Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 160
Filter
1.
Sci Rep ; 13(1): 22552, 2023 12 18.
Article in English | MEDLINE | ID: mdl-38110638

ABSTRACT

Certain intestinal microbiota alterations appear to positively correlate with tumorigenesis of CAC due to the disruption of the balance between the host and microorganisms. It is proven that blocking MyD88 signaling can prevent colitis-associated colorectal cancer (CAC) development in mice. We are aim to reveal the role of MyD88 signaling of maintaining colonic microbiota homeostasis for preventing CAC development. We here analyzed the landscape of gut microbiome in the mice model of AOM/DSS-induced CAC with MyD88 inhibitor treatment. PCoA revealed significant reduction in Lactobacillus load and increase in Escherichia load in the mucosal microbial composition of mice with CAC, compared with normal controls (NCs). Inhibitor-treatment led to almost undetectable Proteobacteria (Escherichia) and the retention of the dominance of Firmicutes and Bacteroidota (Muribaculaceae) in the mucosa. RNA sequencing analysis identified genes were up-regulated (Hp, SAA3 and IL-1F9) and down-regulated (CYP3A44, SLC30A10, GPNMB and OTC) in Inhibitor-treated mice (vs. CAC). Meanwhile, Inhibitor-treated mice had higher percentage of MUC2-positive area in colon sections (vs. CAC, which was less than NCs) by IF staining and decreased Escherichia in the mucus layer (vs. CAC) by FISH. And intestinal microbiota from mice with MyD88 inhibitor treatment could lessen the outcome of CAC by fecal microbiota transplantation. The development of CAC was involved in the increasing and ectopic Escherichia in the decreasing colonic mucus layer. MyD88 signaling blockade may maintain the host-microbiota homeostasis by up-regulating MUC2 production, increasing probiotics and their protective effects, and inhibiting the reproduction of Escherichia.


Subject(s)
Colitis-Associated Neoplasms , Colitis , Gastrointestinal Microbiome , Myeloid Differentiation Factor 88 , Animals , Mice , Colitis/complications , Colitis/microbiology , Colon/metabolism , Dextran Sulfate , Disease Models, Animal , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/antagonists & inhibitors , Myeloid Differentiation Factor 88/metabolism , Signal Transduction
2.
Int Immunopharmacol ; 124(Pt A): 110863, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37703787

ABSTRACT

BACKGROUND: Diabetic cardiomyopathy (DCM) is a common complication of diabetes mellitus and is associated with increased morbidity and mortality due to cardiac dysfunction. Chronic inflammation plays a significant role in the development of DCM, making it a promising target for novel pharmacological strategies. Our previous study has synthesized a novel compound, c17, which exhibited strong anti-inflammatory activity by specifically targeting to myeloid differentiation primary response 88 (MyD88). In this study, we evaluated the therapeutic effect of c17 in DCM. METHODS: The small molecular selective MyD88 inhibitor, c17, was used to evaluate the effect of MyD88 on DCM in both high concentration of glucose- and palmitic acid-stimulated macrophages and streptozotocin (STZ)-induced type 1 diabetes mellitus (T1DM) mice. RESULTS: The treatment of c17 in T1DM mice resulted in improved heart function and reduced cardiac hypertrophy, inflammation and fibrogenesis. RNA sequencing analysis of the heart tissues revealed that c17 effectively suppressed the inflammatory response by regulating the MyD88-dependent pathway. Co-immunoprecipitation experiments further confirmed that c17 disrupted the interaction between MyD88 and Toll-like receptor 4 (TLR4), consequently inhibiting downstream NF-κB activation. In vitro studies demonstrated that c17 exhibited similar anti-inflammatory activity by targeting MyD88 in macrophages, which are the primary regulators of cardiac inflammation. Furthermore, conditioned medium derived from c17-treated macrophages showed reduced capacity to induce hypertrophy, pro-fibrotic reactions, and secondary inflammation in cardiomyocytes. CONCLUSIONS: In conclusion, the small-molecule MyD88 inhibitor, c17, effectively combated the inflammatory DCM, therefore could be a potential candidate for the treatment of this disease.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Diabetic Cardiomyopathies , Myocarditis , Animals , Mice , Anti-Inflammatory Agents/adverse effects , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/drug therapy , Diabetic Cardiomyopathies/drug therapy , Diabetic Cardiomyopathies/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Myeloid Differentiation Factor 88/antagonists & inhibitors , Myeloid Differentiation Factor 88/drug effects , Myocarditis/drug therapy , NF-kappa B/metabolism , Streptozocin
3.
Mediators Inflamm ; 2022: 5184721, 2022.
Article in English | MEDLINE | ID: mdl-36523959

ABSTRACT

Background: Amentoflavone, a natural biflavone, exerts anti-inflammation, antioxidation, and antiapoptosis effects on many diseases. However, the mechanism of amentoflavone on neuroinflammation-related diseases has not been comprehensively examined clearly. Methods: BV2 microglial cells were treated with amentoflavone (10 µM), followed by lipopolysaccharide (LPS). Microglial activation and migration ability and the expression of proinflammatory cytokines and other signaling proteins were determined using immunohistochemistry, immunofluorescence, quantitative real-time polymerase chain reaction, Western blotting, enzyme-linked immunosorbent assay, and wound-healing assays. Results: Amentoflavone restored LPS-induced microglia activation, migration, and inflammation response which depends on regulating toll-like receptor 4 (TLR4)/myeloid differentiation factor 88 (MyD88)/nuclear factor kappa B (NF-κB) pathway. In addition, amentoflavone also enhanced nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) levels in LPS-treated BV2 microglial cells. Conclusions: Amentoflavone ameliorated LPS-induced neuroinflammatory response and oxidative stress in BV2 microglia. These data provide new insight into the mechanism of amentoflavone in the treatment of neuroinflammation-related diseases. Therefore, amentoflavone may be a potential therapeutic option for neurological disorders.


