Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
STAR Protoc ; 5(1): 102888, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38358882

ABSTRACT

Monosodium urate (MSU) crystal deposition in articular joints and bursal tissue causes acute joint inflammation, which is a hallmark of gout. Here, we describe the steps necessary to create a subcutaneous air pouch on the back of mice that resembles this bursa-like space with a synovial lining-like membrane. We then detail the injection of MSU crystals into this pouch, which induces a localized inflammatory response reminiscent of gout and approaches to quantify the inflammatory response. For complete details on the use and execution of this protocol, please refer to Devi et al. (2023),1 de Almeida et al. (2022),2 and Ratsimandresy et al. (2017).3.


Subject(s)
Gout , Uric Acid , Mice , Animals , Uric Acid/adverse effects , Uric Acid/chemistry , Gout/chemically induced
2.
Sci Adv ; 10(9): eadi2742, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38416822

ABSTRACT

Androgen receptor (AR) drives prostate cancer (PC) growth and progression, and targeting AR signaling is the mainstay of pharmacological therapies for PC. Resistance develops relatively fast as a result of refueled AR activity. A major gap in the field is the lack of understanding of targetable mechanisms that induce persistent AR expression in castrate-resistant PC (CRPC). This study uncovers an unexpected function of active Stat5 signaling, a known promoter of PC growth and clinical progression, as a potent inducer of AR gene transcription. Stat5 suppression inhibited AR gene transcription in preclinical PC models and reduced the levels of wild-type, mutated, and truncated AR proteins. Pharmacological Stat5 inhibition by a specific small-molecule Stat5 inhibitor down-regulated Stat5-inducible genes as well as AR and AR-regulated genes and suppressed PC growth. This work introduces the concept of Stat5 as an inducer of AR gene transcription in PC. Pharmacological Stat5 inhibitors may represent a new strategy for suppressing AR and CRPC growth.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Receptors, Androgen , Male , Humans , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism , Signal Transduction , Transcription, Genetic , Cell Line, Tumor , Gene Expression Regulation, Neoplastic
3.
Methods Mol Biol ; 2696: 55-71, 2023.
Article in English | MEDLINE | ID: mdl-37578715

ABSTRACT

Protein oligomerization is a common principle of regulating cellular responses. Oligomerization of NLRs is essential for the formation of NLR signaling platforms and can be detected by several biochemical techniques. Some of these biochemical methods can be combined with functional assays, such as caspase-1 activity assay. Size exclusion chromatography (SEC) allows separation of native protein lysates into different sized complexes by FPLC for follow-up analysis. Using co-immunoprecipitation (co-IP), combined with SEC or on its own, enables subsequent antibody-based purification of NLR complexes and associated proteins, which can then be analyzed by immunoblot and/or subjected to functional caspase-1 activity assay. Native gel electrophoresis also allows detection of the NLR oligomerization state by immunoblot. Chemical cross-linking covalently joins two or more molecules, thus capturing the oligomeric state with high sensitivity and stability. ASC oligomerization has been successfully used as readout for NLR/ALR inflammasome activation in response to various PAMPs and DAMPs in human and mouse macrophages and THP-1 cells. Here, we provide a detailed description of the methods used for NLRP7 oligomerization in response to infection with Staphylococcus aureus (S. aureus) in primary human macrophages, co-immunoprecipitation, and immunoblot analysis of NLRP7 and NLRP3 inflammasome complexes as well as caspase-1 activity assays. Also, ASC oligomerization is shown in response to dsDNA, LPS/ATP, and LPS/nigericin in mouse bone marrow-derived macrophages (BMDMs) and/or THP-1 cells or human primary macrophages.


Subject(s)
Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Mice , Animals , Humans , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Staphylococcus aureus/metabolism , Lipopolysaccharides , Chromatography, Gel , Immunoprecipitation , Caspases/metabolism , Caspase 1/metabolism , Interleukin-1beta/metabolism , Adaptor Proteins, Signal Transducing/metabolism
4.
Cell Rep ; 42(3): 112265, 2023 03 28.
Article in English | MEDLINE | ID: mdl-36930645

ABSTRACT

Inflammatory responses are crucial for controlling infections and initiating tissue repair. However, excessive and uncontrolled inflammation causes inflammatory disease. Processing and release of the pro-inflammatory cytokines interleukin-1ß (IL-1ß) and IL-18 depend on caspase-1 activation within inflammasomes. Assembly of inflammasomes is initiated upon activation of cytosolic pattern recognition receptors (PRRs), followed by sequential polymerization of pyrin domain (PYD)-containing and caspase recruitment domain (CARD)-containing proteins mediated by homotypic PYD and CARD interactions. Small PYD- or CARD-only proteins (POPs and COPs, respectively) evolved in higher primates to target these crucial interactions to limit inflammation. Here, we show the ability of COPs to regulate inflammasome activation by modulating homotypic CARD-CARD interactions in vitro and in vivo. CARD16, CARD17, and CARD18 displace crucial CARD interactions between caspase-1 proteins through competitive binding and ameliorate uric acid crystal-mediated NLRP3 inflammasome activation and inflammatory disease. COPs therefore represent an important family of inflammasome regulators and ameliorate inflammatory disease.


