Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 120
Filter
Add more filters











Publication year range
1.
Int J Mol Sci ; 25(17)2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39273493

ABSTRACT

Lipopolysaccharide (LPS) triggers a severe systemic inflammatory reaction in mammals, with the dimerization of TLR4/MD-2 upon LPS stimulation serving as the pivotal mechanism in the transmission of inflammatory signals. Ginsenoside Rh2 (G-Rh2), one of the active constituents of red ginseng, exerts potent anti-inflammatory activity. However, whether G-Rh2 can block the TLR4 dimerization to exert anti-inflammatory effects remains unclear. Here, we first investigated the non-cytotoxic concentration of G-Rh2 on RAW 264.7 cells, and detected the releases of pro-inflammatory cytokines in LPS-treated RAW 264.7 cells, and then uncovered the mechanisms involved in the anti-inflammatory activity of G-Rh2 through flow cytometry, fluorescent membrane localization, Western blotting, co-immunoprecipitation (Co-IP), molecular docking and surface plasmon resonance (SPR) analysis in LPS-stimulated macrophages. Our results show that G-Rh2 stimulation markedly inhibited the secretion of LPS-induced interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α) and nitric oxide (NO). Additionally, G-Rh2 blocked the binding of LPS with the membrane of RAW 264.7 cells through direct interaction with TLR4 and MD-2 proteins, leading to the disruption of the dimerization of TLR4 and MD-2, followed by suppression of the TLR4/NF-κB signaling pathway. Our results suggest that G-Rh2 acts as a new inhibitor of TLR4 dimerization and may serve as a promising therapeutic agent against inflammation.


Subject(s)
Ginsenosides , Lipopolysaccharides , Lymphocyte Antigen 96 , Toll-Like Receptor 4 , Animals , Mice , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Ginsenosides/pharmacology , Ginsenosides/chemistry , Inflammation/metabolism , Inflammation/drug therapy , Inflammation/chemically induced , Interleukin-6/metabolism , Lymphocyte Antigen 96/metabolism , Lymphocyte Antigen 96/chemistry , Macrophages/drug effects , Macrophages/metabolism , Molecular Docking Simulation , Nitric Oxide/metabolism , Protein Binding , Protein Multimerization/drug effects , RAW 264.7 Cells , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/metabolism
2.
J Chem Inf Model ; 64(13): 5253-5261, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38973303

ABSTRACT

Psychoactive substances, including morphine and methamphetamine, have been shown to interact with the classic innate immune receptor Toll-like receptor 4 (TLR4) and its partner protein myeloid differentiation protein 2 (MD2) in a nonenantioselective manner. (-)-Nicotine, the primary alkaloid in tobacco and a key component of highly addictive cigarettes, targets the TLR4/MD2, influencing TLR4 signaling pathways. Existing as two enantiomers, the stereoselective recognition of nicotine by TLR4/MD2 in the context of the innate immune response remains unclear. In this study, we synthesized (+)-nicotine and investigated its effects alongside (-)-nicotine on lipopolysaccharide (LPS)-induced TLR4 signaling. (-)-Nicotine dose-dependently inhibited proinflammatory factors such as tumor necrosis factor α (TNF-α), interleukin 6 (IL-6), and cyclooxygenase-2 (COX-2). In contrast, (+)-nicotine showed no such inhibitory effects. Molecular dynamics simulations revealed that (-)-nicotine exhibited a stronger affinity with the TLR4 coreceptor MD2 than (+)-nicotine. Additionally, in silico simulations revealed that both nicotine enantiomers initially attach to the entrance of the MD2 cavity, creating a metastable state before they fully enter the cavity. In the metastable state, (-)-nicotine established more stable interactions with the surrounding residues at the entrance of the MD2 cavity compared to those of (+)-nicotine. This highlights the crucial role of the MD2 cavity entrance in the chiral recognition of nicotine. These findings provide valuable insights into the distinct interactions between nicotine enantiomers and the TLR4 coreceptor MD2, underscoring the enantioselective effect of nicotine on modulating TLR4 signaling.


Subject(s)
Lymphocyte Antigen 96 , Molecular Dynamics Simulation , Nicotine , Signal Transduction , Toll-Like Receptor 4 , Toll-Like Receptor 4/metabolism , Nicotine/pharmacology , Nicotine/chemistry , Nicotine/analogs & derivatives , Nicotine/metabolism , Lymphocyte Antigen 96/metabolism , Lymphocyte Antigen 96/chemistry , Signal Transduction/drug effects , Stereoisomerism , Humans , Lipopolysaccharides/pharmacology , Molecular Docking Simulation , Cyclooxygenase 2/metabolism , Cyclooxygenase 2/chemistry
3.
J Chem Inf Model ; 64(13): 5273-5284, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38921627