Subject(s)
Biflavonoids , Microglia , Humans , Cell Line , Heme Oxygenase-1/drug effects , Heme Oxygenase-1/metabolism , Lipopolysaccharides/pharmacology , Lipopolysaccharides/metabolism , Microglia/drug effects , Microglia/metabolism , Myeloid Differentiation Factor 88/antagonists & inhibitors , Myeloid Differentiation Factor 88/metabolism , Neuroinflammatory Diseases/drug therapy , NF-E2-Related Factor 2/agonists , NF-E2-Related Factor 2/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/metabolism , Biflavonoids/pharmacology , Biflavonoids/therapeutic use
4.
J Physiol Pharmacol ; 73(1)2022 Feb.
Article in English | MEDLINE | ID: mdl-35793767

ABSTRACT

Subarachnoid hemorrhage (SAH) is a life-threatening cerebrovascular disease with high rates of morbidity and mortality and a paucity of effective therapies. The development of early brain injury (EBI) is closely related to prognosis in SAH, and inflammation plays an important role in its pathophysiology. A previous experiment showed that ST2825, a selective inhibitor of MyD88, could alleviate EBI in vivo. However, this protective effect in vivo is affected by a variety of pathophysiology processes making the result to some extent uncertain. whether there is a coincident result in vitro ruling out the effect of other factors remains unknown, and further investigation using cultured neurons is necessary. Primary neuronal cells were cultured to construct an in vitro model of SAH. The cells were cultured and then divided into three groups: (1) a blank control group, (2) an oxygenated hemoglobin + vehicle group, and (3) an oxygenated hemoglobin + ST2825 group. In each group, apoptosis of neuronal cells along with changes in the expression of proteins including MyD88, p-JNK, p-Erk, p-p38, NFκB, Bcl-2, and P53 were measured. Results showed that after stimulating neurons with oxygenated hemoglobin, the expression of the MyD88 protein in the vehicle group increased significantly. The quantity of p-JNK, p-p38, and p-Erk also increased significantly, as did the quantity of p65 in the nucleus. Expression of the anti-apoptotic protein Bcl-2 was markedly reduced, while that of the cleaved caspase-3 protein was significantly increased. In addition, in this group, the apoptosis rate of neurons was significantly increased. In the ST2825 group, the expression of p-JNK, p-p38, p-Erk, cleaved caspase-3, and p65 in the nucleus was significantly decreased, the expression of Bcl-2 was significantly increased, and the apoptosis rate of neurons was significantly reduced. The results of this study suggest that in an experimental in vitro SAH model, ST2825, a selective inhibitor of MyD88, can have a neuroprotective effect by inhibiting neuronal apoptosis mediated by the MAPK and NFκB signaling pathways, and this has a certain protective effect on EBI after SAH.


Subject(s)
Myeloid Differentiation Factor 88 , Neurons , Subarachnoid Hemorrhage , Animals , Hemoglobins/metabolism , Mice , Myeloid Differentiation Factor 88/antagonists & inhibitors , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , Neurons/metabolism , Neurons/pathology , Proto-Oncogene Proteins c-bcl-2/metabolism , Subarachnoid Hemorrhage/drug therapy , Subarachnoid Hemorrhage/metabolism , Subarachnoid Hemorrhage/pathology
5.
Int J Mol Sci ; 23(3)2022 Jan 21.
Article in English | MEDLINE | ID: mdl-35163130

ABSTRACT

It is known that plant phenolic compounds exert anti-inflammatory activity through both anti-oxidant effects and modulation of pivotal pro-inflammatory factors. Recently, Olea europaea has been studied as a natural source of bioactive molecules; however, few studies have focused on the biological effect of oleacein (OLC), the most abundant secoiridoid. Therefore, the aim of this study was to investigate the potential anti-oxidant activity of OLC, as well as to study its anti-inflammatory effect in lipopolysaccharide (LPS)-stimulated THP-1-derived macrophages. LPS brought a dramatic increase of both release and gene expression of pro-inflammatory cytokines (IL-6, IL-1ß and TNF-α), as well as a decrease of anti-inflammatory ones (IL-10), the effects of which are reverted by OLC. Moreover, it reduced the levels of COX-2, NO and PGE2 elicited by LPS exposure in THP-1 macrophages. Interestingly, OLC modulated inflammatory signaling pathways through the inhibition of CD14/TLR4/CD14/MyD88 axis and the activation of NF-κB. Finally, OLC showed relevant anti-oxidant capability, assessed by abiotic assays, and reduced the intracellular amount of ROS generated by LPS exposure in THP-1 macrophages. Overall, these results suggest that the anti-oxidant activity and anti-inflammatory effect of OLC may cooperate in its protective effect against inflammatory stressors, thus being a possible alternative pharmacological strategy aimed at reducing the inflammatory process.


