippocampus abdominalis (HaGrx2) was analyzed by molecular, transcriptional, and functional assays. In-silico analysis revealed that HaGrx2 shows the highest homology with Hippocampus comes, while distinctly cluster with fish Grx2 orthologs. Tissue distribution analysis showed that HaGrx2 is ubiquitously expressed in all tissues tested, and the highest expression was observed in the brain and skin. Significant HaGrx2 transcript modulation was identified in blood and liver upon injecting bacterial and Pathogen Associated Molecular Patterns. The redox activity of HaGrx2 was revealed by Dehydroascorbic reduction and insulin disulfide reduction activity assays. Further, the deglutathionylation activity of 1 nM HaGrx2 was found to be equivalent to that of 0.84 nM HaGrx1. HaGrx2 exhibited antiapoptotic activity against H2O2-induced oxidative stress in FHM cells. Altogether, the results of this study suggest that HaGrx2 plays a role in redox homeostasis and innate immune responses in fish.


Subject(s)
Fish Diseases/immunology , Gene Expression Regulation/immunology , Glutaredoxins/genetics , Glutaredoxins/immunology , Immunity, Innate/genetics , Smegmamorpha/genetics , Smegmamorpha/immunology , Amino Acid Sequence , Animals , Base Sequence , Edwardsiella tarda/physiology , Enterobacteriaceae Infections/immunology , Enterobacteriaceae Infections/veterinary , Female , Fish Proteins/chemistry , Fish Proteins/genetics , Fish Proteins/immunology , Gene Expression Profiling/veterinary , Glutaredoxins/chemistry , Homeostasis , Lipopolysaccharides/adverse effects , Male , Oxidation-Reduction , Phylogeny , Poly I-C/adverse effects , Sequence Alignment/veterinary , Streptococcal Infections/immunology , Streptococcal Infections/veterinary , Streptococcus iniae/physiology
4.
Fish Shellfish Immunol ; 90: 40-51, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31015065

ABSTRACT

Glutaredoxins (Grx) are redox enzymes conserved in viruses, eukaryotes, and prokaryotes. In this study, we characterized glutaredoxin 1 (HaGrx1) from big-belly seahorse, Hippocampus abdominalis. In-silico analysis showed that HaGrx1 contained the classical glutaredoxin 1 structure with a CSYC thioredoxin active site motif. According to multiple sequence alignment and phylogenetic reconstruction, HaGrx1 presented the highest homology to the Grx1 ortholog from Hippocampus comes. Transcriptional studies demonstrated the ubiquitous distribution of HaGrx1 transcripts in all the seahorse tissues tested. Significant modulation (p < 0.05) of HaGrx1 transcripts were observed in blood upon stimulation with pathogen-associated molecular patterns and live pathogens. The ß-hydroxyethyl disulfide reduction assay confirmed the antioxidant activity of recombinant HaGrx1. Further, dehydroascorbate reduction and insulin disulfide reduction assays revealed the oxidoreductase activity of HaGrx1. HaGrx1 utilized 1,4-dithiothreitol, l-cysteine, 2-mercaptoethanol, and reduced l-glutathione as reducing agent with different dehydroascorbate reduction activity levels. Altogether, our results suggested a vital role of HaGrx1 in redox homeostasis as well as the host innate immune defense system.


Subject(s)
Fish Diseases/immunology , Gene Expression Regulation/immunology , Glutaredoxins/genetics , Glutaredoxins/immunology , Immunity, Innate/genetics , Smegmamorpha/genetics , Smegmamorpha/immunology , Amino Acid Sequence , Animals , Base Sequence , Edwardsiella tarda/physiology , Enterobacteriaceae Infections/immunology , Enterobacteriaceae Infections/veterinary , Fish Proteins/chemistry , Fish Proteins/genetics , Fish Proteins/immunology , Gene Expression Profiling/veterinary , Glutaredoxins/chemistry , Lipopolysaccharides/pharmacology , Pathogen-Associated Molecular Pattern Molecules , Phylogeny , Poly I-C/pharmacology , Sequence Alignment/veterinary , Streptococcal Infections/immunology , Streptococcal Infections/veterinary , Streptococcus iniae/physiology
5.
J Biol Chem ; 293(13): 4893-4900, 2018 03 30.
Article in English | MEDLINE | ID: mdl-29414783