ABSTRACT

Toll-like receptor 4 (TLR4) is pivotal as an innate immune receptor, playing a critical role in mediating neuropathic pain and drug addiction through its regulation of the neuroinflammatory response. The nonclassical (+)-opioid isomers represent a unique subset of TLR4 antagonists known for their effective blood-brain barrier permeability. Despite growing interest in the structure-activity relationship of these (+)-opioid-based TLR4 antagonists, the specific impact of heteroatoms on their TLR4 antagonistic activities has not been fully explored. This study investigated the influence of the hydroxyl group at C14 in six (+)-opioid TLR4 antagonists (1-6) using wet-lab experiments and in silico simulations. The corresponding C14-deoxy derivatives (7-12) were synthesized, and upon comparison with their corresponding counterparts (1-6), it was discovered that their TLR4 antagonistic activities were significantly diminished. Molecular dynamics simulations showed that the (+)-opioid TLR4 antagonists (1-6) possessed more negative binding free energies to the TLR4 coreceptor MD2, which was responsible for ligand recognition. This was primarily attributed to the formation of a hydrogen bond between the hydroxyl group at the C-14 position of the antagonists (1-6) and the R90 residue of MD2 during the binding process. Such an interaction facilitated the entry and subsequent binding of these molecules within the MD2 cavity. In contrast, the C14-deoxy derivatives (7-12), lacking the hydroxyl group at the C-14 position, missed this crucial hydrogen bond interaction with the R90 residue of MD2, leading to their egression from the MD2 cavity during simulations. This study underscores the significant role of the C14 hydroxyl moiety in enhancing the effectiveness of (+)-opioid TLR4 antagonists, which provides insightful guidance for designing future (+)-isomer opioid-derived TLR4 antagonists.


Subject(s)
Molecular Dynamics Simulation , Toll-Like Receptor 4 , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/metabolism , Analgesics, Opioid/chemistry , Analgesics, Opioid/pharmacology , Humans , Structure-Activity Relationship , Molecular Docking Simulation , Lymphocyte Antigen 96/antagonists & inhibitors , Lymphocyte Antigen 96/metabolism , Lymphocyte Antigen 96/chemistry
4.
Int J Biol Macromol ; 273(Pt 1): 132931, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38942665

ABSTRACT

PCP-W1, the Poria cocos polysaccharide with the strong immunomodulatory activity, was isolated through column chromatography and screened for in vitro immune activity in RAW 264.7 cells in this study. The structure analysis results revealed that the PCP-W1 were composed of galactose, glucose, fucose and mannose in a molar percentage of 35.87: 28.56: 21.77: 13.64. And it exhibited a random coil and branched conformational features with a molecular weight of 18.38 kDa. The main chain consisted of residues→3)-ß-D-Glcp-(1 â†’ 3,6)-ß-D-Glcp-(1 â†’ 3)-ß-D-Glcp-(1 â†’ 6)-ß-D-Glcp-(1 â†’ 6)-α-D-Galp-(1 â†’ 6)-α-D-Galp-(1 â†’ 2,6)-α-D-Galp-(1→6)-α-D-Galp-(1 â†’ 6)-α-D-Galp-(1 â†’ , while branching occurred at ß-D-Glcp-(1→, α-D-Manp-(1→, and α-L-Fucp-(1 â†’ 3)- α-L-Fucp-(1→. The pharmacodynamic studies demonstrated that PCP-W1 activated the release of NO, IL-6, IL-ß, TNF-α, CD86, and ROS to induce polarization of RAW 264.7 murine macrophages towards M1-type through modulation of the TLR4/MD2/NF-κB pathway. The molecular docking results showed that PCP-W1 could primarily dock onto the hydrophobic binding site of TLR4/MD2 complex via its galactose chain. Furthermore, molecular dynamics simulation displayed stable modeling for TLR4-MD2-PCP-W1 complex. Overall, we screened the most immunoactive components of the polysaccharide, analyzed its structure, demonstrated its impact on TLR4/MD2/NF-kB pathway, and studied the interaction between TLR4/MD2 and the polysaccharide fragments. These results provide further support for the structure-activity relationship study of the immunomodulatory effects of Poria cocos polysaccharide.


Subject(s)
NF-kappa B , Polysaccharides , Signal Transduction , Toll-Like Receptor 4 , Wolfiporia , Animals , Mice , Toll-Like Receptor 4/metabolism , RAW 264.7 Cells , NF-kappa B/metabolism , Polysaccharides/pharmacology , Polysaccharides/chemistry , Signal Transduction/drug effects , Wolfiporia/chemistry , Lymphocyte Antigen 96/metabolism , Lymphocyte Antigen 96/chemistry , Immunologic Factors/pharmacology , Immunologic Factors/chemistry , Molecular Docking Simulation
5.
Int J Mol Sci ; 25(10)2024 May 17.
Article in English | MEDLINE | ID: mdl-38791489

ABSTRACT

The SARS-CoV-2 Omicron variants have replaced all earlier variants, due to increased infectivity and effective evasion from infection- and vaccination-induced neutralizing antibodies. Compared to earlier variants of concern (VoCs), the Omicron variants show high TMPRSS2-independent replication in the upper airway organs, but lower replication in the lungs and lower mortality rates. The shift in cellular tropism and towards lower pathogenicity of Omicron was hypothesized to correlate with a lower toll-like receptor (TLR) activation, although the underlying molecular mechanisms remained undefined. In silico analyses presented here indicate that the Omicron spike protein has a lower potency to induce dimerization of TLR4/MD-2 compared to wild type virus despite a comparable binding activity to TLR4. A model illustrating the molecular consequences of the different potencies of the Omicron spike protein vs. wild-type spike protein for TLR4 activation is presented. Further analyses indicate a clear tendency for decreasing TLR4 dimerization potential during SARS-CoV-2 evolution via Alpha to Gamma to Delta to Omicron variants.