Subject(s)
Aldehydes/pharmacology , Inflammation/drug therapy , Lipopolysaccharides/adverse effects , Macrophages/drug effects , Myeloid Differentiation Factor 88/antagonists & inhibitors , NF-kappa B/antagonists & inhibitors , Phenols/pharmacology , Toll-Like Receptor 4/antagonists & inhibitors , Anti-Inflammatory Agents/pharmacology , Cytokines/metabolism , Humans , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Signal Transduction , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
6.
Invest New Drugs ; 40(3): 506-518, 2022 06.
Article in English | MEDLINE | ID: mdl-35089465

ABSTRACT

BACKGROUND: In cancer, myeloid-derived suppressor cells (MDSCs) are known to escape the host immune system by developing a highly suppressive environment. However, little is known about the molecular mechanism behind MDSC-mediated tumor cell evasion of the immune system. Toll-like receptor (TLR) signaling elicited in the tumor microenvironment has the potential to induce MDSC differentiations in different organs. Therefore, MDSC elimination by blocking the action of myeloid differentiation factor 88 (MyD88), which is a key adaptor-signaling molecule that affects TLR activity, seems to be an ideal tumor immunotherapy. Previous studies have proven that blocking MyD88 signaling with a novel MyD88 inhibitor (TJ-M2010-5, synthesized by Zhou's group) completely prevented colitis-associated colorectal cancer (CAC) development in mice. METHODS: In the present study, we investigated the impact of the novel MyD88 inhibitor on the number, phenotype, and function of MDSC in the mice model of CAC. RESULTS: We showed that CAC growth inhibition was involved in diminished MDSC generation, expansion, and suppressive function and that MDSC-mediated immune escape was dependent on MyD88 signaling pathway activation. MyD88 inhibitor treatment decreased the accumulation of CD11b+Gr1+ MDSCs in mice with CAC, thereby reducing cytokine (GM-CSF, G-CSF, IL-1ß, IL-6 and TGF-ß) secretion associated with MDSC accumulation, and reducing the expression of molecules (iNOS, Arg-1 and IDO) associated with the suppressive capacity of MDSCs. In addition, MyD88 inhibitor treatment reduced the differentiation of MDSCs from myeloid cells and the suppressive capacity of MDSCs on the proliferation of activated CD4+ T cells in vitro. CONCLUSION: MDSCs are primary cellular targets of a novel MyD88 inhibitor during CAC development. Our findings prove that MyD88 signaling is involved in the regulation of the immunosuppressive functions of MDSCs. The novel MyD88 inhibitor TJ-M2010-5 is a new and effective agent that modulates MyD88 signaling to overcome MDSC suppressive functions, enabling the development of successful antitumor immunotherapy.


Subject(s)
Colitis-Associated Neoplasms , Myeloid Differentiation Factor 88 , Myeloid-Derived Suppressor Cells , Piperazines , Thiazoles , Animals , Colitis-Associated Neoplasms/drug therapy , Colitis-Associated Neoplasms/metabolism , Colitis-Associated Neoplasms/pathology , Cytokines/metabolism , Mice , Myeloid Differentiation Factor 88/antagonists & inhibitors , Myeloid Differentiation Factor 88/metabolism , Myeloid-Derived Suppressor Cells/drug effects , Myeloid-Derived Suppressor Cells/metabolism , Myeloid-Derived Suppressor Cells/pathology , Piperazines/pharmacology , Signal Transduction , Thiazoles/pharmacology , Tumor Microenvironment
7.
Acta Pharmacol Sin ; 43(2): 354-366, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34552217

ABSTRACT

Emerging evidence shows that chronic inflammation mediated by toll-like receptors (TLRs) contributes to diabetic nephropathy. Myeloid differentiation primary-response protein-88 (MyD88) is an essential adapter protein of all TLRs except TLR3 in innate immunity. It is unclear whether MyD88 could be a therapeutic target for diabetic nephropathy. Here, we used a new small-molecule MyD88 inhibitor, LM8, to examine the pharmacological inhibition of MyD88 in protecting kidneys from inflammatory injury in diabetes. We showed that MyD88 was significantly activated in the kidney of STZ-induced type 1 diabetic mice in tubular epithelial cells as well as in high glucose-treated rat tubular epithelial cells NRK-52E. In cultured tubular epithelial cells, we show that LM8 (2.5-10 µM) or MyD88 siRNA attenuated high-concentration glucose-induced inflammatory and fibrogenic responses through inhibition of MyD88-TLR4 interaction and downstream NF-κB activation. Treatment with LM8 (5, 10 mg/kg, i.g.) significantly reduced renal inflammation and fibrosis and preserved renal function in both type 1 and type 2 diabetic mice. These renoprotective effects were associated with reduced MyD88-TLR4 complex formation, suppressed NF-κB signaling, and prevention of inflammatory factor expression. Collectively, our results show that hyperglycemia activates MyD88 signaling cascade to induce renal inflammation, fibrosis, and dysfunction. Pharmacological inhibition of MyD88 may be a therapeutic approach to mitigate diabetic nephropathy and the inhibitor LM8 could be a potential candidate for such therapy.


Subject(s)
Diabetic Nephropathies/prevention & control , Hypoglycemic Agents/therapeutic use , Kidney Tubules/drug effects , Myeloid Differentiation Factor 88/antagonists & inhibitors , Animals , Blotting, Western , Diabetes Mellitus, Experimental/complications , Diabetic Nephropathies/metabolism , Immunoprecipitation , Kidney/drug effects , Kidney/pathology , Kidney Tubules/pathology , Male , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction , Surface Plasmon Resonance
8.
Int J Mol Sci ; 22(19)2021 Sep 23.
Article in English | MEDLINE | ID: mdl-34638569

ABSTRACT

Renal ischemia reperfusion injury (RIRI) is one of the main causes of acute kidney injury (AKI), which can lead to acute renal failure. The development of RIRI is so complicated that it involves many factors such as inflammatory response, oxidative stress and cell apoptosis. Ganoderic acids (GAs), as one of the main pharmacological components of Ganoderma lucidum, have been reported to possess anti-inflammatory, antioxidant, and other pharmacological effects. The study is aimed to investigate the protective effect of GAs on RIRI and explore related underlying mechanisms. The mechanisms involved were assessed by a mouse RIRI model and a hypoxia/reoxygenation model. Compared with sham-operated group, renal dysfunction and morphological damages were relieved markedly in GAs-pretreatment group. GAs pretreatment could reduce the production of pro-inflammatory factors such as IL-6, COX-2 and iNOS induced by RIRI through inhibiting TLR4/MyD88/NF-kB signaling pathway. Furthermore, GAs reduced cell apoptosis via the decrease of the ratios of cleaved caspase-8 and cleaved caspase-3. The experimental results suggest that GAs prevent RIRI by alleviating tissue inflammation and apoptosis and might be developed as a candidate drug for preventing RIRI-induced AKI.