ABSTRACT

Inflammasomes are cytosolic complexes that mature and secrete the inflammatory cytokines interleukin 1ß (IL-1ß) and IL-18 and induce pyroptosis. The NLRP3 (NACHT, LRR, and PYD domains-containing protein 3) inflammasome detects many pathogen- and danger-associated molecular patterns, and reactive oxygen species (ROS)/reactive nitrogen species (RNS) have been implicated in its activation. The phenazine pyocyanin (PCN) is a virulence factor of Pseudomonas aeruginosa and generates superoxide in cells. Here we report that PCN inhibits IL-1ß and IL-18 release and pyroptosis upon NLRP3 inflammasome activation in macrophages by preventing speck formation and Caspase-1 maturation. Of note, PCN did not regulate the AIM2 (absent in melanoma 2) or NLRC4 inflammasomes or tumor necrosis factor (TNF) secretion. Imaging of the fluorescent glutathione redox potential sensor Grx1-roGFP2 indicated that PCN provokes cytosolic and nuclear but not mitochondrial redox changes. PCN-induced intracellular ROS/RNS inhibited the NLRP3 inflammasome posttranslationally, and hydrogen peroxide or peroxynitrite alone were sufficient to block its activation. We propose that cytosolic ROS/RNS inhibit the NLRP3 inflammasome and that PCN's anti-inflammatory activity may help P. aeruginosa evade immune recognition.


Subject(s)
Inflammasomes/immunology , Macrophages/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , Pyocyanine/immunology , Reactive Nitrogen Species/immunology , Reactive Oxygen Species/immunology , Animals , Apoptosis Regulatory Proteins/immunology , Calcium-Binding Proteins/immunology , Caspase 1/immunology , Cell Line , DNA-Binding Proteins/immunology , Glutaredoxins/immunology , Immune Evasion , Interleukin-18/immunology , Interleukin-1beta/immunology , Macrophages/microbiology , Macrophages/pathology , Mice , Pseudomonas Infections/pathology
6.
J Cell Biol ; 216(12): 4073-4090, 2017 12 04.
Article in English | MEDLINE | ID: mdl-29150539

ABSTRACT

The antimicrobial defense activity of neutrophils partly depends on their ability to form neutrophil extracellular traps (NETs), but the underlying mechanism controlling NET formation remains unclear. We demonstrate that inhibiting cytoskeletal dynamics with pharmacological agents or by genetic manipulation prevents the degranulation of neutrophils and mitochondrial DNA release required for NET formation. Wiskott-Aldrich syndrome protein-deficient neutrophils are unable to polymerize actin and exhibit a block in both degranulation and DNA release. Similarly, neutrophils with a genetic defect in NADPH oxidase fail to induce either actin and tubulin polymerization or NET formation on activation. Moreover, neutrophils deficient in glutaredoxin 1 (Grx1), an enzyme required for deglutathionylation of actin and tubulin, are unable to polymerize either cytoskeletal network and fail to degranulate or release DNA. Collectively, cytoskeletal dynamics are achieved as a balance between reactive oxygen species-regulated effects on polymerization and glutathionylation on the one hand and the Grx1-mediated deglutathionylation that is required for NET formation on the other.


Subject(s)
Cytoskeleton/immunology , Extracellular Traps/immunology , Glutathione/immunology , Neutrophils/immunology , Reactive Oxygen Species/immunology , Actins/genetics , Actins/immunology , Animals , Cell Degranulation/drug effects , Cell Degranulation/immunology , Cytoskeleton/ultrastructure , DNA, Mitochondrial/immunology , DNA, Mitochondrial/metabolism , Extracellular Traps/chemistry , Extracellular Traps/drug effects , Gene Expression Regulation , Glutaredoxins/genetics , Glutaredoxins/immunology , Glutathione/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Homeodomain Proteins/immunology , Humans , Mice , Mice, Transgenic , NADPH Oxidases/genetics , NADPH Oxidases/immunology , Neutrophils/cytology , Neutrophils/drug effects , Oxidation-Reduction , Primary Cell Culture , Reactive Oxygen Species/metabolism , Signal Transduction , Tubulin/genetics , Tubulin/immunology , Wiskott-Aldrich Syndrome Protein/deficiency , Wiskott-Aldrich Syndrome Protein/genetics , Wiskott-Aldrich Syndrome Protein/immunology
7.
Immunity ; 37(6): 1037-49, 2012 Dec 14.
Article in English | MEDLINE | ID: mdl-23159440

ABSTRACT

The regulation of actin dynamics is pivotal for cellular processes such as cell adhesion, migration, and phagocytosis and thus is crucial for neutrophils to fulfill their roles in innate immunity. Many factors have been implicated in signal-induced actin polymerization, but the essential nature of the potential negative modulators are still poorly understood. Here we report that NADPH oxidase-dependent physiologically generated reactive oxygen species (ROS) negatively regulate actin polymerization in stimulated neutrophils via driving reversible actin glutathionylation. Disruption of glutaredoxin 1 (Grx1), an enzyme that catalyzes actin deglutathionylation, increased actin glutathionylation, attenuated actin polymerization, and consequently impaired neutrophil polarization, chemotaxis, adhesion, and phagocytosis. Consistently, Grx1-deficient murine neutrophils showed impaired in vivo recruitment to sites of inflammation and reduced bactericidal capability. Together, these results present a physiological role for glutaredoxin and ROS- induced reversible actin glutathionylation in regulation of actin dynamics in neutrophils.