Subject(s)
COVID-19 , Lymphocyte Antigen 96 , Protein Multimerization , SARS-CoV-2 , Toll-Like Receptor 4 , Humans , Computer Simulation , COVID-19/virology , Lymphocyte Antigen 96/metabolism , Lymphocyte Antigen 96/genetics , Lymphocyte Antigen 96/chemistry , Protein Binding , SARS-CoV-2/genetics , SARS-CoV-2/pathogenicity , Spike Glycoprotein, Coronavirus/metabolism , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/genetics , Toll-Like Receptor 4/metabolism
6.
Chemistry ; 30(32): e202400429, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38587187

ABSTRACT

Agonists of Toll like receptors (TLRs) have attracted interest as adjuvants and immune modulators. A crystal structure of TLR4/MD2 with E. coli LPS indicates that the fatty acid at C-2 of the lipid A component of LPS induces dimerization of two TLR4-MD2 complexes, which in turn initiates cell signaling leading to the production of (pro)inflammatory cytokines. To probe the importance of the (R)-3-hydroxymyristate at C-2 of lipid A, a range of bis- and mono-phosphoryl lipid A derivatives with different modifications at C-2 were prepared by a strategy in which 2-methylnaphthyl ethers were employed as permanent protecting group that could be readily removed by catalytic hydrogenation. The C-2 amine was protected as 9-fluorenylmethyloxycarbamate, which at a later stage could be removed to give a free amine that was modified by different fatty acids. LPS and the synthetic lipid As induced the same cytokines, however, large differences in activity were observed. A compound having a hexanoyl moiety at C-2 still showed agonistic properties, but further shortening to a butanoyl abolished activity. The modifications had a larger influence on monophosphoryl lipid As. The lipid As having a butanoyl moiety at C-2 could selectively antagonize TRIF associated cytokines induced by LPS or lipid A.


Subject(s)
Cytokines , Lipid A , Lipopolysaccharides , Lipid A/chemistry , Lipid A/pharmacology , Lipid A/analogs & derivatives , Lipid A/chemical synthesis , Cytokines/metabolism , Lipopolysaccharides/pharmacology , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/chemistry , Humans , Lymphocyte Antigen 96/metabolism , Lymphocyte Antigen 96/chemistry , Drug Design , Structure-Activity Relationship , Signal Transduction/drug effects
7.
Mol Pharmacol ; 103(3): 158-165, 2023 03.
Article in English | MEDLINE | ID: mdl-36460345

ABSTRACT

Cisplatin is an effective chemotherapeutic agent, yet its use is limited by several adverse drug reactions, known as cisplatin-induced toxicities (CITs). We recently demonstrated that cisplatin could elicit proinflammatory responses associated with CITs through Toll-like receptor 4 (TLR4). TLR4 is best recognized for binding bacterial lipopolysaccharide (LPS) via its coreceptor, MD-2. TLR4 is also proposed to directly bind transition metals, such as nickel. Little is known about the nature of the cisplatin-TLR4 interaction. Here, we show that soluble TLR4 was capable of blocking cisplatin-induced, but not LPS-induced, TLR4 activation. Cisplatin and nickel, but not LPS, were able to directly bind soluble TLR4 in a microscale thermophoresis binding assay. Interestingly, TLR4 histidine variants that abolish nickel binding reduced, but did not eliminate, cisplatin-induced TLR4 activation. This was corroborated by binding data that showed cisplatin, but not nickel, could directly bind mouse TLR4 that lacks these histidine residues. Altogether, our findings suggest that TLR4 can directly bind cisplatin in a manner that is enhanced by, but not dependent on, histidine residues that facilitate binding to transition metals. SIGNIFICANCE STATEMENT: This work describes how the xenobiotic cisplatin interacts with Toll-like receptor 4 (TLR4) to initiate proinflammatory signaling that underlies cisplatin toxicities, which are severe adverse outcomes in cisplatin treatment. Here, this study provides a mechanistic bridge between cisplatin extracellular interactions with TLR4 and previous observations that genetic and chemical inhibition of TLR4 mitigates cisplatin-induced toxicity.


Subject(s)
Cisplatin , Toll-Like Receptor 4 , Animals , Mice , Allergens , Cisplatin/toxicity , Histidine , Lipopolysaccharides/pharmacology , Lymphocyte Antigen 96/chemistry , Lymphocyte Antigen 96/genetics , Lymphocyte Antigen 96/metabolism , Toll-Like Receptor 4/drug effects , Toll-Like Receptor 4/metabolism
8.
Molecules ; 26(18)2021 Sep 20.
Article in English | MEDLINE | ID: mdl-34577169

ABSTRACT

Artemisinin (also known as Qinghaosu), an active component of the Qinghao extract, is widely used as antimalarial drug. Previous studies reveal that artemisinin and its derivatives also have effective anti-inflammatory and immunomodulatory properties, but the direct molecular target remains unknown. Recently, several reports mentioned that myeloid differentiation factor 2 (MD-2, also known as lymphocyte antigen 96) may be the endogenous target of artemisinin in the inhibition of lipopolysaccharide signaling. However, the exact interaction between artemisinin and MD-2 is still not fully understood. Here, experimental and computational methods were employed to elucidate the relationship between the artemisinin and its inhibition mechanism. Experimental results showed that artemether exhibit higher anti-inflammatory activity performance than artemisinin and artesunate. Molecular docking results showed that artemisinin, artesunate, and artemether had similar binding poses, and all complexes remained stable throughout the whole molecular dynamics simulations, whereas the binding of artemisinin and its derivatives to MD-2 decreased the TLR4(Toll-Like Receptor 4)/MD-2 stability. Moreover, artemether exhibited lower binding energy as compared to artemisinin and artesunate, which is in good agreement with the experimental results. Leu61, Leu78, and Ile117 are indeed key residues that contribute to the binding free energy. Binding free energy analysis further confirmed that hydrophobic interactions were critical to maintain the binding mode of artemisinin and its derivatives with MD-2.