Subject(s)
Acute Kidney Injury/prevention & control , Apoptosis/drug effects , Inflammation/drug therapy , Protective Agents/pharmacology , Reperfusion Injury/prevention & control , Triterpenes/pharmacology , Acute Kidney Injury/etiology , Acute Kidney Injury/pathology , Animals , Cell Line , Cyclooxygenase 2/metabolism , Disease Models, Animal , Inflammation/metabolism , Interleukin-6/metabolism , Male , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/antagonists & inhibitors , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/metabolism , Oxidative Stress/drug effects , Protective Agents/therapeutic use , Rats , Reperfusion Injury/complications , Reperfusion Injury/pathology , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/metabolism , Triterpenes/therapeutic use
9.
Bioorg Med Chem ; 49: 116442, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34600241

ABSTRACT

Acquired paclitaxel (PTX) chemoresistance in triple-negative breast cancer (TNBC) can be inferred from the overexpression of toll-like receptor 4 (TLR4) and myeloid differentiation primary response 88 (MyD88) proteins and the activation of the TLR4/MyD88 cascading signalling pathway. Finding a new inhibitor that can attenuate the activation of this pathway is a novel strategy for reducing PTX chemoresistance. In this study, a series of small molecule compounds were synthesised and tested in combination with PTX against TNBC cells. The trimethoxy-substituted compound significantly decreased MyD88 overexpression and improved PTX activity in MDA-MB-231TLR4+ cells but not in HCCTLR4- cells. On the contrary, the trifluoromethyl-substituted compound with PTX synergistically improved the growth inhibition in both TNBC subtypes. The fluorescence titrations indicated that both compounds could bind with MD2 with good and comparable binding affinities. This was further supported by docking analysis, in which both compounds fit perfectly well and form some critical binding interactions with MD2, an essential lipid-binding accessory to TLR4 involved in activating the TLR-4/MyD88-dependent pathway.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Myeloid Differentiation Factor 88/antagonists & inhibitors , Paclitaxel/pharmacology , Small Molecule Libraries/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Antineoplastic Agents, Phytogenic/chemical synthesis , Antineoplastic Agents, Phytogenic/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Myeloid Differentiation Factor 88/genetics , Paclitaxel/chemical synthesis , Paclitaxel/chemistry , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/genetics , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology
10.
J Cell Mol Med ; 25(20): 9533-9542, 2021 10.
Article in English | MEDLINE | ID: mdl-34562309

ABSTRACT

Progressive photoreceptor death occurs in blinding diseases such as retinitis pigmentosa. Myeloid differentiation primary response protein 88 (MyD88) is a central adaptor protein for innate immune system Toll-like receptors (TLR) and induces cytokine secretion during retinal disease. We recently demonstrated that inhibiting MyD88 in mouse models of retinal degeneration led to increased photoreceptor survival, which was associated with altered cytokines and increased neuroprotective microglia. However, the identity of additional molecular changes associated with MyD88 inhibitor-induced neuroprotection is not known. In this study, we used isobaric tags for relative and absolute quantification (iTRAQ) labelling followed by LC-MS/MS for quantitative proteomic analysis on the rd10 mouse model of retinal degeneration to identify protein pathways changed by MyD88 inhibition. Quantitative proteomics using iTRAQ LC-MS/MS is a high-throughput method ideal for providing insight into molecular pathways during disease and experimental treatments. Forty-two proteins were differentially expressed in retinas from mice treated with MyD88 inhibitor compared with control. Notably, increased expression of multiple crystallins and chaperones that respond to cellular stress and have anti-apoptotic properties was identified in the MyD88-inhibited mice. These data suggest that inhibiting MyD88 enhances chaperone-mediated retinal protection pathways. Therefore, this study provides insight into molecular events contributing to photoreceptor protection from modulating inflammation.


Subject(s)
Myeloid Differentiation Factor 88/antagonists & inhibitors , Neuroprotection , Proteome , Proteomics , Retinal Degeneration/etiology , Retinal Degeneration/metabolism , Animals , Biomarkers , Chromatography, High Pressure Liquid , Computational Biology/methods , Disease Models, Animal , Disease Susceptibility , Female , Gene Expression Regulation , Gene Ontology , Male , Mass Spectrometry , Mice , Photoreceptor Cells/metabolism , Photoreceptor Cells/pathology , Protein Interaction Mapping/methods , Proteomics/methods , Retinal Degeneration/pathology , Toll-Like Receptors/metabolism
11.
Int J Mol Sci ; 22(10)2021 May 13.
Article in English | MEDLINE | ID: mdl-34068193