Subject(s)
Actins/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Reactive Oxygen Species/metabolism , Animals , Bacterial Infections/genetics , Bacterial Infections/immunology , Cells, Cultured , Chemotaxis/immunology , Glutaredoxins/genetics , Glutaredoxins/immunology , Humans , Mice , Mice, Knockout , NADPH Oxidases/metabolism , Protein Binding , Pseudopodia/metabolism
8.
Free Radic Biol Med ; 51(6): 1249-57, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21762778

ABSTRACT

The transcription factor nuclear factor κB (NF-κB) is a critical regulator of inflammation and immunity and is negatively regulated via S-glutathionylation. The inhibitory effect of S-glutathionylation is overcome by glutaredoxin-1 (Grx1), which under physiological conditions catalyzes deglutathionylation and enhances NF-κB activation. The mechanisms whereby expression of the Glrx1 gene is regulated remain unknown. Here we examined the role of NF-κB in regulating activation of Glrx1. Transgenic mice that express a doxycycline-inducible constitutively active version of inhibitory κB kinase-ß (CA-IKKß) demonstrate elevated expression of Grx1. Transient transfection of CA-IKKß also resulted in significant induction of Grx1. A 2-kb region of the Glrx1 promoter that contains two putative NF-κB binding sites was activated by CA-IKKß, RelA/p50, and lipopolysaccharide (LPS). Chromatin immunoprecipitation experiments confirmed binding of RelA to the promoter of Glrx1 in response to LPS. Stimulation of C10 lung epithelial cells with LPS caused transient increases in Grx1 mRNA expression and time-dependent increases in S-glutathionylation of IKKß. Overexpression of Grx1 decreased S-glutathionylation of IKKß, prolonged NF-κB activation, and increased levels of proinflammatory mediators. Collectively, this study demonstrates that the Glrx1 gene is positively regulated by NF-κB and suggests a feed-forward mechanism to promote NF-κB signaling by decreasing S-glutathionylation.


Subject(s)
Epithelial Cells/metabolism , Glutaredoxins/metabolism , NF-kappa B/metabolism , Animals , Cell Line , Epithelial Cells/immunology , Epithelial Cells/pathology , Gene Expression Regulation/immunology , Glutaredoxins/genetics , Glutaredoxins/immunology , I-kappa B Kinase/genetics , Immunization , Inflammation Mediators/metabolism , Lipopolysaccharides/immunology , Lipopolysaccharides/metabolism , Lung/pathology , Mice , Mice, Transgenic , NF-kappa B/genetics , Promoter Regions, Genetic/genetics , Signal Transduction/genetics , Transcription Factor RelA/metabolism , Transcriptional Activation/genetics , Transgenes/genetics
9.
Biochim Biophys Acta ; 1810(1): 2-92, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20682242

ABSTRACT

BACKGROUND: Oxidoreductases of the thioredoxin family of proteins have been thoroughly studied in numerous cellular and animal models mimicking human diseases. Despite of their well documented role in various disease conditions, no systematic information on the presence of these proteins is available. METHODS: Here, we have systematically analyzed the presence of some of the major constituents of the glutaredoxin (Grx)-, peroxiredoxin (Prx)-, and thioredoxin (Trx)-systems, i.e. Grx1, Grx2, Grx3 (TXNL-2/PICOT), Grx5, nucleoredoxin (Nrx), Prx1, Prx2, Prx3, Prx4, Prx5, Prx6, Trx1, thioredoxin reductase 1 (TrxR1), Trx2, TrxR2, and γ-glutamyl cysteine synthetase (γ-GCS) in various tissues of the mouse using immunohistochemistry. RESULTS: The identification of the Trx family proteins in the central nervous system, sensory organs, digestive system, lymphatic system, reproductive system, urinary system, respiratory system, endocrine system, skin, heart, and muscle revealed a number of significant differences between these proteins with respect to their distribution in these tissues. CONCLUSION: Our results imply more specific functions and interactions between the proteins of this family than previously assumed. GENERAL SIGNIFICANCE: Crucial functions of Trx family proteins have been demonstrated in various disease conditions. A detailed overview on their distribution in various tissues will be helpful to fully comprehend their potential role and the interactions of these proteins in the most thoroughly studied model for human diseases-the laboratory mouse. This article is part of a Special Issue entitled Human and Murine Redox Protein Atlases.


Subject(s)
Glutaredoxins/metabolism , Mice/metabolism , Peroxiredoxins/metabolism , Thioredoxins/metabolism , Animals , Atlases as Topic , Female , Glutaredoxins/genetics , Glutaredoxins/immunology , Humans , Immunohistochemistry , Male , Mice/genetics , Mice/immunology , Models, Biological , Oxidation-Reduction , Peroxiredoxins/genetics , Peroxiredoxins/immunology , Pregnancy , Thioredoxins/genetics , Thioredoxins/immunology , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...