Subject(s)
Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Artemisinins/chemistry , Artemisinins/pharmacology , Lymphocyte Antigen 96/antagonists & inhibitors , Lymphocyte Antigen 96/chemistry , Animals , Artemether/pharmacology , Artesunate/pharmacology , Binding Sites/drug effects , Cell Line , Cell Survival/drug effects , Fatty Acid-Binding Proteins/metabolism , Hydrophobic and Hydrophilic Interactions , Immunomodulation/drug effects , In Vitro Techniques , Lipopolysaccharides/toxicity , Mice , Microglia/drug effects , Molecular Docking Simulation , Molecular Dynamics Simulation , Nitric Oxide/metabolism , Protein Binding , Thermodynamics , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/chemistry , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/metabolism
9.
Int J Mol Sci ; 22(12)2021 Jun 17.
Article in English | MEDLINE | ID: mdl-34204506

ABSTRACT

Ergosta-7, 9 (11), 22-trien-3ß-ol (EK100) was isolated from Cordyceps militaris, which has been used as a traditional anti-inflammatory medicine. EK100 has been reported to attenuate inflammatory diseases, but its anti-inflammatory mechanism is still unclear. We were the first to investigate the effect of EK100 on the Toll-like receptor 4 (TLR4)/nuclear factor of the κ light chain enhancer of B cells (NF-κB) signaling in the lipopolysaccharide (LPS)-stimulated RAW264.7 cells and the green fluorescent protein (GFP)-labeled NF-κB reporter gene of Drosophila. EK100 suppressed the release of the cytokine and attenuated the mRNA and protein expression of pro-inflammatory mediators. EK100 inhibited the inhibitor kappa B (IκB)/NF-κB signaling pathway. EK100 also inhibited phosphatidylinositol-3-kinase (PI3K)/Protein kinase B (Akt) signal transduction. Moreover, EK100 interfered with LPS docking to the LPS-binding protein (LBP), transferred to the cluster of differentiation 14 (CD14), and bonded to TLR4/myeloid differentiation-2 (MD-2) co-receptors. Compared with the TLR4 antagonist, resatorvid (CLI-095), and dexamethasone (Dexa), EK100 suppressed the TLR4/AKT signaling pathway. In addition, we also confirmed that EK100 attenuated the GFP-labeled NF-κB reporter gene expression in Drosophila. In summary, EK100 might alter LPS docking to LBP, CD14, and TLR4/MD-2 co-receptors, and then it suppresses the TLR4/NF-κB inflammatory pathway in LPS-stimulated RAW264.7 cells and Drosophila.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Drosophila/metabolism , Lipopolysaccharide Receptors/metabolism , Lymphocyte Antigen 96/metabolism , NF-kappa B/metabolism , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism , Animals , Anti-Inflammatory Agents/chemistry , Cell Survival/drug effects , Cyclooxygenase 2/metabolism , Cytokines/metabolism , Inflammation Mediators/metabolism , Lipopolysaccharide Receptors/chemistry , Lipopolysaccharides/chemistry , Lipopolysaccharides/immunology , Lymphocyte Antigen 96/chemistry , Macrophages/immunology , Macrophages/metabolism , Mice , Models, Molecular , Molecular Conformation , Nitric Oxide Synthase Type II/metabolism , Phosphorylation , Protein Binding , Structure-Activity Relationship , Toll-Like Receptor 4/chemistry
10.
Chem Commun (Camb) ; 57(50): 6209-6212, 2021 Jun 22.
Article in English | MEDLINE | ID: mdl-34059855

ABSTRACT

Toll-like receptor 4 (TLR4) recognizes various protein ligands; however, the protein-TLR4 binding model is unclear. Here we demonstrate a Crenomytilus grayanus lectin (CGL)-TLR4/MD2 model to show that CGL interacts with a TLR4/myeloid differentiation factor 2 (MD2) complex independently of sugar-binding properties. CGL could suppress lipopolysaccharide-induced immune responses significantly, suggesting that TLR4 itself has potential as a therapeutic target.