ABSTRACT

In this study, we investigate the immunomodulatory effects of a novel antimicrobial peptide, YD1, isolated from Kimchi, in both in vitro and in vivo models. We establish that YD1 exerts its anti-inflammatory effects via up-regulation of the Nrf2 pathway, resulting in the production of HO-1, which suppresses activation of the NF-κB pathway, including the subsequent proinflammatory cytokines IL-1ß, IL-6, and TNF-α. We also found that YD1 robustly suppresses nitric oxide (NO) and prostaglandin E2 (PGE2) production by down-regulating the expression of the upstream genes, iNOS and COX-2, acting as a strong antioxidant. Collectively, YD1 exhibits vigorous anti-inflammatory and antioxidant activity, presenting it as an interesting potential therapeutic agent.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Gene Expression Regulation/drug effects , Heme Oxygenase-1/metabolism , Inflammation/prevention & control , Membrane Proteins/metabolism , NF-E2-Related Factor 2/metabolism , Pore Forming Cytotoxic Proteins/pharmacology , Animals , Cytokines/metabolism , Edema/chemically induced , Edema/metabolism , Edema/pathology , Edema/prevention & control , Heme Oxygenase-1/genetics , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Lipopolysaccharides/toxicity , Membrane Proteins/genetics , Mice , Myeloid Differentiation Factor 88/antagonists & inhibitors , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , NF-E2-Related Factor 2/genetics , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , NF-kappa B/metabolism , Nitric Oxide/metabolism , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
12.
Ann Clin Lab Sci ; 51(3): 376-384, 2021 May.
Article in English | MEDLINE | ID: mdl-34162568

ABSTRACT

OBJECTIVE: To investigate the effect of dexmedetomidine (DEX) on myocardial injury induced by renal ischemia/reperfusion (I/R) and to explore the role of the HMGB1-TLR4-MyD88-NF-κB signaling pathway. METHODS: Adult male Wistar rats were randomly allocated into the control group, renal I/R group, renal I/R group pretreated with a low dose of DEX (L-Dex+I/R), renal I/R group pretreated with a medium dose of DEX (M-Dex+I/R), and renal I/R group pretreated with a high dose of DEX (H-Dex+I/R). Outcome measures included the plasma concentrations of HMGB1, IL-6, IL-10, IL-17, and TnI, the expression levels of HMGB1, TLR4, MyD88, NF-κBp65, and P-NF-κBp65, the pathological change, and the cell apoptosis. RESULTS: Renal I/R led to severe myocardial histological injury and cell apoptosis. DEX reduced the plasma concentration of IL-17 and TnI in a dose-dependent manner in the renal I/R model rats and inhibited the protein expression of TLR4 and NF-κBp65 in a dose-dependent manner in the myocardial tissue. Additionally, mRNA expression of MyD88 was elevated in the I/R group compared with the control group, and DEX significantly reduced mRNA expression of MyD88 in the renal I/R model rats. DEX inhibited myocardial cell apoptosis in the renal I/R model rats. CONCLUSION: DEX could attenuate myocardial injury induced by renal I/R in a dose-dependent manner. The potential mechanisms are associated with inhibition of the HMGB1-TLR4-NF-κB signaling pathway and myocardial cell apoptosis.


Subject(s)
Dexmedetomidine/pharmacology , HMGB1 Protein/antagonists & inhibitors , Ischemia/complications , Myeloid Differentiation Factor 88/antagonists & inhibitors , Myocardial Infarction/drug therapy , NF-kappa B/antagonists & inhibitors , Reperfusion Injury/complications , Toll-Like Receptor 4/antagonists & inhibitors , Adrenergic alpha-2 Receptor Agonists/pharmacology , Animals , Male , Myocardial Infarction/etiology , Myocardial Infarction/pathology , Rats , Rats, Wistar
13.
Inflammation ; 44(5): 1895-1907, 2021 Oct.
Article in English | MEDLINE | ID: mdl-33860870

ABSTRACT

Asthma is an inflammatory airway disease wherein bronchoconstriction, airway inflammation, and airway obstruction during asthma attacks are the main problems. It is recognized that imbalance of Th1/Th2 and Th17/Treg is a critical factor in asthma pathogenesis. Manipulation of these with signaling molecules such as mTOR, PI3K, Akt, and MyD88 can control asthma. Mouse model of allergic asthma was produced and treated with ketamine, metformin, metformin and ketamine, triciribine, LY294002, and torin2. MCh challenge test, BALf's Eos Count, the IL-4, 5, INF-γ, eicosanoid, total IgE levels were determined. The MUC5a, Foxp3, RORγt, PI3K, mTOR, Akt, PU.1, and MyD88 gene expressions and histopathology study were done. Asthma groups that were treated with all six components had reduced Penh value, total IgE, IL-4 and IL-5 levels, MUC5a, RORγt, MyD88 and mTOR expression, goblet cell hyperplasia, and mucus hyper-secretion. The eosinophil percentage and Cys-LT level were decreased by metformin and ketamine, triciribine, LY294002, and torin2. The level of IFN-γ was increased in triciribine, LY294002, and torin2. Metformin, metformin and ketamine, triciribine, LY294002, and torin2 reduced Akt and PI3K expression, peribronchial and perivascular inflammation, and increased expression of Foxp3. Torin2 had an effect on PU.1 expression. Inhibition of PI3K/AKT/mTOR and TLR4/MyD88/NF-κB signaling with targeted molecules can attenuate asthma pathology and play an important role in airways protection.