Subject(s)
Gout , Inflammasomes , Animals , Inflammasomes/metabolism , Inflammation/metabolism , Caspase 1/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Interleukin-1beta/metabolism , CARD Signaling Adaptor Proteins/metabolism
5.
Arch Microbiol ; 204(11): 684, 2022 Nov 01.
Article in English | MEDLINE | ID: mdl-36318321

ABSTRACT

Inflammatory bowel disease includes ulcerative colitis and Crohn's disease, and is globally increasing. An appropriate model system is required to dissect the disease pathogenesis and drugs screening for adequate treatment. In the present study, we established a novel model of gut inflammation by injecting peptidoglycan from Staphylococcus aureus using laparotomic procedure. For this, three different doses of peptidoglycan, i.e., 2.5, 5 and 10 mg/kg body weight were used. The treatment effect was evaluated by studying the macrophage phagocytic function, spleen lymphocytes' proliferation and qRT-PCR for the assessment of peritoneal cells' gene expression. In addition, histological analysis of gut sections, gastric acidity, immunoglobulins and cytokines were assessed. There was significant increase in phagocytic activity in 10 mg/kg body weight PGN group. A dose dependent increase in spleen lymphocytes' proliferation and a significant increase in total acid secretion in 5 and 10 mg/kg body weight PGN treated rats were observed. In addition, a significant increment in TLR-2 and CD-14 mRNA expression in peritoneal cells, TNF-α, IL-6 and IFN-γ level and maximum distortion of gut architecture was observed in 10 mg/kg body weight PGN group. Hence, peptidoglycan from S. aureus can be used for establishing the screening model to study the action and mechanism of anti-inflammatory food products and drugs.


Subject(s)
Peptidoglycan , Staphylococcal Infections , Rats , Animals , Peptidoglycan/metabolism , Staphylococcus aureus/metabolism , Cytokines/genetics , Body Weight
6.
Front Immunol ; 13: 912069, 2022.
Article in English | MEDLINE | ID: mdl-36225929

ABSTRACT

Canonical inflammasomes are innate immune protein scaffolds that enable the activation of inflammatory caspase-1, and subsequently the processing and release of interleukin (IL)-1ß, IL-18, and danger signals, as well as the induction of pyroptotic cell death. Inflammasome assembly and activation occurs in response to sensing of infectious, sterile and self-derived molecular patterns by cytosolic pattern recognition receptors, including the Nod-like receptor NLRP3. While these responses are essential for host defense, excessive and uncontrolled NLRP3 inflammasome responses cause and contribute to a wide spectrum of inflammatory diseases, including gout. A key step in NLRP3 inflammasome assembly is the sequentially nucleated polymerization of Pyrin domain (PYD)- and caspase recruitment domain (CARD)-containing inflammasome components. NLRP3 triggers polymerization of the adaptor protein ASC through PYD-PYD interactions, but ASC polymerization then proceeds in a self-perpetuating manner and represents a point of no return, which culminates in the activation of caspase-1 by induced proximity. In humans, small PYD-only proteins (POPs) lacking an effector domain regulate this key process through competitive binding, but limited information exists on their physiological role during health and disease. Here we demonstrate that POP1 expression in macrophages is sufficient to dampen MSU crystal-mediated inflammatory responses in animal models of gout. Whether MSU crystals are administered into a subcutaneous airpouch or into the ankle joint, the presence of POP1 significantly reduces neutrophil infiltration. Also, airpouch exudates have much reduced IL-1ß and ASC, which are typical pro-inflammatory indicators that can also be detected in synovial fluids of gout patients. Exogenous expression of POP1 in mouse and human macrophages also blocks MSU crystal-induced NLRP3 inflammasome assembly, resulting in reduced IL-1ß and IL-18 secretion. Conversely, reduced POP1 expression in human macrophages enhances IL-1ß secretion. We further determined that the mechanism for the POP1-mediated inhibition of NLRP3 inflammasome activation is through its interference with the crucial NLRP3 and ASC interaction within the inflammasome complex. Strikingly, administration of an engineered cell permeable version of POP1 was able to ameliorate MSU crystal-mediated inflammation in vivo, as measured by neutrophil infiltration. Overall, we demonstrate that POP1 may play a crucial role in regulating inflammatory responses in gout.


Subject(s)
Gout , Inflammasomes , Ribonucleoproteins/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Caspase 1/metabolism , Gout/metabolism , Humans , Inflammasomes/metabolism , Interleukin-18/metabolism , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
7.
Nat Immunol ; 23(6): 892-903, 2022 06.
Article in English | MEDLINE | ID: mdl-35624206

ABSTRACT

Intracellular sensing of stress and danger signals initiates inflammatory innate immune responses by triggering inflammasome assembly, caspase-1 activation and pyroptotic cell death as well as the release of interleukin 1ß (IL-1ß), IL-18 and danger signals. NLRP3 broadly senses infectious patterns and sterile danger signals, resulting in the tightly coordinated and regulated assembly of the NLRP3 inflammasome, but the precise mechanisms are incompletely understood. Here, we identified NLRP11 as an essential component of the NLRP3 inflammasome in human macrophages. NLRP11 interacted with NLRP3 and ASC, and deletion of NLRP11 specifically prevented NLRP3 inflammasome activation by preventing inflammasome assembly, NLRP3 and ASC polymerization, caspase-1 activation, pyroptosis and cytokine release but did not affect other inflammasomes. Restored expression of NLRP11, but not NLRP11 lacking the PYRIN domain (PYD), restored inflammasome activation. NLRP11 was also necessary for inflammasome responses driven by NLRP3 mutations that cause cryopyrin-associated periodic syndrome (CAPS). Because NLRP11 is not expressed in mice, our observations emphasize the specific complexity of inflammasome regulation in humans.