Subject(s)
Carbohydrates/chemistry , Lectins/chemistry , Lymphocyte Antigen 96/chemistry , Toll-Like Receptor 4/chemistry , Animals , Binding Sites , Bivalvia , Carbohydrates/immunology , Humans , Lectins/immunology , Lymphocyte Antigen 96/immunology , Toll-Like Receptor 4/immunology
11.
Front Immunol ; 12: 631797, 2021.
Article in English | MEDLINE | ID: mdl-33815382

ABSTRACT

Pro-inflammatory signaling mediated by Toll-like receptor 4 (TLR4)/myeloid differentiation-2 (MD-2) complex plays a crucial role in the instantaneous protection against infectious challenge and largely contributes to recovery from Gram-negative infection. Activation of TLR4 also boosts the adaptive immunity which is implemented in the development of vaccine adjuvants by application of minimally toxic TLR4 activating ligands. The modulation of pro-inflammatory responses via the TLR4 signaling pathway was found beneficial for management of acute and chronic inflammatory disorders including asthma, allergy, arthritis, Alzheimer disease pathology, sepsis, and cancer. The TLR4/MD-2 complex can recognize the terminal motif of Gram-negative bacterial lipopolysaccharide (LPS)-a glycophospholipid lipid A. Although immense progress in understanding the molecular basis of LPS-induced TLR4-mediated signaling has been achieved, gradual, and predictable TLR4 activation by structurally defined ligands has not yet been attained. We report on controllable modulation of cellular pro-inflammatory responses by application of novel synthetic glycolipids-disaccharide-based lipid A mimetics (DLAMs) having picomolar affinity for TLR4/MD-2. Using crystal structure inspired design we have developed endotoxin mimetics where the inherently flexible ß(1 → 6)-linked diglucosamine backbone of lipid A is replaced by a conformationally restricted α,α-(1↔1)-linked disaccharide scaffold. The tertiary structure of the disaccharide skeleton of DLAMs mirrors the 3-dimensional shape of TLR4/MD-2 bound E. coli lipid A. Due to exceptional conformational rigidity of the sugar scaffold, the specific 3D organization of DLAM must be preserved upon interaction with proteins. These structural factors along with specific acylation and phosphorylation pattern can ensure picomolar affinity for TLR4 and permit efficient dimerization of TLR4/MD-2/DLAM complexes. Since the binding pose of lipid A in the binding pocket of MD-2 (±180°) is crucial for the expression of biological activity, the chemical structure of DLAMs was designed to permit a predefined binding orientation in the binding groove of MD-2, which ensured tailored and species-independent (human and mice) TLR4 activation. Manipulating phosphorylation and acylation pattern at the sugar moiety facing the secondary dimerization interface allowed for adjustable modulation of the TLR4-mediated signaling. Tailored modulation of cellular pro-inflammatory responses by distinct modifications of the molecular structure of DLAMs was attained in primary human and mouse immune cells, lung epithelial cells and TLR4 transfected HEK293 cells.


Subject(s)
Biomimetic Materials/pharmacology , Disaccharides/pharmacology , Immunomodulation , Lipid A/pharmacology , Animals , Biomimetic Materials/chemical synthesis , Biomimetic Materials/chemistry , Cytokines/immunology , Disaccharides/chemistry , Escherichia coli , HEK293 Cells , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Lipid A/chemistry , Lipopolysaccharides/chemistry , Lipopolysaccharides/pharmacology , Lymphocyte Antigen 96/chemistry , Lymphocyte Antigen 96/immunology , Macrophages/drug effects , Macrophages/immunology , Mice , NF-kappa B/immunology , Signal Transduction/drug effects , THP-1 Cells , Toll-Like Receptor 4/chemistry , Toll-Like Receptor 4/immunology
12.
Mol Med Rep ; 23(3)2021 03.
Article in English | MEDLINE | ID: mdl-33495830

ABSTRACT

Trienones are curcuminoid analogues and are minor constituents in the rhizomes of numerous Curcuma plant species. Studies investigating the biological activities of trienones, particularly their anti­inflammatory activities, are limited. In the present study, the trienone 1,7­bis(4­hydroxy­3­methoxyphenyl)­1,4,6­heptatrien­3­one (HMPH) was structurally modified from curcumin using a novel and concise method. HMPH was shown to exhibit potential anti­inflammatory effects on lipopolysaccharide (LPS)­activated RAW264.7 macrophages. Furthermore, LPS­induced nitric oxide secretion in RAW264.7 cells was markedly and dose­dependently inhibited by HMPH; in addition, HMPH had a greater efficacy compared with curcumin. This inhibition was accompanied by the suppression of inducible nitric oxide synthase and cyclooxygenase­2 expression, as well as pro­inflammatory cytokine secretion. To elucidate the molecular mechanism underlying the anti­inflammatory effects of HMPH, the effects of this compound on nuclear factor­κB (NF­κB) translocation were assessed. HMPH significantly inhibited the translocation of p65 NF­κB into the nucleus to a greater extent than curcumin, thus indicating that HMPH has more potent anti­inflammatory activity than curcumin. In addition, an in silico modelling study revealed that HMPH possessed stronger binding energy to myeloid differentiation factor 2 (MD2) compared with that of curcumin, and indicated that the anti­inflammatory effects of HMPH may be through upstream inhibition of the inflammatory pathway. In conclusion, HMPH may be considered a promising compound for reducing inflammation via targeting p65 NF­κB translocation and interfering with MD2 binding.