Subject(s)
Asthma/metabolism , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Toll-Like Receptor 4/metabolism , Animals , Anti-Asthmatic Agents/pharmacology , Anti-Asthmatic Agents/therapeutic use , Asthma/chemically induced , Asthma/drug therapy , Asthma/pathology , Chromones/pharmacology , Chromones/therapeutic use , Female , Metformin/pharmacology , Metformin/therapeutic use , Mice , Mice, Inbred BALB C , Morpholines/pharmacology , Morpholines/therapeutic use , Myeloid Differentiation Factor 88/antagonists & inhibitors , NF-kappa B/antagonists & inhibitors , Naphthyridines/pharmacology , Naphthyridines/therapeutic use , Ovalbumin/toxicity , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Phosphoinositide-3 Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/physiology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Toll-Like Receptor 4/antagonists & inhibitors
14.
Can J Physiol Pharmacol ; 99(10): 1007-1015, 2021 Oct.
Article in English | MEDLINE | ID: mdl-33852805

ABSTRACT

Sepsis often leads to systemic multiple organ dysfunction, with the majority of deaths attributable to acute myocardial injury (AMI). In this study, we aimed to explore the functional role of miR-365a-3p in sepsis-induced AMI. The sepsis myocardial injury model was constructed using lipopolysaccharide (LPS) both in vitro and in vivo with selective regulation of miR-365a-3p expression. Real-time PCR or Western blot was employed to detect the expressions of miR-365a-3p, inflammatory cytokines (tumor necrosis factor α (TNF-α), interleukin-1ß (IL-1ß), and IL-6), and inflammation-related proteins (nuclear factor-κB (NF-κB), I-κB, myeloid differentiation factor 88 (MyD88)) in myocardial tissues and cells. Also, cell counting kit-8 (CCK8) and flow cytometry assays were used to measure cardiomyocyte proliferation and apoptosis, respectively. Furthermore, the targeting relationship between miR-365a-3p and MyD88 was verified with the dual luciferase activity assay. miR-365a-3p was downregulated in LPS-induced myocardial injury model. miR-365a-3p overexpression attenuated cardiomyocyte apoptosis and suppressed the expressions of inflammatory cytokines and proteins. Inhibiting miR-365a-3p, however, produced the opposite effects. Mechanistically, miR-365a-3p targeted the 3'-untranslated region of MyD88, thereby inactivating MyD88-mediated NF-κB pathway. miR-365a-3p overexpression mitigated sepsis-mediated myocardial injury by inhibiting MyD88-mediated NF-κB activation.


Subject(s)
MicroRNAs/metabolism , Myeloid Differentiation Factor 88/antagonists & inhibitors , Myocardial Infarction/metabolism , NF-kappa B/antagonists & inhibitors , Sepsis/metabolism , Animals , Apoptosis/physiology , Lipopolysaccharides/pharmacology , MicroRNAs/genetics , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/pathology , NF-kappa B/genetics , NF-kappa B/metabolism , Random Allocation , Rats , Rats, Sprague-Dawley , Sepsis/genetics , Sepsis/pathology , Signal Transduction , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
15.
J Med Microbiol ; 70(4)2021 Apr.
Article in English | MEDLINE | ID: mdl-33830910

ABSTRACT

Introduction. Clostridioides difficile infection (CDI) causes toxin-mediated enteropathy, such as antibiotic-associated diarrhoea and pseudomembranous colitis. Rho-glucosylating toxin A (TcdA) and toxin B (TcdB) have been clearly implicated in pathogenesis, whereas the virulence of binary toxin (CDT) is still debated.Hypothesis statement. We hypothesized that CDT is involved in the host immune response and plays a pivotal role in establishing virulence by modulating pro-inflammatory cytokine production; this is achieved through the integral Toll-like receptor (TLR) signalling pathways.Aim. The aim of the present study was to determine whether and how CDT impacts macrophages compared to TcdA or TcdB by examining the induction of CXC chemokine ligand 2 (CXCL2) and tumour necrosis factor-α (TNF-α), both of which are crucial in mediating local and systematic inflammatory responses.Methodology. RAW264.7 cells or transfected human embryonic kidney (HEK) 293 T cells were incubated with TcdA, TcdB, or CDT. In some experiments, a neutralizing antibody against TLR2 or TLR4, or myeloid differentiation 88 inhibitory peptide were added. The amount of CXCL2 and TNF-α secreted was then measured.Results. In RAW264.7 macrophages, CXCL2 and TNF-α were produced via the Toll-like receptor 2 (TLR2) or Toll-like receptor 4 (TLR4) pathway in a TcdA, TcdB, or CDT dose-dependent manner. Interleukin-8 secretion was induced in TLR4/MD2/CD14-transfected, but not in TLR2-transfected, HEK 293 T cells following TcdB or CDT exposure.Conclusion. Our results showed that C. difficile toxins, including CDT, enhanced macrophage-mediated CXCL2 and TNF-α production via TLR2 and TLR4, indicating that CDT affects host immune responses.


Subject(s)
Bacterial Toxins/pharmacology , Chemokine CXCL2/metabolism , Clostridioides difficile/pathogenicity , Macrophages/drug effects , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , HEK293 Cells , Humans , Macrophages/metabolism , Mice , Myeloid Differentiation Factor 88/antagonists & inhibitors , Myeloid Differentiation Factor 88/metabolism , RAW 264.7 Cells , Signal Transduction/drug effects , Toll-Like Receptor 2/antagonists & inhibitors , Toll-Like Receptor 4/antagonists & inhibitors , Virulence
16.
Immunol Res ; 69(2): 117-128, 2021 04.
Article in English | MEDLINE | ID: mdl-33834387