Subject(s)
Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Animals , Caspase 1/genetics , Caspases/metabolism , Humans , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Licensure , Macrophages , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
8.
Int J Mol Sci ; 21(18)2020 Sep 20.
Article in English | MEDLINE | ID: mdl-32962268

ABSTRACT

Inflammasomes are protein scaffolds required for the activation of caspase-1 and the subsequent release of interleukin (IL)-1ß, IL-18, and danger signals, as well as the induction of pyroptotic cell death to restore homeostasis following infection and sterile tissue damage. However, excessive inflammasome activation also causes detrimental inflammatory disease. Therefore, extensive control mechanisms are necessary to prevent improper inflammasome responses and inflammatory disease. Inflammasomes are assembled by sequential nucleated polymerization of Pyrin domain (PYD) and caspase recruitment domain (CARD)-containing inflammasome components. Once polymerization is nucleated, this process proceeds in a self-perpetuating manner and represents a point of no return. Therefore, regulation of this key step is crucial for a controlled inflammasome response. Here, we provide an update on two single domain protein families containing either a PYD or a CARD, the PYD-only proteins (POPs) and CARD-only proteins (COPs), respectively. Their structure allows them to occupy and block access to key protein-protein interaction domains necessary for inflammasome assembly, thereby regulating the threshold of these nucleated polymerization events, and consequently, the inflammatory host response.


Subject(s)
CARD Signaling Adaptor Proteins/metabolism , Inflammasomes/metabolism , Protein Multimerization , CARD Signaling Adaptor Proteins/genetics , Caspase 1/genetics , Caspase 1/metabolism , Humans , Inflammasomes/genetics , Inflammation/genetics , Inflammation/metabolism , Interleukin-18/genetics , Interleukin-18/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Pyrin Domain
9.
Mol Cancer Ther ; 19(1): 231-246, 2020 01.
Article in English | MEDLINE | ID: mdl-31548294

ABSTRACT

The second-generation antiandrogen, enzalutamide, is approved for castrate-resistant prostate cancer (CRPC) and targets androgen receptor (AR) activity in CRPC. Despite initial clinical activity, acquired resistance to enzalutamide arises rapidly and most patients develop terminal disease. Previous work has established Stat5 as a potent inducer of prostate cancer growth. Here, we investigated the significance of Jak2-Stat5 signaling in resistance of prostate cancer to enzalutamide. The levels of Jak2 and Stat5 mRNA, proteins and activation were evaluated in prostate cancer cells, xenograft tumors, and clinical prostate cancers before and after enzalutamide therapy. Jak2 and Stat5 were suppressed by genetic knockdown using lentiviral shRNA or pharmacologic inhibitors. Responsiveness of primary and enzalutamide-resistant prostate cancer to pharmacologic inhibitors of Jak2-Stat5 signaling was assessed in vivo in mice bearing prostate cancer xenograft tumors. Patient-derived prostate cancers were tested for responsiveness to Stat5 blockade as second-line treatment after enzalutamide ex vivo in tumor explant cultures. Enzalutamide-liganded AR induces sustained Jak2-Stat5 phosphorylation in prostate cancer leading to the formation of a positive feed-forward loop, where activated Stat5, in turn, induces Jak2 mRNA and protein levels contributing to further Jak2 activation. Mechanistically, enzalutamide-liganded AR induced Jak2 phosphorylation through a process involving Jak2-specific phosphatases. Stat5 promoted prostate cancer growth during enzalutamide treatment. Jak2-Stat5 inhibition induced death of prostate cancer cells and patient-derived prostate cancers surviving enzalutamide treatment and blocked enzalutamide-resistant tumor growth in mice. This work introduces a novel concept of a pivotal role of hyperactivated Jak2-Stat5 signaling in enzalutamide-resistant prostate cancer, which is readily targetable by Jak2 inhibitors in clinical development.