Subject(s)
Curcumin/analogs & derivatives , Lipopolysaccharides/toxicity , Lymphocyte Antigen 96 , Macrophages/metabolism , Transcription Factor RelA , Animals , Curcumin/pharmacology , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Lymphocyte Antigen 96/chemistry , Lymphocyte Antigen 96/metabolism , Macrophages/pathology , Mice , RAW 264.7 Cells , Transcription Factor RelA/chemistry , Transcription Factor RelA/metabolism
13.
Hum Immunol ; 81(10-11): 596-605, 2020.
Article in English | MEDLINE | ID: mdl-32718721

ABSTRACT

Query fever is a zoonotic disease caused by Coxiella burnetii. There is no universal method for the prevention of this disease. Recombinant vaccine is a potent strategy that can be utilized for this purpose. The current study was conducted to develop a multi-epitope vaccine against Coxiella burnetii. Hence, OmpA, Tuf2, GroEL, Mip and sucB antigens were used for the prediction of epitopes. Then, a multi-epitope vaccine was developed based on a molecular adjuvant and fragments that contained the best MHCI, B cell, MHCII and IFN-γ epitopes. The features of the developed vaccine including physicochemical parameters, antigenicity and protein structures were assessed. Also, interaction between the developed vaccine and TLR4/MD2 receptor along with molecular dynamics of the ligand-receptor complex were investigated. Finally, the codon adaptation and cloning were conducted for the developed vaccine. According to the results, molecular weight, instability index, antigenicity and random coil percentage of the developed vaccine were 54.4 kDa, 32.84, 1.1936 and 34.92%, respectively. Besides, residues distribution in core region of the refined model was 85%. The results demonstrated that the developed vaccine could dock to its receptor with the lowest energy of -976.7 as well as RMSD value of the complex was between 0.15 and 0.22 nm. Also, the results showed that CIA index of the codon adapted sequence was 0.95. Finally, cloning results revealed that nucleotide sequence of the developed vaccine could be successfully cloned into pET-21a (+). Based on these results, it seems that the developed vaccine can be a suitable candidate to prevent Coxiella burnetii.


Subject(s)
Bacterial Vaccines/immunology , Bacterial Zoonoses/prevention & control , Coxiella burnetii/immunology , Epitopes, B-Lymphocyte/immunology , Molecular Docking Simulation/methods , Molecular Dynamics Simulation , Q Fever/prevention & control , Amino Acid Sequence , Animals , Antigens, Bacterial/immunology , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/immunology , Bacterial Vaccines/chemistry , Chaperonin 60/chemistry , Chaperonin 60/immunology , Epitopes, B-Lymphocyte/chemistry , Histocompatibility Antigens Class I/chemistry , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class II/chemistry , Histocompatibility Antigens Class II/immunology , Humans , Ketoglutarate Dehydrogenase Complex/chemistry , Ketoglutarate Dehydrogenase Complex/immunology , Lymphocyte Antigen 96/chemistry , Protein Structure, Secondary , Protein Structure, Tertiary , Q Fever/immunology , Q Fever/microbiology , Toll-Like Receptor 4/chemistry
14.
Biomolecules ; 10(4)2020 03 27.
Article in English | MEDLINE | ID: mdl-32230927

ABSTRACT

Inflammation is a well-organized innate immune response that plays an important role during the pathogen attacks and mechanical injuries. The Toll-like receptors (TLR)/nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) is a major signal transduction pathway observed in RAW 264.7 macrophages during the inflammatory responses. Here, we investigated the anti-inflammatory effects of Octominin; a bio-active peptide developed from Octopus minor in RAW 264.7 macrophages in vitro. Octominin was found to inhibit lipopolysaccharides (LPS)-stimulated transcriptional activation of NF-κB in RAW 264.7 cells and dose-dependently decreased the mRNA expression levels of TLR4. Specifically, in silico docking results demonstrated that Octominin has a potential to inhibit TLR4 mediated inflammatory responses via blocking formation of TLR4/MD-2/LPS complex. We also demonstrated that Octominin could significantly inhibit LPS-induced secretion of pro-inflammatory cytokine (interleukin-ß; IL-1ß, IL-6, and tumor necrosis factor-α) and chemokines (CCL3, CCL4, CCL5, and CXCL10) from RAW 264.7 cells. Additionally, Octominin repressed the LPS-induced pro-inflammatory mediators including nitric oxide (NO), prostaglandin E2, inducible NO synthase, and cyclooxygenase 2 in macrophages. These results suggest that Octominin is a potential inhibitor of TLRs/NF-κB signal transduction pathway and is a potential candidate for the treatment of inflammatory diseases.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Cytokines/metabolism , Octopodiformes/chemistry , Peptide Fragments/pharmacology , Peptides/pharmacology , Signal Transduction/drug effects , Animals , Chemokines/genetics , Chemokines/metabolism , Cytokines/genetics , Dinoprostone/metabolism , Inflammation Mediators/metabolism , Lipopolysaccharides/pharmacology , Lymphocyte Antigen 96/chemistry , Lymphocyte Antigen 96/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice , NF-kappa B/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Peptides/genetics , Phosphorylation/drug effects , RAW 264.7 Cells , Toll-Like Receptor 4/classification , Toll-Like Receptor 4/metabolism
15.
J Biol Chem ; 295(51): 17842-17851, 2020 12 18.
Article in English | MEDLINE | ID: mdl-33454018

ABSTRACT

Animals can sense the presence of microbes in their tissues and mobilize their own defenses by recognizing and responding to conserved microbial structures (often called microbe-associated molecular patterns (MAMPs)). Successful host defenses may kill the invaders, yet the host animal may fail to restore homeostasis if the stimulatory microbial structures are not silenced. Although mice have many mechanisms for limiting their responses to lipopolysaccharide (LPS), a major Gram-negative bacterial MAMP, a highly conserved host lipase is required to extinguish LPS sensing in tissues and restore homeostasis. We review recent progress in understanding how this enzyme, acyloxyacyl hydrolase (AOAH), transforms LPS from stimulus to inhibitor, reduces tissue injury and death from infection, prevents prolonged post-infection immunosuppression, and keeps stimulatory LPS from entering the bloodstream. We also discuss how AOAH may increase sensitivity to pulmonary allergens. Better appreciation of how host enzymes modify LPS and other MAMPs may help prevent tissue injury and hasten recovery from infection.