ABSTRACT

The continuous emergence of infectious pathogens along with antimicrobial resistance creates a need for an alternative approach to treat infectious diseases. Targeting host factor(s) which are critically involved in immune signaling pathways for modulation of host immunity offers to treat a broad range of infectious diseases. Upon pathogen-associated ligands binding to the Toll-like/ IL-1R family, and other cellular receptors, followed by recruitment of intracellular signaling adaptor proteins, primarily MyD88, trigger the innate immune responses. But activation of host innate immunity strongly depends on the correct function of MyD88 which is tightly regulated. Dysregulation of MyD88 may cause an imbalance that culminates to a wide range of inflammation-associated syndromes and diseases. Furthermore, recent reports also describe that MyD88 upregulation with many viral infections is linked to decreased antiviral type I IFN response, and MyD88-deficient mice showed an increase in survivability. These reports suggest that MyD88 is also negatively involved via MyD88-independent pathways of immune signaling for antiviral type I IFN response. Because of its expanding role in controlling host immune signaling pathways, MyD88 has been recognized as a potential drug target in a broader drug discovery paradigm. Targeting BB-loop of MyD88, small molecule inhibitors were designed by structure-based approach which by blocking TIR-TIR domain homo-dimerization have shown promising therapeutic efficacy in attenuating MyD88-mediated inflammatory impact, and increased antiviral type I IFN response in experimental mouse model of diseases. In this review, we highlight the reports on MyD88-linked immune response and MyD88-targeted therapeutic approach with underlying mechanisms for controlling inflammation and antiviral type I IFN response. HIGHLIGHTS: • Host innate immunity is activated upon PAMPs binding to PRRs followed by immune signaling through TIR domain-containing adaptor proteins mainly MyD88. • Structure-based approach led to develop small-molecule inhibitors which block TIR domain homodimerization of MyD88 and showed therapeutic efficacy in limiting severe inflammation-associated impact in mice. • Therapeutic intervention of MyD88 also showed an increase in antiviral effect with strong type I IFN signaling linked to increased phosphorylation of IRFs via MyD88-independent pathway. • MyD88 inhibitors might be potentially useful as a small-molecule therapeutics for modulation of host immunity against inflammatory diseases and antiviral therapy. • However, prior clinical use of more in-depth efforts should be focused for suitability of the approach in deploying to complex diseases including COPD and COVID-19 in limiting inflammation-associated syndrome to infection.


Subject(s)
Drug Delivery Systems , Immunity, Innate/drug effects , Myeloid Differentiation Factor 88 , Virus Diseases , Animals , Disease Models, Animal , Humans , Mice , Myeloid Differentiation Factor 88/antagonists & inhibitors , Myeloid Differentiation Factor 88/immunology , Virus Diseases/drug therapy , Virus Diseases/immunology
17.
Mol Med Rep ; 23(4)2021 04.
Article in English | MEDLINE | ID: mdl-33655339

ABSTRACT

Toll­like receptor (TLR) 2/4 serves an important regulatory role in nerve tissue injury. However, the downstream and potential mechanisms remain to be elucidated. The present study was designed to investigate the roles of the TLR2/4­major myeloid differentiation response gene 88 (MyD88)­NF­κB signaling pathway in the development of intracranial aneurysm. The expression of TLR2, TLR4 and MyD88 in the blood of normal controls and patients with intracranial aneurysm were detected by quantitative PCR and ELISA. Human brain vascular smooth muscle cells were treated by Angiotensin II (Ang II) to evaluate the involvement of TLR2/4­MyD88­NF­κB signaling pathway in the process. The in vitro experiment was divided into four groups: The control group, an Ang â…¡ group, an Ang â…¡ + small interfering (si)RNA control group and an Ang â…¡ + TLR2­group. Cell viability, migration, apoptosis and expression of TLR2, TLR4, MyD88, NF­κB and phosphorylated (p­)p65 expression were detected. The results demonstrated that the expression of TLR2, TLR4, MyD88 and NF­κB at mRNA and protein levels in patients with intracranial aneurysm was significantly higher compared with corresponding protein in normal controls (P<0.05). In vitro experiments demonstrated that Ang â…¡ treatment increased the cell proliferation and migration rate but reduced the apoptotic rate compared with the control (P<0.05). The expression of TLR2, TLR4, MyD88, NF­κB and p­p65 was significantly increased in the Ang II group (vs. control; P<0.05). By contrast, TLR2­short interfering RNA reduced the cell proliferation and migration rate, and reduced the expression of TLR2, TLR4, MyD88, NF­κB and p­p65 (vs. Ang â…¡ + short interfering RNA control; P<0.05). In conclusion, the data of the present study indicated that the TLR2/4­MyD88­NF­κB signaling pathway is involved in the pathogenesis of intracranial aneurysm.


Subject(s)
Intracranial Aneurysm/genetics , Myeloid Differentiation Factor 88/genetics , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/genetics , Angiotensin II/pharmacology , Female , Gene Expression Regulation/genetics , Humans , Intracranial Aneurysm/pathology , Intracranial Aneurysm/therapy , Male , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Myeloid Differentiation Factor 88/antagonists & inhibitors , NF-kappa B/genetics , RNA, Messenger/genetics , RNA, Small Interfering/pharmacology , Signal Transduction/drug effects , Toll-Like Receptor 2/antagonists & inhibitors , Toll-Like Receptor 4/antagonists & inhibitors , Transcription Factor RelA/antagonists & inhibitors , Transcription Factor RelA/genetics
18.
Virulence ; 12(1): 704-722, 2021 12.
Article in English | MEDLINE | ID: mdl-33517839

ABSTRACT

Toll-like receptors (TLRs) are essential for the protection of the host from pathogen infections by initiating the integration of contextual cues to regulate inflammation and immunity. However, without tightly controlled immune responses, the host will be subjected to detrimental outcomes. Therefore, it is important to balance the positive and negative regulations of TLRs to eliminate pathogen infection, yet avert harmful immunological consequences. This study revealed a distinct mechanism underlying the regulation of the TLR network. The expression of sex-determining region Y-box 4 (Sox4) is induced by virus infection in viral infected patients and cultured cells, which subsequently represses the TLR signaling network to facilitate viral replication at multiple levels by a distinct mechanism. Briefly, Sox4 inhibits the production of myeloid differentiation primary response gene 88 (MyD88) and most of the TLRs by binding to their promoters to attenuate gene transcription. In addition, Sox4 blocks the activities of the TLR/MyD88/IRAK4/TAK1 and TLR/TRIF/TRAF3/TBK1 pathways by repressing their key components. Moreover, Sox4 represses the activation of the nuclear factor kappa-B (NF-κB) through interacting with IKKα/α, and attenuates NF-kB and IFN regulatory factors 3/7 (IRF3/7) abundances by promoting protein degradation. All these contributed to the down-regulation of interferons (IFNs) and IFN-stimulated gene (ISG) expression, leading to facilitate the viral replications. Therefore, we reveal a distinct mechanism by which viral pathogens evade host innate immunity and discover a key regulator in host defense.


Subject(s)
Immunity, Innate/genetics , SOXC Transcription Factors/genetics , SOXC Transcription Factors/immunology , Signal Transduction/immunology , Toll-Like Receptors/metabolism , Viruses/immunology , Enterovirus A, Human/immunology , Enterovirus A, Human/pathogenicity , Hep G2 Cells , Humans , Immunity, Innate/immunology , Influenza A virus/immunology , Influenza A virus/pathogenicity , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/virology , Myeloid Differentiation Factor 88/antagonists & inhibitors , Myeloid Differentiation Factor 88/immunology , Signal Transduction/genetics , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology , Virus Replication , Viruses/pathogenicity
19.
Immunopharmacol Immunotoxicol ; 43(2): 153-159, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33435756

ABSTRACT

OBJECTIVE: This current study evaluated the underlying mechanisms of LF against the inflammatory microRNAs (miRNAs), HMGB1 expression, and TLR4-MyD88-NF-кB pathway in LPS-activated murine RAW264.7 cells. METHODS: MTT assay was used to assess cell metabolism and the cell culture levels of the cytokines (TNF-α, IL-6) were evaluated by Enzyme-linked immunosorbent assay (ELISA). The expression of miRNAs was quantified by using qPCR and the expression of HMGB1, TLR4, MyD88, and phosphorylated NF-κB (P-p65) were determined with Western blot and qPCR, respectively. RESULTS: The results indicated that LF downregulates IL-6 and TNF-α expression. LF exhibited the degradation of P-p65 and reduced the production of HMGB1, TLR4, and MyD88 in LPS-induced inflammatory response. Importantly, in parallel with the suppression of cytokines and HMGB1-TLR4-MyD88-NF-кB pathway, LF could induce a decrease in inflammatory selected miRNAs, mmu-mir-155, and mmu-mir-146a expression. CONCLUSIONS: Altogether, these findings provide LF as a prominent anti-inflammatory agent that could modulate HMGB1, mmu-mir-155, mmu-mir-146a, and TLR4/MyD88/NF-кB pathway.


Subject(s)
HMGB1 Protein/antagonists & inhibitors , Lactoferrin/pharmacology , MicroRNAs/antagonists & inhibitors , Myeloid Differentiation Factor 88/antagonists & inhibitors , NF-kappa B/antagonists & inhibitors , Toll-Like Receptor 4/antagonists & inhibitors , Animals , Dose-Response Relationship, Drug , Gene Expression , HMGB1 Protein/biosynthesis , Lipopolysaccharides/toxicity , Mice , MicroRNAs/biosynthesis , Myeloid Differentiation Factor 88/biosynthesis , NF-kappa B/biosynthesis , RAW 264.7 Cells , Signal Transduction/drug effects , Signal Transduction/physiology , Toll-Like Receptor 4/biosynthesis
20.
Immunopharmacol Immunotoxicol ; 43(1): 51-57, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33251898

ABSTRACT

OBJECTIVE: Pretreatment of J774.1 cells with etidronate, a non-nitrogen-containing bisphosphonate (non-NBP) used as an antibone resorptive drug, was previously reported to inhibit Toll-like receptor (TLR) 2 agonist-induced proinflammatory cytokine production. The present study aimed to examine the effects of etidronate on chemokine production by human monocytic U937 cells incubated with Pam3Cys-Ser-(Lys)4 (Pam3CSK4, a TLR2 ligand) and lipid A (a TLR4 ligand). METHODS: U937 cells were pretreated with or without etidronate, and then incubated with or without Pam3CSK4 or lipid A. Levels of secreted human interleukin (IL)-8 and monocyte chemoattractant protein-1 (MCP-1) in culture supernatants and activation of nuclear factor-κB (NF-κB) p65 were measured by enzyme-linked immunosorbent assay (ELISA). Cytotoxicity was determined by measuring lactate dehydrogenase (LDH) activity in supernatants. Expression of intracellular adhesion molecule (ICAM)-1 and MyD88 was analyzed by flow cytometry and Western blot analysis, respectively. RESULTS: Etidronate down-regulated IL-8 and MCP-1 production and NF-κB p65 activation induced by Pam3CSK4, but not lipid A, in U937 cells. Etidronate also inhibited MyD88 expression in U937 cells incubated with Pam3CSK4. CONCLUSION: Etidronate down-regulates IL-8 and MCP-1 production in U937 cells by inhibiting both the expression of MyD88 and activation of NF-κB p65 in the TLR2, but not TLR4, pathway.


Subject(s)
Bone Density Conservation Agents/pharmacology , Chemokines/antagonists & inhibitors , Etidronic Acid/pharmacology , Myeloid Differentiation Factor 88/antagonists & inhibitors , NF-kappa B/antagonists & inhibitors , Toll-Like Receptor 2/antagonists & inhibitors , Chemokines/metabolism , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Down-Regulation/physiology , Gene Expression , Humans , Ligands , Myeloid Differentiation Factor 88/biosynthesis , NF-kappa B/metabolism , Toll-Like Receptor 2/metabolism , U937 Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...