Subject(s)
Janus Kinase 2/antagonists & inhibitors , Phenylthiohydantoin/analogs & derivatives , Prostatic Neoplasms, Castration-Resistant/drug therapy , Animals , Benzamides , Humans , Male , Mice , Mice, Nude , Nitriles , Phenylthiohydantoin/pharmacology , Phenylthiohydantoin/therapeutic use , Prostatic Neoplasms, Castration-Resistant/pathology , Signal Transduction , Xenograft Model Antitumor Assays
10.
J Cell Biochem ; 120(6): 9677-9691, 2019 06.
Article in English | MEDLINE | ID: mdl-30592315

ABSTRACT

Prolonged passaging of primary fibroblast cells totally shapes the natural biological phenomena and leads to the appearance of features related to senescence. As a result, it is a good natural tool to delineate the molecular mechanism of cellular aging. The present investigation revealed the antiaging effect of milk-derived novel bioactive peptide (VLPVPQK). The peptide played an important role in downregulating apoptosis-related markers in late passages of cultured fibroblast cells. The peptide treatment to aged fibroblasts caused enhancement in cell migration, DNA integrity, and decrease in the lipid peroxidation, reactive oxygen species, nitric oxide production as well as pro-inflammatory cytokines, TNF-α and IL-6. Moreover, the peptide decreased the expression of apoptotic caspases, Bax, and senescence-associated ß-galactosidase (SA-ß-gal) proteins. The peptide pretreatment also enhanced the extracellular collagen protein and antiapoptotic, Bcl-xL. In addition, the peptide treatment reversed the senescence-related activity in fibroblasts by stimulating Nrf2 mediated antioxidative defense system and inhibiting the action of NFkB/p38MAPK signaling, similar to the commercially available inhibitor (SB203580) of p38MAPK. Thus, the peptide exhibits the antiaging effect in dermal fibroblast cells.


Subject(s)
Cellular Senescence/drug effects , Fibroblasts/metabolism , MAP Kinase Signaling System/drug effects , Milk Proteins/chemistry , Milk/chemistry , NF-E2-Related Factor 2/metabolism , Peptides/pharmacology , Animals , Peptides/chemistry , Rats
11.
Exp Toxicol Pathol ; 69(7): 485-495, 2017 Sep 05.
Article in English | MEDLINE | ID: mdl-28483348

ABSTRACT

Oxidative stress is one of a critical pathogenic factor in the progression of aging and chronic diseases such as cancer, myocardial inflammation and diabetes. In the present scenario, peptides with short half life and more biological specificities are gaining much attention as prodrugs. Thus, the present investigation carried out to screen potential antioxidative peptide, VLPVPQK to cope with the cellular oxidative damage. Our results showed that treatment of rat fibroblast cells with 0.2mM H2O2 for 6h significantly declined different oxidative stress biomarkers such as SOD, CAT, GSH, and promoted LDH activity. In addition, ROS and TNF-α levels were also increased upon H2O2 exposure for 6h and thereby, it induced cell death. Amazingly, pretreatment of the peptide (VLPVPQK) significantly elevated cell survivability, by reversing all H2O2 induced alterations in fibroblast cells. Therefore, our results indicated that, the peptide (VLPVPQK) acted as a potential cytoprotective agent, who restored redox balance and cell homeostasis in cultured fibroblast cells, even after H2O2 exposure, suggesting that the peptide can be valuable as an effective remedy in treatment of oxidative stress related diseases and skin inflammation related disorders.


Subject(s)
Caseins/pharmacology , Fibroblasts/drug effects , Oxidative Stress/drug effects , Animals , Buffaloes , Hydrogen Peroxide/toxicity , Necrosis , Oxidants/toxicity , Peptides/pharmacology , Rats
12.
Appl Environ Microbiol ; 83(1)2017 01 01.
Article in English | MEDLINE | ID: mdl-27815276

ABSTRACT

Some life-threatening, foodborne, and zoonotic infections are transmitted through poultry birds. Inappropriate and indiscriminate use of antimicrobials in the livestock industry has led to an increased prevalence of multidrug-resistant bacteria with epidemic potential. Here, we present a functional molecular epidemiological analysis entailing the phenotypic and whole-genome sequence-based characterization of 11 H. pullorum isolates from broiler and free-range chickens sampled from retail wet markets in Hyderabad City, India. Antimicrobial susceptibility tests revealed all of the isolates to be resistant to multiple antibiotic classes such as fluoroquinolones, cephalosporins, sulfonamides, and macrolides. The isolates were also found to be extended-spectrum ß-lactamase producers and were even resistant to clavulanic acid. Whole-genome sequencing and comparative genomic analysis of these isolates revealed the presence of five or six well-characterized antimicrobial resistance genes, including those encoding a resistance-nodulation-division efflux pump(s). Phylogenetic analysis combined with pan-genome analysis revealed a remarkable degree of genetic diversity among the isolates from free-range chickens; in contrast, a high degree of genetic similarity was observed among broiler chicken isolates. Comparative genomic analysis of all publicly available H. pullorum genomes, including our isolates (n = 16), together with the genomes of 17 other Helicobacter species, revealed a high number (8,560) of H. pullorum-specific protein-encoding genes, with an average of 535 such genes per isolate. In silico virulence screening identified 182 important virulence genes and also revealed high strain-specific gene content in isolates from free-range chickens (average, 34) compared to broiler chicken isolates. A significant prevalence of prophages (ranging from 1 to 9) and a significant presence of genomic islands (0 to 4) were observed in free-range and broiler chicken isolates. Taken together, these observations provide significant baseline data for functional molecular infection epidemiology of nonpyloric Helicobacter species such as H. pullorum by unraveling their evolution in chickens and their possible zoonotic transmission to humans. IMPORTANCE: Globally, the poultry industry is expanding with an ever-growing consumer base for chicken meat. Given this, food-associated transmission of multidrug-resistant bacteria represents an important health care issue. Our study involves a critical baseline approach directed at genome sequence-based epidemiology and transmission dynamics of H. pullorum, a poultry pathogen having established zoonotic potential. We believe our studies would facilitate the development of surveillance systems that ensure the safety of food for humans and guide public health policies related to the use of antibiotics in animal feed in countries such as India. We sequenced 11 new genomes of H. pullorum as a part of this study. These genomes would provide much value in addition to the ongoing comparative genomic studies of helicobacters.


Subject(s)
Chickens/microbiology , DNA, Bacterial/genetics , Drug Resistance, Multiple, Bacterial , Genome, Bacterial , Helicobacter Infections/veterinary , Helicobacter/genetics , Poultry Diseases/microbiology , Animals , Anti-Bacterial Agents/pharmacology , Cephalosporins/pharmacology , Fluoroquinolones/pharmacology , Food Microbiology , Genomic Islands , Helicobacter/drug effects , Helicobacter/isolation & purification , Helicobacter Infections/epidemiology , Helicobacter Infections/microbiology , High-Throughput Nucleotide Sequencing , Humans , India/epidemiology , Microbial Sensitivity Tests , Molecular Epidemiology , Phylogeny , Poultry Diseases/epidemiology , Prophages/genetics , Prophages/isolation & purification , beta-Lactamases/biosynthesis , beta-Lactamases/genetics
13.
Drug Des Devel Ther ; 10: 3611-3632, 2016.
Article in English | MEDLINE | ID: mdl-27853354

ABSTRACT

Continuous usage of synthetic chemotherapeutic drugs causes adverse effects, which prompted for the development of alternative therapeutics for gastric cancer from natural source. This study was carried out with a specific aim to screen gastroprotective compounds from the fruits of Syzygium alternifolium (Myrtaceae). Three flavonoids, namely, 1) 5-hydroxy-7,4'-dimethoxy-6,8-di-C-methylflavone, 2) kaempferol-3-O-ß-d-glucopyranoside, and 3) kaempferol-3-O-α-l-rhamnopyranoside were isolated from the above medicinal plant by employing silica gel column chromatography and are characterized by NMR techniques. Antigastric cancer activity of these flavonoids was examined on AGS cell lines followed by cell cycle progression assay. In addition, pharmacophore-based screening and molecular dynamics of protein-ligand complex were carried out to identify potent scaffolds. The results showed that compounds 2 and 3 exhibited significant cytotoxic effect, whereas compound 1 showed moderate effect on AGS cells by inhibiting G2/M phase of cell cycle. Molecular docking analysis revealed that compound 2 has higher binding energies on human growth factor receptor-2 (HER2). The constructed pharmacophore models reveal that the compounds have more number of H-bond Acc/Don features which contribute to the inhibition of HER2 activity. By selecting these features, 34 hits were retrieved using the query compound 2. Molecular dynamic simulations (MDS) of protein-ligand complexes demonstrated conspicuous inhibition of HER2 as evidenced by dynamic trajectory analysis. Based on these results, the compound ZINC67903192 was identified as promising HER2 inhibitor against gastric cancer. The present work provides a basis for the discovery a new class of scaffolds from natural products for gastric carcinoma.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Flavonoids/pharmacology , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Kinase Inhibitors/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Stomach Neoplasms/drug therapy , Syzygium/chemistry , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/isolation & purification , Antineoplastic Agents, Phytogenic/metabolism , Binding Sites , Cell Line, Tumor , Cell Proliferation/drug effects , Chromatography, Gel , Dose-Response Relationship, Drug , Drug Discovery/methods , Flavonoids/chemistry , Flavonoids/isolation & purification , Flavonoids/metabolism , Fruit , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Inhibitory Concentration 50 , Magnetic Resonance Spectroscopy , Molecular Targeted Therapy , Phytotherapy , Plants, Medicinal , Protein Binding , Protein Conformation , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/isolation & purification , Protein Kinase Inhibitors/metabolism , Quantitative Structure-Activity Relationship , Receptor, ErbB-2/chemistry , Receptor, ErbB-2/metabolism , Signal Transduction/drug effects , Stomach Neoplasms/enzymology , Stomach Neoplasms/pathology
14.
Nucleic Acids Res ; 44(19): 9393-9412, 2016 Nov 02.
Article in English | MEDLINE | ID: mdl-27550181

ABSTRACT

Helicobacter pylori portrays a classical paradigm of persistent bacterial infections. A well balanced homeostasis of bacterial effector functions and host responses is purported to be the key in achieving long term colonization in specific hosts. H. pylori nucleases have been shown to assist in natural transformation, but their role in virulence and colonization remains elusive. Therefore, it is imperative to understand the involvement of these nucleases in the pathogenesis of H. pylori Here, we report the multifaceted role of a TNFR-1 interacting endonuclease A (TieA) from H. pylori. tieA expression is differentially regulated in response to environmental stress and post adherence to gastric epithelial cells. Studies with isogenic knockouts of tieA revealed it to be a secretory protein which translocates into the host gastric epithelial cells independent of a type IV secretion system, gets phosphorylated by DNA-PK kinase and auto-phosphorylates as serine kinase. Furthermore, TieA binds to and cleaves DNA in a non-specific manner and promotes Fas mediated apoptosis in AGS cells. Additionally, TieA induced pro-inflammatory cytokine secretion via activation of transcription factor AP-1 and signaled through MAP kinase pathway. Collectively, TieA with its multipronged and moonlighting functions could facilitate H. pylori in maintaining a balance of bacterial adaptation, and elimination by the host responses.