Subject(s)
Carboxylic Ester Hydrolases/metabolism , Gram-Negative Bacteria/metabolism , Lipopolysaccharides/metabolism , Animals , Carboxylic Ester Hydrolases/chemistry , Carboxylic Ester Hydrolases/genetics , Humans , Lung/immunology , Lung/metabolism , Lymphocyte Antigen 96/chemistry , Lymphocyte Antigen 96/metabolism , Neutrophils/metabolism , Toll-Like Receptor 4/chemistry , Toll-Like Receptor 4/metabolism
16.
Molecules ; 24(23)2019 Dec 03.
Article in English | MEDLINE | ID: mdl-31816891

ABSTRACT

Salvianolic acid B is one of the main water-soluble components of Salvia miltiorrhiza Bge. Many reports have shown that it has significant anti-myocardial ischemia effect. However, the underlying mechanism remains unclear. Our present study demonstrated that Sal B could alleviate myocardial ischemic injury by inhibiting the priming phase of NLRP3 inflammasome. In vivo, serum c-troponin I (cTn), lactate dehydrogenase (LDH) levels, the cardiac function and infract size were examined. We found that Sal B could notably reduce the myocardial ischemic injury caused by ligation of the left anterior descending coronary artery. In vitro, Sal B down-regulated the TLR4/NF-κB signaling cascades in lipopolysaccharide (LPS)-stimulated H9C2 cells. Furthermore, Sal B reduced the expression levels of IL-1ß and NLRP3 inflammasome in a dose-dependent manner. In short, our study provided evidence that Sal B could attenuate myocardial ischemic injury via inhibition of TLR4/NF-κB/NLRP3 signaling pathway. And in an upstream level, MD-2 may be the potential target.


Subject(s)
Benzofurans/administration & dosage , Lipopolysaccharides/adverse effects , Myocardial Ischemia/drug therapy , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Toll-Like Receptor 4/metabolism , Animals , Benzofurans/chemistry , Benzofurans/pharmacology , Cell Line , Disease Models, Animal , Dose-Response Relationship, Drug , Down-Regulation , Lymphocyte Antigen 96/antagonists & inhibitors , Lymphocyte Antigen 96/chemistry , Male , Models, Molecular , Myocardial Ischemia/etiology , Myocardial Ischemia/immunology , Rats , Signal Transduction
17.
FASEB J ; 33(12): 14528-14541, 2019 12.
Article in English | MEDLINE | ID: mdl-31675483

ABSTRACT

General anesthesia has been the requisite component of surgical procedures for over 150 yr. Although immunomodulatory effects of volatile anesthetics have been growingly appreciated, the molecular mechanism has not been understood. In septic mice, the commonly used volatile anesthetic isoflurane attenuated the production of 5-lipoxygenase products and IL-10 and reduced CD11b and intercellular adhesion molecule-1 expression on neutrophils, suggesting the attenuation of TLR4 signaling. We confirmed the attenuation of TLR4 signaling in vitro and their direct binding to TLR4-myeloid differentiation-2 (MD-2) complex by photolabeling experiments. The binding sites of volatile anesthetics isoflurane and sevoflurane were located near critical residues for TLR4-MD-2 complex formation and TLR4-MD-2-LPS dimerization. Additionally, TLR4 activation was not attenuated by intravenous anesthetics, except for a high concentration of propofol. Considering the important role of TLR4 system in the perioperative settings, these findings suggest the possibility that anesthetic choice may modulate the outcome in patients or surgical cases in which TLR4 activation is expected.-Okuno, T., Koutsogiannaki, S., Hou, L., Bu, W., Ohto, U., Eckenhoff, R. G., Yokomizo, T., Yuki, K. Volatile anesthetics isoflurane and sevoflurane directly target and attenuate Toll-like receptor 4 system.


Subject(s)
Anesthetics, Inhalation/pharmacology , Isoflurane/pharmacology , Sevoflurane/pharmacology , Toll-Like Receptor 4/metabolism , Animals , Binding Sites , Cells, Cultured , Lymphocyte Antigen 96/chemistry , Lymphocyte Antigen 96/metabolism , Male , Mice , Mice, Inbred C57BL , Molecular Docking Simulation , Protein Binding , Protein Multimerization , Toll-Like Receptor 4/chemistry
18.
Int J Mol Sci ; 20(13)2019 Jun 26.
Article in English | MEDLINE | ID: mdl-31247943