Subject(s)
Bacterial Proteins/metabolism , Endonucleases/metabolism , Helicobacter Infections/microbiology , Helicobacter pylori/enzymology , Antibodies, Bacterial/immunology , Apoptosis/genetics , Bacterial Adhesion , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Bacterial Proteins/isolation & purification , Cell Line, Tumor , Cluster Analysis , Endonucleases/genetics , Endonucleases/immunology , Endonucleases/isolation & purification , Gastric Mucosa/immunology , Gastric Mucosa/metabolism , Gastric Mucosa/microbiology , Gene Deletion , Gene Expression Profiling , Gene Expression Regulation, Bacterial , Genome, Bacterial , Helicobacter Infections/genetics , Helicobacter Infections/immunology , Helicobacter pylori/immunology , Host-Pathogen Interactions , Humans , Hydrogen-Ion Concentration , Hydrolysis , Immune Sera/immunology , Immunity, Innate , Macrophages/immunology , Macrophages/metabolism , Mitogen-Activated Protein Kinases/metabolism , Mutation , Protein Binding , Type IV Secretion Systems , fas Receptor/metabolism
15.
Sci Rep ; 5: 15049, 2015 Oct 12.
Article in English | MEDLINE | ID: mdl-26456705

ABSTRACT

Evasion of innate immune recognition is one of the key strategies for persistence of Helicobacter pylori, by virtue of its ability to modulate or escape the host innate immune receptors and signaling pathways. C-type lectin receptors (CLRs) predominantly expressed by macrophages are pivotal in tailoring immune response against pathogens. The recognition of glyco or carbohydrate moieties by Mincle (Macrophage inducible C-type lectin) is emerging as a crucial element in anti-fungal and anti-mycobacterial immunity. Herein, we demonstrate the role of Mincle in modulation of innate immune response against H. pylori infection. Our results revealed an upregulated expression of Mincle which was independent of direct host cell contact. Upon computational modelling, Mincle was observed to interact with the Lewis antigens of H. pylori LPS and possibly activating an anti-inflammatory cytokine production, thereby maintaining a balance between pro- and anti-inflammatory cytokine production. Furthermore, siRNA mediated knockdown of Mincle in human macrophages resulted in up regulation of pro-inflammatory cytokines and consequent down regulation of anti-inflammatory cytokines. Collectively, our study demonstrates a novel mechanism employed by H. pylori to escape clearance by exploiting functional plasticity of Mincle to strike a balance between pro-and anti-inflammatory responses ensuring its persistence in the host.


Subject(s)
Helicobacter pylori/immunology , Immune Evasion , Lectins, C-Type/immunology , Lipopolysaccharides/immunology , Macrophages/microbiology , Receptors, Immunologic/immunology , Amino Acid Sequence , Binding Sites , Cell Line , Gene Expression Regulation , Humans , Interleukin-10/biosynthesis , Interleukin-10/immunology , Lectins, C-Type/antagonists & inhibitors , Lectins, C-Type/genetics , Lipopolysaccharides/chemistry , Macrophage Activation , Macrophages/immunology , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Interaction Domains and Motifs , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Immunologic/antagonists & inhibitors , Receptors, Immunologic/genetics , Sequence Alignment , Signal Transduction , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/immunology
16.
Int J Med Microbiol ; 304(8): 1066-76, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25172221

ABSTRACT

The Helicobacter pylori gene JHP0940 has been shown to encode a serine/threonine kinase which can induce cytokines in gastric epithelial cells relevant to chronic gastric inflammation. Here we demonstrate that JHP0940 can be secreted by the bacteria, triggers apoptosis in cultured mouse macrophages and acts as an auto-phosphorylating tyrosine kinase. Recombinant JHP0940 protein was found to decrease the viability of RAW264.7 cells (a mouse macrophage cell line) up to 55% within 24h of co-incubation. The decreased cellular viability was due to apoptosis, which was confirmed by TUNEL assay and Fas expression analysis by flow-cytometry. Further, we found that caspase-1 and IL-1beta were activated upon treatment with JHP0940. These results point towards possible action through the host inflammasome. Our in vitro studies using tyrosine kinase assays further demonstrated that JHP0940 acts as auto-phosphorylating tyrosine kinase and induces pro-inflammatory cytokines in RAW264.7 cells. Upon exposure with JHP0940, these cells secreted IL-1beta, TNF-alpha and IL-6, in a dose- and time-dependent manner, as detected by ELISA and transcript profiling by q-RT-PCR. The pro-inflammatory, pro-apoptotic and other regulatory responses triggered by JHP0940 lead to the assumption of its possible role in inducing chronic inflammation for enhanced bacterial persistence and escape from host innate immune responses by apoptosis of macrophages.


Subject(s)
Apoptosis , Bacterial Proteins/metabolism , Helicobacter pylori/enzymology , Host-Pathogen Interactions , Macrophages/microbiology , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Virulence Factors/metabolism , Animals , Cell Line , Cell Survival , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Macrophages/physiology , Mice , Phosphorylation , Protein Processing, Post-Translational
17.
Cytokine ; 68(2): 110-7, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24767863

ABSTRACT

HP0986 protein of Helicobacter pylori has been shown to trigger induction of proinflammatory cytokines (IL-8 and TNF-α) through the activation of NF-κB and also to induce Fas mediated apoptosis of human macrophage cells (THP-1). In this study, we unravel mechanistic details of the biological effects of this protein in a murine macrophage environment. Up regulation of MCP-1 and TNF-α in HP0986-induced RAW 264.7 cells occurred subsequent to the activation and translocation of NF-κB to the cell nucleus. Further, HP0986 induced apoptosis of RAW 264.7 cells through Fas activation and this was in agreement with previous observations made with THP-1 cells. Our studies indicated activation of TNFR1 through interaction with HP0986 and this elicited the aforementioned responses independent of TLR2, TLR4 or TNFR2. We found that mouse TNFR1 activation by HP0986 facilitates formation of a complex comprising of TNFR1, TRADD and TRAF2, and this occurs upstream of NF-κB activation. Furthermore, FADD also forms a second complex, at a later stage, together with TNFR1 and TRADD, resulting in caspase-8 activation and thereby the apoptosis of RAW 264.7 cells. In summary, our observations reveal finer details of the functional activity of HP0986 protein in relation to its behavior in a murine macrophage cell environment. These findings reconfirm the proinflammatory and apoptotic role of HP0986 signifying it to be an important trigger of innate responses. These observations form much needed baseline data entailing future in vivo studies of the functions of HP0986 in a murine model.


Subject(s)
Apoptosis , Bacterial Proteins/metabolism , Helicobacter pylori/metabolism , Inflammation/pathology , Macrophages/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction , Animals , Cell Membrane/metabolism , Cells, Cultured , Chemokine CCL2/metabolism , Fas-Associated Death Domain Protein/metabolism , Fluorescein-5-isothiocyanate/metabolism , Humans , I-kappa B Proteins/metabolism , Immunoprecipitation , Mice , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Propidium/metabolism , Protein Binding , Proteolysis , TNF Receptor-Associated Death Domain Protein/metabolism , TNF Receptor-Associated Factor 2/metabolism , Tumor Necrosis Factor-alpha/metabolism , fas Receptor/metabolism
18.
Helicobacter ; 19(1): 26-36, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24205801

ABSTRACT

BACKGROUND: The envisaged roles and partly understood functional properties of Helicobacter pylori protein HP0986 are significant in the context of proinflammatory and or proapoptotic activities, the two important facilitators of pathogen survival and persistence. In addition, sequence analysis of this gene predicts a restriction endonuclease function which remained unknown thus far. To evaluate the role of HP0986 in gastric inflammation, we studied its expression profile using a large number of clinical isolates but a limited number of biopsies and patient sera. Also, we studied antigenic role of HP0986 in altering cytokine responses of human gastric epithelial (AGS) cells including its interaction with and localization within the AGS cells. MATERIALS AND METHODS: For in vitro expression study of HP0986, 110 H. pylori clinical isolates were cultured from patients with functional dyspepsia. For expression analysis by qRT PCR of HP0986, 10 gastric biopsy specimens were studied. HP0986 was also used to detect antibodies in patient sera. AGS cells were incubated with recombinant HP0986 to determine cytokine response and NF-κB activation. Transient transfection with HP0986 cloned in pEGFPN1 was used to study its subcellular localization or homing in AGS cells. RESULTS: Out of 110 cultured H. pylori strains, 34 (31%) were positive for HP0986 and this observation was correlated with in vitro expression profiles. HP0986 mRNA was detected in 7 of the 10 biopsy specimens. Further, HP0986 induced IL-8 secretion in gastric epithelial cells in a dose and time-dependent manner via NF-κB pathway. Serum antibodies against HP0986 were positively associated with H. pylori positive patients. Transient transfection of AGS cells revealed both cytoplasmic and nuclear localization of HP0986. CONCLUSION: HP0986 was moderately prevalent in clinical isolates and its expression profile in cultures and gastric biopsies points to its being naturally expressed. Collective observations including the induction of IL-8 via TNFR1 and NF-κB, subcellular localization, and seropositivity data point to a significant role of HP0986 in gastroduodenal inflammation. We propose to name the HP0986 gene/protein as 'TNFR1 interacting endonuclease A (TieA or tieA)'.


Subject(s)
Antigens, Bacterial/immunology , Epithelial Cells/immunology , Helicobacter pylori/immunology , Host-Pathogen Interactions , Interleukin-8/immunology , Receptors, Tumor Necrosis Factor, Type I/immunology , Virulence Factors/immunology , Antigens, Bacterial/genetics , Antigens, Bacterial/metabolism , Biopsy , Dyspepsia/microbiology , Epithelial Cells/microbiology , Female , Gene Expression Regulation, Bacterial , Helicobacter Infections/microbiology , Helicobacter pylori/genetics , Helicobacter pylori/isolation & purification , Helicobacter pylori/metabolism , Humans , Interleukin-8/metabolism , Male , Middle Aged , NF-kappa B/immunology , NF-kappa B/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Virulence Factors/genetics , Virulence Factors/metabolism
19.
Cytokine ; 64(1): 258-64, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23819907

ABSTRACT

Mycobacterium tuberculosis, the cause of tuberculosis in humans, is present approximately in one third of the world's population, mostly in a dormant state. The proteins encoded by the dormancy survival regulon (DosR regulon) are mainly responsible for survival of the bacilli in a latent form. To maintain latency, mycobacteria orchestrate a balanced interplay of different cytokines secreted by immune cells during the granulomatous stage. The function of most of the DosR regulon proteins of M. tuberculosis is unknown. In this study, we have shown that one of the DosR regulon proteins, DATIN, encoded by the gene Rv0079, can stimulate macrophages and peripheral blood mononuclear cells (PBMC) to secrete important cytokines that may be significant in granuloma formation and its maintenance. The expression level of DATIN in Mycobacterium bovis BCG was found to be upregulated in pH stress and microaerobic conditions. Computational modeling, docking and simulation study suggested that DATIN might interact with TLR2. This was further confirmed through the interaction of recombinant DATIN with TLR2 expressed by HEK293 cells. When in vitro differentiated THP-1 cells were treated with recombinant DATIN, increased secretion of TNF-α, IL-1ß and IL-8 was observed in a dose dependent manner. When differentiated THP-1 cells were infected with a modified BCG strain that overexpressed DATIN, augmented secretions of TNF-α, IL-1ß and IL-8 were observed as compared to a reference BCG strain containing empty vector. Similarly, human PBMCs when infected with M. bovis BCG that overexpressed DATIN, upregulated secretion of proinflammatory cytokines IFN-γ, TNF-α, IL-1ß and IL-8. The cytokine profiles dissected herein point to a possible role of DATIN in maintenance of latency with the help of the proinflammatory responses.


Subject(s)
Bacterial Proteins/metabolism , Macrophages/immunology , Toll-Like Receptor 2/metabolism , Cell Line , HEK293 Cells , Humans , Inflammation/immunology , Interferon-gamma/metabolism , Interleukin-1beta/metabolism , Interleukin-8/metabolism , Macrophages/metabolism , Mycobacterium bovis/immunology , Mycobacterium bovis/metabolism , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/metabolism , Tumor Necrosis Factor-alpha/metabolism
20.
PLoS One ; 6(7): e22530, 2011.
Article in English | MEDLINE | ID: mdl-21789261

ABSTRACT

Helicobacter pylori induces cytokine mediated changes in gastroduodenal pathophysiology, wherein, the activated macrophages at the sub-mucosal space play a central role in mounting innate immune response against the antigens. The bacterium gains niche through persistent inflammation and local immune-suppression causing peptic ulcer disease or chronic gastritis; the latter being a significant risk factor for the development of gastric adenocarcinoma. What favors persistence of H. pylori in the gastric niches is not clearly understood. We report detailed characterization of a functionally unknown gene (HP986), which was detected in patient isolates associated with peptic ulcer and gastric carcinoma. Expression and purification of recombinant HP986 (rHP986) revealed a novel, ∼29 kDa protein in biologically active form which associates with significant levels of humoral immune responses in diseased individuals (p<0.001). Also, it induced significant levels of TNF-α and Interleukin-8 in cultured human macrophages concurrent to the translocation of nuclear transcription factor-κB (NF-κB). Further, the rHP986 induced apoptosis of cultured macrophages through a Fas mediated pathway. Dissection of the underlying signaling mechanism revealed that rHP986 induces both TNFR1 and Fas expression to lead to apoptosis. We further demonstrated interaction of HP986 with TNFR1 through computational and experimental approaches. Independent proinflammatory and apoptotic responses triggered by rHP986 as shown in this study point to its role, possibly as a survival strategy to gain niche through inflammation and to counter the activated macrophages to avoid clearance.


Subject(s)
Apoptosis , Bacterial Proteins/metabolism , Helicobacter Infections/metabolism , Helicobacter pylori/metabolism , Inflammation Mediators/metabolism , Amino Acid Sequence , Bacterial Proteins/chemistry , Gastrointestinal Diseases/metabolism , Gastrointestinal Diseases/microbiology , Gastrointestinal Diseases/pathology , Genetic Loci/genetics , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Helicobacter pylori/isolation & purification , Humans , Immunity, Humoral , Interleukin-8/metabolism , Models, Biological , Molecular Sequence Data , NF-kappa B/metabolism , Protein Binding , Receptors, Tumor Necrosis Factor, Type I/metabolism , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Analysis, Protein , Time Factors , Tumor Necrosis Factor-alpha/metabolism , fas Receptor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...