ABSTRACT

While precise mechanisms underlying cardiovascular diseases (CVDs) are still not fully understood, previous studies suggest that the innate immune system, through Toll-like receptor 4 (TLR4), plays a crucial part in the pathways leading to these diseases, mainly because of its interplay with endogenous molecules. The Heat-shock protein 70 family (HSP70-70kDa) is of particular interest in cardiovascular tissues as it may have dual effects when interacting with TLR4 pathways. Although the hypothesis of the HSP70 family members acting as TLR4 ligands is becoming widely accepted, to date no co-crystal structure of this complex is available and it is still unknown whether this process requires the co-adaptor MD2. In this study, we aimed at investigating the interplay between the TLR4/MD2 complex and HSP70 family members in the human cardiovascular system through transcriptomic data analysis and at proposing a putative interaction model between these proteins. We report compelling evidence of correlated expression levels between TLR4 and MD2 with HSP70 cognate family members, especially in heart tissue. In our molecular docking simulations, we found that HSP70 in the ATP-bound state presents a better docking score towards the TLR4/MD2 complex compared to the ADP-bound state (-22.60 vs. -10.29 kcal/mol, respectively). Additionally, we show via a proximity ligation assay for HSP70 and TLR4, that cells stimulated with ATP have higher formation of fluorescent spots and that MD2 might be required for the complexation of these proteins. The insights provided by our computational approach are potential scaffolds for future in vivo studies investigating the interplay between the TLR4/MD2 complex and HSP70 family members in the cardiovascular system.


Subject(s)
Cardiovascular System/metabolism , Computational Biology , HSP70 Heat-Shock Proteins/metabolism , Lymphocyte Antigen 96/metabolism , Models, Biological , Toll-Like Receptor 4/metabolism , Adult , Aged , Aged, 80 and over , Computational Biology/methods , Female , Gene Expression Profiling , HSP70 Heat-Shock Proteins/chemistry , HSP70 Heat-Shock Proteins/genetics , Humans , Lymphocyte Antigen 96/chemistry , Lymphocyte Antigen 96/genetics , Male , Middle Aged , Models, Molecular , Multiprotein Complexes/metabolism , Protein Binding , Structure-Activity Relationship , Toll-Like Receptor 4/chemistry , Toll-Like Receptor 4/genetics , Transcriptome , Young Adult
19.
Protein Sci ; 28(7): 1350-1358, 2019 07.
Article in English | MEDLINE | ID: mdl-31075178

ABSTRACT

Toll-like receptor 4 (TLR4) is a critical innate immune protein that activates inflammation in response to extracellular cues. Much of the work to understand how the protein works in humans has been done using mouse models. Although human and mouse TLR4 have many shared features, they have also diverged significantly since their last common ancestor, acquiring 277 sequence differences. Functional differences include the extent of ligand-independent activation, whether lipid IVa acts as an antagonist or agonist, and the relative species cross-compatibility of their MD-2 cofactor. We set out to understand the evolutionary origins for these functional differences between human and mouse TLR4. Using a combination of phylogenetics, ancestral sequence reconstruction, and functional characterization, we found that evolutionary changes to the human TLR4, rather than changes to the mouse TLR4, were largely responsible for these functional changes. Human TLR4 repressed ancestral ligand-independent activity and gained antagonism to lipid IVa. Additionally, mutations to the human TLR4 cofactor MD-2 led to lineage-specific incompatibility between human and opossum TLR4 complex members. These results were surprising, as mouse TLR4 has acquired many more mutations than human TLR4 since their last common ancestor. Our work has polarized this set of transitions and sets up work to study the mechanistic underpinnings for the evolution of new functions in TLR4.


Subject(s)
Toll-Like Receptor 4/metabolism , Humans , Lymphocyte Antigen 96/chemistry , Lymphocyte Antigen 96/metabolism , Models, Molecular , Mutation , Toll-Like Receptor 4/chemistry , Toll-Like Receptor 4/genetics
20.
J Med Chem ; 62(11): 5453-5469, 2019 06 13.
Article in English | MEDLINE | ID: mdl-30998353

ABSTRACT

Acute lung injury (ALI) is often caused by systemic inflammatory responses. Targeting the myeloid differentiation protein 2/toll-like receptor 4 (MD2-TLR4) complex may be a promising way to treat Gram-negative bacterial-induced inflammatory disorders. In this study, we report the design and synthesis of a new series of 3-(indol-5-yl)-indazoles, which were evaluated for their anti-inflammatory activities in macrophages. Among the analogues generated, the promising 3-(indol-5-yl)-indazole analogue 22m inhibited lipopolysaccharide (LPS)-induced expression of tumor necrosis factor alpha (TNF-α) and interleukin-6 (IL-6) in macrophages with IC50 values of 0.89 and 0.53 µM, respectively. Compound 22m was then identified as an MD2-TLR4 antagonist in suppressing LPS-induced inflammatory responses. In vivo administration of 22m significantly inhibited macrophage infiltration and ameliorated histopathological changes in lung tissues of LPS-challenged mice. Our studies have identified a new 3-(indol-5-yl)-indazole, 22m, as a potent MD2-TLR4 inhibitor and lay the groundwork for future drug development of anti-inflammatory agents for the treatment of ALI.


Subject(s)
Acute Lung Injury/drug therapy , Drug Design , Indazoles/chemistry , Indazoles/pharmacology , Lymphocyte Antigen 96/antagonists & inhibitors , Toll-Like Receptor 4/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/chemical synthesis , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Indazoles/chemical synthesis , Indazoles/therapeutic use , Lymphocyte Antigen 96/chemistry , Mice , Models, Molecular , Protein Conformation , Toll-Like Receptor 4/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL