Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.535
Filter
1.
Biomolecules ; 14(8)2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39199394

ABSTRACT

Cystic echinococcosis (CE) is a zoonotic disease caused by the parasite Echinococcus granulosus (E. granulosus), which can lead to the formation of liver lesions. Research indicates that E. granulosus releases both Toll-like receptor 2 (TLR2) and Interleukin-9 (IL-9), which can potentially impair the body's innate immune defenses and compromise the liver's ability to fight against diseases. To investigate the role of TLR2 and IL-9 in liver damage caused by E. granulosus infection, samples were initially collected from individuals diagnosed with CE. Subsequently, BALB/c mice were infected with E. granulosus at multiple time points (4 weeks, 12 weeks, 32 weeks) and the expression levels of these markers was then assessed at each of these phases. Furthermore, a BALB/c mouse model was generated and administered anti-IL-9 antibody via intraperitoneal injection. The subsequent analysis focused on the TLR2/MyD88/NF-κB signaling pathway and the expression of IL-9 in E. granulosus was examined. A co-culture experiment was conducted using mouse mononuclear macrophage cells (RAW264.7) and hepatic stellate cells (HSCs) in the presence of E. granulosus Protein (EgP). The findings indicated elevated levels of IL-9 and TLR2 in patients with CE, with the activation of the signaling pathway significantly increased as the duration of infection progressed. Administration of anti-IL-9 in mice reduced the activation of the TLR2/MyD88/NF-κB signaling pathway, exacerbating liver injury. Moreover, EgP stimulates the TLR2/MyD88/NF-κB signaling pathway, resulting in the synthesis of α-SMA and Collagen I. The data suggest that infection with E. granulosus may stimulate the production of IL-9 through the activation of the TLR2/MyD88/NF-κB signaling pathway, which is mediated by TLR2. This activation stimulates RAW264.7 and HSCs, exacerbating liver injury and fibrosis.


Subject(s)
Echinococcosis , Echinococcus granulosus , Interleukin-9 , Mice, Inbred BALB C , Myeloid Differentiation Factor 88 , Toll-Like Receptor 2 , Animals , Mice , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 2/genetics , Humans , Echinococcosis/pathology , Echinococcosis/immunology , Echinococcosis/metabolism , Interleukin-9/metabolism , Myeloid Differentiation Factor 88/metabolism , Myeloid Differentiation Factor 88/genetics , RAW 264.7 Cells , Liver/parasitology , Liver/metabolism , Liver/pathology , Female , Signal Transduction , NF-kappa B/metabolism , Male , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/parasitology , Adult , Disease Models, Animal
2.
Arch Biochem Biophys ; 759: 110112, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39111613

ABSTRACT

Inflammation is the body's response to injuries, which depends on numerous regulatory factors. Among them, miRNAs have gained much attention for their role in regulating inflammatory gene expression at multiple levels. In particular, miR-21 is up-regulated during the inflammatory response and reported to be involved in the resolution of inflammation by down-regulating pro-inflammatory mediators, including MyD88. Herein, we evaluated the regulatory effects of miR-21 on the TLR-4/MyD88 pathway in an in vitro model of 6-mer HA oligosaccharides-induced inflammation in human chondrocytes. The exposition of chondrocytes to 6-mer HA induced the activation of the TLR4/MyD88 pathway, which culminates in NF-kB activation. Changes in miR-21, TLR-4, MyD88, NLRP3 inflammasome, IL-29, Caspase1, MMP-9, iNOS, and COX-2 mRNA expression of 6-mer HA-stimulated chondrocytes were examined by qRT-PCR. Protein amounts of TLR-4, MyD88, NLRP3 inflammasome, p-ERK1/2, p-AKT, IL-29, caspase1, MMP-9, p-NK-kB p65 subunit, and IKB-a have been evaluated by ELISA kits. NO and PGE2 levels have been assayed by colorimetric and ELISA kits, respectively. HA oligosaccharides induced a significant increase in the expression of the above parameters, including NF-kB activity. The use of a miR-21 mimic attenuated MyD88 expression levels and the downstream effectors. On the contrary, treatment with a miR-21 inhibitor induced opposite effects. Interestingly, the use of a MyD88 siRNA confirmed MyD88 as the target of miR-21 action. Our results suggest that miR-21 expression could increase in an attempt to reduce the inflammatory response, targeting MyD88.


Subject(s)
Chondrocytes , Hyaluronic Acid , Inflammation , MicroRNAs , Myeloid Differentiation Factor 88 , Oligosaccharides , Humans , Myeloid Differentiation Factor 88/metabolism , Myeloid Differentiation Factor 88/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Chondrocytes/metabolism , Chondrocytes/drug effects , Hyaluronic Acid/pharmacology , Hyaluronic Acid/metabolism , Inflammation/metabolism , Inflammation/genetics , Oligosaccharides/pharmacology , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/genetics , Signal Transduction/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NF-kappa B/metabolism , Cells, Cultured
3.
Int Immunopharmacol ; 140: 112843, 2024 Oct 25.
Article in English | MEDLINE | ID: mdl-39098224

ABSTRACT

Neutrophils and macrophages confine pathogens by entrapping them in extracellular traps (ETs) through activating TLR9 function. However, plasmodial parasites secreted TatD-like DNases (TatD) to counteract ETs-mediated immune clearance. We found that TLR9 mutant mice increased susceptibility to rodent malaria, suggesting TLR9 is a key protein for host defense. We found that the proportion of neutrophils and macrophages in response to plasmodial parasite infection in the TLR9 mutant mice was significantly reduced compared to that of the WT mice. Importantly, PbTatD can directly bind to the surface TLR9 (sTLR9) on macrophages, which blocking the phosphorylation of mitogen-activated protein kinase and nuclear factor-κB, negatively regulated the signaling of ETs formation by both macrophages and neutrophils. Such, P. berghei TatD is a parasite virulence factor that can inhibit the proliferation of macrophages and neutrophils through directly binding to TLR9 receptors on the cell surface, thereby blocking the activation of the downstream MyD88-NF-kB pathways.


Subject(s)
Deoxyribonucleases , Immunity, Innate , Macrophages , Malaria , Mice, Inbred C57BL , NF-kappa B , Neutrophils , Plasmodium berghei , Signal Transduction , Toll-Like Receptor 9 , Toll-Like Receptor 9/metabolism , Animals , NF-kappa B/metabolism , Plasmodium berghei/immunology , Neutrophils/immunology , Macrophages/immunology , Macrophages/metabolism , Mice , Malaria/immunology , Malaria/parasitology , Deoxyribonucleases/metabolism , Extracellular Traps/immunology , Extracellular Traps/metabolism , Mice, Knockout , Protozoan Proteins/metabolism , Protozoan Proteins/immunology , Protozoan Proteins/genetics , Myeloid Differentiation Factor 88/metabolism , Myeloid Differentiation Factor 88/genetics , Humans
4.
J Cell Mol Med ; 28(15): e18583, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39123292

ABSTRACT

In this study, we investigated whether the ability of aucubin to mitigate the pathology of GONFH involves suppression of TLR4/NF-κB signalling and promotion of macrophage polarization to an M2 phenotype. In necrotic bone tissues from GONFH patients, we compared levels of pro-inflammatory M1 macrophages and anti-inflammatory M2 macrophages as well as levels of TLR4/NF-κB signalling. In a rat model of GONFH, we examined the effects of aucubin on these parameters. We further explored its mechanism of action in a cell culture model of M1 macrophages. Necrotic bone tissues from GONFH patients contained a significantly increased macrophage M1/M2 ratio, and higher levels of TLR4, MYD88 and NF-κB p65 than bone tissues from patients with hip osteoarthritis. Treating GONFH rats with aucubin mitigated bone necrosis and demineralization as well as destruction of trabecular bone and marrow in a dose-dependent manner, based on micro-computed tomography. These therapeutic effects were associated with a decrease in the overall number of macrophages, decrease in the proportion of M1 macrophages, increase in the proportion of M2 macrophages, and downregulation of TLR4, MYD88 and NF-κB p65. These effects in vivo were confirmed by treating cultures of M1 macrophage-like cells with aucubin. Aucubin mitigates bone pathology in GONFH by suppressing TLR4/NF-κB signalling to shift macrophages from a pro- to anti-inflammatory phenotype.


Subject(s)
Iridoid Glucosides , Macrophages , Myeloid Differentiation Factor 88 , Signal Transduction , Toll-Like Receptor 4 , Animals , Female , Humans , Male , Middle Aged , Rats , Disease Models, Animal , Femur Head Necrosis/chemically induced , Femur Head Necrosis/pathology , Femur Head Necrosis/metabolism , Femur Head Necrosis/drug therapy , Glucocorticoids/pharmacology , Iridoid Glucosides/pharmacology , Macrophages/metabolism , Macrophages/drug effects , Myeloid Differentiation Factor 88/metabolism , Myeloid Differentiation Factor 88/genetics , NF-kappa B/metabolism , Phenotype , Rats, Sprague-Dawley , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism
5.
Nat Commun ; 15(1): 7037, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39147750

ABSTRACT

The quest for targeted therapies is critical in the battle against cancer. The RAS/MAP kinase pathway is frequently implicated in neoplasia, with ERK playing a crucial role as the most distal kinase in the RAS signaling cascade. Our previous research demonstrated that the interaction between ERK and MYD88, an adaptor protein in innate immunity, is crucial for RAS-dependent transformation and cancer cell survival. In this study, we examine the biological consequences of disrupting the ERK-MYD88 interaction through the ERK D-recruitment site (DRS), while preserving ERK's kinase activity. Our results indicate that EI-52, a small-molecule benzimidazole targeting ERK-MYD88 interaction induces an HRI-mediated integrated stress response (ISR), resulting in immunogenic apoptosis specific to cancer cells. Additionally, EI-52 exhibits anti-tumor efficacy in patient-derived tumors and induces an anti-tumor T cell response in mice in vivo. These findings suggest that inhibiting the ERK-MYD88 interaction may be a promising therapeutic approach in cancer treatment.


Subject(s)
Benzimidazoles , Extracellular Signal-Regulated MAP Kinases , Myeloid Differentiation Factor 88 , Myeloid Differentiation Factor 88/metabolism , Myeloid Differentiation Factor 88/genetics , Humans , Animals , Mice , Extracellular Signal-Regulated MAP Kinases/metabolism , Cell Line, Tumor , Benzimidazoles/pharmacology , Apoptosis/drug effects , Immunogenic Cell Death/drug effects , Neoplasms/immunology , Neoplasms/drug therapy , Neoplasms/pathology , Neoplasms/metabolism , Female , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/immunology , Xenograft Model Antitumor Assays , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use
6.
Food Funct ; 15(17): 8916-8934, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39143863

ABSTRACT

Rosacea, a chronic inflammatory dermatological condition, is characterized by facial erythema and pustules. Recent investigations have delved into the interplay between the gut microbiota and rosacea pathogenesis, unveiling promising avenues for therapeutic intervention. In this study, we screened and isolated strains Ligilactobacillus salivarius 23-006 and Lacticaseibacillus paracasei 23-008 from the feces of healthy volunteers and evaluated the intervention effects of probiotics on rosacea by constructing an LL37 induced rosacea-like mouse model. Our results showed that both L. salivarius 23-006 and L. paracasei 23-008 were probiotic strains with favourable properties. In specific, we observed that both L. salivarius 23-006 and L. paracasei 23-008 alleviated skin lesions, reduced skin inflammatory infiltrates, and decreased the expression of inflammatory factors in mice, with the combination of L. salivarius 23-006 and L. paracasei 23-008 having the most significant effect. Moreover, the combination of strains reduced the expression of cathelicidin LL37 and rosacea-associated factors by inhibiting the TLR2/MyD88/NF-κB pathway. The 16S rRNA analysis showed that the combination enhanced the intestinal barrier, restored intestinal microbiota homeostasis, and up-regulated the abundance of Lactobacillus while down-regulating the abundance of Coprococcus and Oscillospira. We also explored the effects of postbiotics of L. salivarius 23-006 and L. paracasei 23-008 on rosacea. While postbiotics could also ameliorate the rosacea-like phenotype in mice via the TLR2/MyD88/NF-κB pathway, the effects were not as pronounced as those observed with probiotic treatment. However, the postbiotics still enhanced the intestinal barrier, up-regulated the Lactobacillus abundance, and modulated the intestinal microbiota. In conclusion, our study revealed that L. salivarius 23-006 and L. paracasei 23-008 improved rosacea by regulating the TLR2/MyD88/NF-κB pathway and intestinal microbiota, providing a theoretical basis for the treatment of rosacea.


Subject(s)
Cathelicidins , Myeloid Differentiation Factor 88 , NF-kappa B , Probiotics , Rosacea , Signal Transduction , Toll-Like Receptor 2 , Probiotics/pharmacology , Probiotics/administration & dosage , Rosacea/microbiology , Animals , Myeloid Differentiation Factor 88/metabolism , Toll-Like Receptor 2/metabolism , Mice , NF-kappa B/metabolism , Humans , Ligilactobacillus salivarius/physiology , Skin/microbiology , Antimicrobial Cationic Peptides , Female , Lacticaseibacillus paracasei/physiology , Male , Disease Models, Animal , Gastrointestinal Microbiome/drug effects , Mice, Inbred C57BL , Inflammation
7.
Int J Mol Sci ; 25(15)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39125989

ABSTRACT

Nearly six million people worldwide have died from the coronavirus disease (COVID-19) outbreak caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Although COVID-19 vaccines are largely successful in reducing the severity of the disease and deaths, the decline in vaccine-induced immunity over time and the continuing emergence of new viral variants or mutations underscore the need for an alternative strategy for developing broad-spectrum host-mediated therapeutics against SARS-CoV-2. A key feature of severe COVID-19 is dysregulated innate immune signaling, culminating in a high expression of numerous pro-inflammatory cytokines and chemokines and a lack of antiviral interferons (IFNs), particularly type I (alpha and beta) and type III (lambda). As a natural host defense, the myeloid differentiation primary response protein, MyD88, plays pivotal roles in innate and acquired immune responses via the signal transduction pathways of Toll-like receptors (TLRs), a type of pathogen recognition receptors (PRRs). However, recent studies have highlighted that infection with viruses upregulates MyD88 expression and impairs the host antiviral response by negatively regulating type I IFN. Galectin-3 (Gal3), another key player in viral infections, has been shown to modulate the host immune response by regulating viral entry and activating TLRs, the NLRP3 inflammasome, and NF-κB, resulting in the release of pro-inflammatory cytokines and contributing to the overall inflammatory response, the so-called "cytokine storm". These studies suggest that the specific inhibition of MyD88 and Gal3 could be a promising therapy for COVID-19. This review presents future directions for MyD88- and Gal3-targeted antiviral drug discovery, highlighting the potential to restore host immunity in SARS-CoV-2 infections.


Subject(s)
Antiviral Agents , COVID-19 , Galectin 3 , Myeloid Differentiation Factor 88 , SARS-CoV-2 , Humans , Myeloid Differentiation Factor 88/metabolism , Myeloid Differentiation Factor 88/genetics , SARS-CoV-2/physiology , SARS-CoV-2/immunology , COVID-19/immunology , COVID-19/virology , Antiviral Agents/therapeutic use , Antiviral Agents/pharmacology , Galectin 3/metabolism , COVID-19 Drug Treatment , Immunity, Innate , Signal Transduction , Animals
8.
Food Funct ; 15(17): 8797-8809, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39114922

ABSTRACT

Probiotics can alleviate alcoholic liver disease. However, whether inactive counterparts can produce similar outcomes requires further investigation. We investigated the effects of viable (V) and dead (D) Lactobacillus paracasei CCFM1120 on alcohol-induced ALD mice. The results showed that CCFM1120V and D ameliorated the disease symptoms and intestinal injury. Specifically, these interventions strengthened the intestinal barrier, as evidenced by the increased expression of ZO-1 (zonula occludens 1), occludin, and claudin-1 in the colon and the restored ileal microstructure, including the villi and crypts. In addition, they enhanced the antioxidant capacity of the liver by reducing the production of malondialdehyde and increasing the levels of glutathione and superoxide dismutase. The activation of Nrf2 and HO-1 may be responsible for recovering the antioxidant capacity. Interventions can decrease mouse TNF-α, IL-6 and IL-1ß content in serum, probably through the TLR4/MyD88/NF-κB pathway. Furthermore, they possess the ability to restore the quantities of bacteria responsible for producing butyric acid, such as Lactobacillus, Blautia, Bifidobacterium, Ruminococcaceae, Faecalibaculum and Lachnospiraceae. Taken together, CCFM1120V and D apparently can modify the composition of the gut microbiota, foster the gastrointestinal equilibrium, fortify the intestinal barrier, augment the antioxidant capacity of the liver, and effectively shield it from ethanol-induced injury.


Subject(s)
Gastrointestinal Microbiome , Lacticaseibacillus paracasei , Liver Diseases, Alcoholic , Myeloid Differentiation Factor 88 , NF-E2-Related Factor 2 , NF-kappa B , Probiotics , Toll-Like Receptor 4 , Animals , Liver Diseases, Alcoholic/metabolism , Mice , NF-E2-Related Factor 2/metabolism , Toll-Like Receptor 4/metabolism , NF-kappa B/metabolism , Myeloid Differentiation Factor 88/metabolism , Probiotics/pharmacology , Male , Lacticaseibacillus paracasei/metabolism , Signal Transduction , Mice, Inbred C57BL , Liver/metabolism , Antioxidants/metabolism , Membrane Proteins , Heme Oxygenase-1
9.
Discov Med ; 36(187): 1627-1640, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39190378

ABSTRACT

BACKGROUND: Tumor necrosis factor alpha induced protein 3 (TNFAIP3) is reportedly to have significant implications for autophagy regulation in various cancers. The current study aimed to decipher the role and mechanism of TNFAIP3 in diffuse large B-cell lymphoma (DLBCL) by modulating autophagy. METHODS: Information pertaining to the differential expression and prognostic role of TNFAIP3 in DLBCL was gleaned from the Gene Expression Omnibus (GEO) database. The TNFAIP3 expression levels in human DLBCL cells were detected by quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting. Cell counting kit-8 (CCK-8) and colony formation assays were employed to determine cell proliferation. Transwell assay and flow cytometry were applied to detect cell migration and apoptosis, respectively. Immunofluorescence and transmission electron microscope were used for the assessment of cell autophagy. The levels of apoptotic markers (caspase-3, cleaved-caspase-3, Bcl-2 Associated X (Bax), and B cell lymphoma-2 (Bcl-2)), autophagy indicators (the ratio of microtubule-associated proteins 1A/1B light chain 3 II and I (LC3II/LC3I), Sequestosome (p62)), and pathway proteins (toll-like receptor 4 (TLR4), myeloid differentiation primary response 88 (MyD88), Transcription Factor NF-Kappa-B P65 Subunit (p65), and phosphorylated-p65 (p-p65)) were assessed via Western blotting. Immunohistochemistry was employed to detect Ki67 expression in tumor tissues. RESULTS: TNFAIP3 expression in DLBCL samples was downregulated, correlating with poor prognosis. TNFAIP3 expression was also downregulated in DLBCL cells. It was found that TNFAIP3 impeded cell proliferation and migration, and enhanced apoptosis of OCI-LY3 cells. Intervention with autophagy inhibitor 3-methyladenine (3-MA) markedly reversed apoptosis of OCI-LY3 cells induced by TNFAIP3. Besides, TNFAIP3 induced autophagy via modulating the TLR4/MyD88/nuclear factor kappa B (NF-κB) signaling pathway. In vivo experiments showed that TNFAIP3 expression in DLBCL was downregulated, and upregulation of TNFAIP3 could inhibit tumor growth. CONCLUSION: TNFAIP3 inhibits DLBCL progression by inducing TLR4/MyD88/NF-κB pathway-mediated autophagy.


Subject(s)
Autophagy , Lymphoma, Large B-Cell, Diffuse , Myeloid Differentiation Factor 88 , NF-kappa B , Signal Transduction , Toll-Like Receptor 4 , Tumor Necrosis Factor alpha-Induced Protein 3 , Humans , Lymphoma, Large B-Cell, Diffuse/pathology , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/genetics , Myeloid Differentiation Factor 88/metabolism , Myeloid Differentiation Factor 88/genetics , Autophagy/genetics , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/genetics , NF-kappa B/metabolism , Cell Line, Tumor , Animals , Gene Expression Regulation, Neoplastic , Mice , Cell Proliferation , Apoptosis/genetics , Male , Female , Disease Progression , Cell Movement
10.
Sci Rep ; 14(1): 18618, 2024 08 10.
Article in English | MEDLINE | ID: mdl-39127850

ABSTRACT

This study aimed to investigate whether class A1 scavenger receptor (SR-A1) regulated macrophage polarization and gut microbial alteration during intestinal inflammation of colitis. A murine colitis model was established by feeding with dextran sulfate sodium (DSS), and treatment groups were injected intravenously with SR-A1 antibody. Results showed a preventive effect on colitis symptoms and fewer inflammatory cell infiltrates in treatment groups. Down-regulation of inflammatory cytokines and up-regulation of anti-inflammatory cytokine related to macrophages were seen in murine PBMC and LPMC after injected with SR-A1 antibody. The percentage of M2 macrophages was also elevated in treatment groups. In addition, SR-A1 antibody treatment resulted in the decreased apoptosis and increased proliferation of colonic epithelial cells. Other findings indicated that SR-A1 antibody injection could mediate its anti-inflammatory effect via inhibiting TLR4-MyD88-NF-kB signaling pathway and alterating the gut microbiota composition. Our research identified SR-A1 as a potential therapeutic target in inflammatory bowel disease (IBD).


Subject(s)
Colitis , Gastrointestinal Microbiome , Macrophages , Scavenger Receptors, Class A , Animals , Gastrointestinal Microbiome/drug effects , Colitis/immunology , Colitis/chemically induced , Colitis/microbiology , Colitis/metabolism , Mice , Macrophages/immunology , Macrophages/metabolism , Scavenger Receptors, Class A/metabolism , Dextran Sulfate/toxicity , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/immunology , Myeloid Differentiation Factor 88/metabolism , Signal Transduction , Disease Models, Animal , Cytokines/metabolism , Antibodies , NF-kappa B/metabolism , Mice, Inbred C57BL , Male , Apoptosis/drug effects
11.
Cell Mol Biol (Noisy-le-grand) ; 70(7): 206-211, 2024 Jul 28.
Article in English | MEDLINE | ID: mdl-39097873

ABSTRACT

The objective of this study was to investigate the impact of ethyl pyruvate (EP), an HMGB1 inhibitor, on ESCC cells both in vitro and in vivo. The viability of ESCC cells was assessed using the MTT method to evaluate the correlation between EP and cell viability. A scratch test was used to investigate the relationship between EP and cell migration and invasion. The effects of EP on tumor growth and survival in cancerous nude mice were examined using a tumor formation model. Immunohistochemical staining was performed to evaluate the expression levels of HMGB1, TLR4, and MyD88 in tumor tissues. EP, an anti-HMGB1 inhibitor, inhibited ESCC cell proliferation and metastasis in vitro and in vivo. Furthermore, compared with the control treatment, EP improved the activity, diet, and drinking behaviour of nude mice; inhibited tumour growth; and led to lower protein expression levels of HMGB1, TLR4, and MyD88. EP has the potential to regulate the HMGB1/TLR4-MyD88 signaling pathway, thereby inhibiting the proliferation and metastasis of ESCC, suppressing tumor growth, improving quality of life, and serving as an effective drug for ESCC treatment.


Subject(s)
Carcinoma, Squamous Cell , Cell Proliferation , Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , HMGB1 Protein , Mice, Nude , Myeloid Differentiation Factor 88 , Pyruvates , Toll-Like Receptor 4 , Animals , Pyruvates/pharmacology , Humans , HMGB1 Protein/metabolism , HMGB1 Protein/genetics , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/genetics , Myeloid Differentiation Factor 88/metabolism , Myeloid Differentiation Factor 88/genetics , Cell Line, Tumor , Esophageal Squamous Cell Carcinoma/drug therapy , Esophageal Squamous Cell Carcinoma/pathology , Esophageal Squamous Cell Carcinoma/metabolism , Esophageal Neoplasms/pathology , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/metabolism , Cell Proliferation/drug effects , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/metabolism , Xenograft Model Antitumor Assays , Cell Movement/drug effects , Mice , Signal Transduction/drug effects , Mice, Inbred BALB C , Cell Survival/drug effects , Male
12.
FASEB J ; 38(16): e23882, 2024 Aug 31.
Article in English | MEDLINE | ID: mdl-39143727

ABSTRACT

Phillygenin (PHI) is an active ingredient derived from the leaf of Forsythia suspensa that has been found to alleviate inflammation and peroxidation response. Avian infectious bronchitis (IB) is a major threat to poultry industry viral respiratory tract disease that infected with infectious bronchitis virus (IBV). This study investigated the protection of PHI to CEK cell and broiler's tracheal injury triggered by avian infectious bronchitis virus (IBV). The results showed that IBV infection did not cause serious clinical symptoms and slowing-body weight in PHI-treated broilers. The expression of virus loads, pro-inflammation factors (IL-6, TNF-α, and IL-1ß) in CEK cell, and tracheas were decreased compared to the IBV group, exhibiting its potent anti-inflammation. Mechanistically, the study demonstrated that the inhibition of TLR7/MyD88/NF-κB pathway was mainly involved in the protection effect of PHI to inflammation injury. Interestingly, a higher abundance of Firmicutes and Lactobacillus in respiratory tract was observed in PHI-treated broilers than in the IBV group. Significant differences were observed between the IBV group and PHI-treated group in the Ferroptosis, Tryptophan metabolism, and Glutathione metabolism pathways. PHI exhibited potent protection effect on IBV infection and alleviated inflammation injury, mainly through inhibiting TLR7/MyD88/NF-κB pathway. The study encourages further development of PHI, paving the way to its clinical use as a new candidate drug to relieve IBV-induced respiratory symptoms.


Subject(s)
Chickens , Coronavirus Infections , Infectious bronchitis virus , Myeloid Differentiation Factor 88 , NF-kappa B , Poultry Diseases , Toll-Like Receptor 7 , Animals , NF-kappa B/metabolism , Myeloid Differentiation Factor 88/metabolism , Poultry Diseases/drug therapy , Poultry Diseases/microbiology , Poultry Diseases/metabolism , Toll-Like Receptor 7/metabolism , Coronavirus Infections/drug therapy , Coronavirus Infections/veterinary , Microbiota/drug effects , Signal Transduction/drug effects
13.
Food Chem Toxicol ; 191: 114893, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39067743

ABSTRACT

Particulate matter (PM) poses significant health risks, especially fine particles (PM2.5) that can cause severe lung injuries. Lupeol, a phytosterol from medicinal plants, has potential anti-cancer properties. This study investigated lupeol's protective effects against PM2.5-induced lung damage. Mice received lupeol following intratracheal PM2.5 exposure. Results showed lupeol reduced lung damage, lowered wet/dry (W/D) weight ratio, and suppressed increased permeability caused by PM2.5. Additionally, lupeol decreased plasma inflammatory cytokines, total protein concentration in bronchoalveolar lavage fluid (BALF), and PM2.5-induced lymphocyte proliferation. Lupeol also reduced expression of toll-like receptor 4 (TLR4), myeloid differentiation primary response 88 (MyD88), and autophagy-related proteins microtubule-associated protein 1 A/1 B-light chain 3 (LC3) II and Beclin 1, while increasing phosphorylated mammalian target of rapamycin (mTOR) phosphorylation. These findings suggest lupeol's potential as a therapeutic agent for PM2.5-induced lung damage via modulation of the TLR4-MyD88 and mTOR-autophagy pathways.


Subject(s)
Particulate Matter , Pentacyclic Triterpenes , Pneumonia , Toll-Like Receptor 4 , Animals , Pentacyclic Triterpenes/pharmacology , Particulate Matter/toxicity , Toll-Like Receptor 4/metabolism , Pneumonia/drug therapy , Pneumonia/metabolism , Pneumonia/chemically induced , Mice , Male , Myeloid Differentiation Factor 88/metabolism , Myeloid Differentiation Factor 88/genetics , TOR Serine-Threonine Kinases/metabolism , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Autophagy/drug effects , Mice, Inbred C57BL , Cytokines/metabolism , Lung/drug effects , Lung/metabolism , Lung/pathology , Beclin-1/metabolism , Microtubule-Associated Proteins/metabolism , Lupanes
14.
J Ethnopharmacol ; 334: 118520, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-38964626

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Inflammation is directly related to disease progression and contributes significantly to the global burden of disease. Pothos chinensis (Raf.) Merr. (PCM) is commonly used in Yao medicine in China to treat tumors, and orthopedic illnesses such as knee osteoarthritis, and rheumatic bone discomfort. PCM was found to have significant anti-inflammatory properties in previous studies. AIM OF THE STUDY: To explore the active compounds of PCM and their anti-inflammatory pharmacological mechanisms through an integrated strategy of serum pharmacochemistry, network pharmacology, and serum metabolomics. MATERIALS AND METHODS: The qualitative and quantitative analyses of the chemical components of PCM were performed using UPLC-QTOF-MS/MS and UPLC, respectively, and the prototype components of PCM absorbed into the blood were analyzed. Based on the characterized absorbed into blood components, potential targets and signaling pathways of PCM anti-inflammatory were found using network pharmacology. Furthermore, metabolomics studies using UPLC-QTOF-MS/MS identified biomarkers and metabolic pathways related to the anti-inflammatory effects of PCM. Finally, the hypothesized mechanisms were verified by in vivo and in vitro experiments. RESULTS: Forty chemical components from PCM were identified for the first time, and seven of them were quantitatively analyzed, while five serum migratory prototype components were found. Network pharmacology KEGG enrichment analysis revealed that arachidonic acid metabolism, Tyrosine metabolism, TNF signaling pathway, NF-κB signaling pathway, and phenylalanine metabolism were the main signaling pathways of PCM anti-inflammatory. Pharmacodynamic results showed that PCM ameliorated liver injury and inflammatory cell infiltration and downregulated protein expression of IL-1ß, NF-κB p65, and MyD88 in the liver. Metabolomics studies identified 53 different serum metabolites, mainly related to purine and pyrimidine metabolism, phenylalanine metabolism, primary bile acid biosynthesis, and glycerophospholipid metabolism. The comprehensive results demonstrated that the anti-inflammatory modulatory network of PCM was related to 5 metabolites, 3 metabolic pathways, 7 targets, and 4 active components of PCM. In addition, molecular docking identified the binding ability between the active ingredients and the core targets, and the anti-inflammatory efficacy of the active ingredients was verified by in vitro experiments. CONCLUSION: Our study demonstrated the anti-inflammatory effect of PCM, and these findings provide new insights into the active ingredients and metabolic mechanisms of PCM in anti-inflammation.


Subject(s)
Anti-Inflammatory Agents , Metabolomics , Myeloid Differentiation Factor 88 , NF-kappa B , Network Pharmacology , Signal Transduction , Toll-Like Receptor 4 , Anti-Inflammatory Agents/pharmacology , Animals , NF-kappa B/metabolism , Toll-Like Receptor 4/metabolism , Myeloid Differentiation Factor 88/metabolism , Male , Signal Transduction/drug effects , Mice , Rats, Sprague-Dawley , Tandem Mass Spectrometry , Drugs, Chinese Herbal/pharmacology , Animals, Outbred Strains
15.
Int Immunopharmacol ; 139: 112660, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39018688

ABSTRACT

Cardiac tissue remodeling is characterized by altered heart tissue architecture and dysfunction, leading to heart failure. Sustained activation of the renin-angiotensin-aldosterone system (RAAS) greatly promotes the development of myocardial remodeling. Angiotensin II (Ang II), which is the major component of RAAS, can directly lead to cardiac remodeling by inducing an inflammatory response. Schisandrin B (Sch B), the active component extracted from the fruit of Schisandra chinensis (Turcz.) Baill has been shown to exhibit anti-inflammatory activity through its ability to target TLR4 and its adaptor protein, MyD88. In this study, we explored whether Sch B alleviates Ang II-induced myocardial inflammation and remodeling via targeting MyD88. Sch B significantly suppressed Ang II-induced inflammation as well as increased the expression of several genes of tissue remodeling (ß-Mhc, Tgfb, Anp, α-Ska) both in vivo and in vitro. These protective effects of Sch B were due to the inhibition of recruitment of MyD88 to TLR2 and TLR4, suppressing the Ang II-induced NF-κB activation and reducing the following inflammatory responses. Moreover, the knockdown of Myd88 in cardiomyocytes abrogated the Ang II-induced increases in the production of inflammatory cytokines and expression of remodeling genes. These findings provide new evidence that the mechanism of Sch B protection was attributed to selective inhibition of MyD88 signaling. This finding could pave the way for novel therapeutic strategies for myocardial inflammatory diseases.


Subject(s)
Angiotensin II , Cyclooctanes , Lignans , Mice, Inbred C57BL , Myeloid Differentiation Factor 88 , Myocytes, Cardiac , Polycyclic Compounds , Toll-Like Receptor 4 , Animals , Cyclooctanes/pharmacology , Cyclooctanes/therapeutic use , Lignans/pharmacology , Lignans/therapeutic use , Myeloid Differentiation Factor 88/metabolism , Polycyclic Compounds/pharmacology , Polycyclic Compounds/therapeutic use , Angiotensin II/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/genetics , Mice , Male , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 2/genetics , Ventricular Remodeling/drug effects , Signal Transduction/drug effects , Cells, Cultured , NF-kappa B/metabolism
16.
Fish Shellfish Immunol ; 151: 109734, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38950759

ABSTRACT

Toll-like receptors (TLRs) are pattern recognition receptors that trigger host immune responses against various pathogens by detecting evolutionarily conserved pathogen-associated molecular patterns (PAMPs). TLR21 is a member of the Toll-like receptor family, and emerging data suggest that it recognises unmethylated CpG DNA and is considered a functional homologue of mammalian TLR9. However, little is known regarding the role of TLR21 in the fish immune response. In the present study, we isolated the cDNA sequence of TLR21 from the largemouth bass (Micropterus salmoides) and termed it MsTLR21. The MsTLR21 gene contained an open reading frame (ORF) of 2931 bp and encodes a polypeptide of 976 amino acids. The predicted MsTLR21 protein has two conserved domains, a conserved leucine-rich repeats (LRR) domain and a C-terminal Toll-interleukin (IL) receptor (TIR) domain, similar to those of other fish and mammals. In healthy largemouth bass, the TLR21 transcript was broadly expressed in all the examined tissues, with the highest expression levels in the gills. After challenge with Nocardia seriolae and polyinosinic polycytidylic acid (Poly[I:C]), the expression of TLR21 mRNA was upregulated or downregulated in all tissues tested. Overexpression of TLR21 in 293T cells showed that it has a positive regulatory effect on nuclear factor-kappaB (NF-κB) and interferons-ß (IFN-ß) activity. Subcellular localisation analysis showed that TLR21 was expressed in the cytoplasm. We performed pull-down assays and determined that TLR21 did not interact with myeloid differentiation primary response gene 88 (Myd88); however, it interacted with TIR domain-containing adaptor inducing interferon-ß (TRIF). Taken together, these findings suggest that MsTLR21 plays important roles in TLR/IL-1R signalling pathways and the immune response to pathogen invasion.


Subject(s)
Adaptor Proteins, Vesicular Transport , Amino Acid Sequence , Bass , Fish Diseases , Fish Proteins , NF-kappa B , Phylogeny , Animals , Bass/immunology , Bass/genetics , Fish Proteins/genetics , Fish Proteins/immunology , Fish Proteins/chemistry , NF-kappa B/genetics , NF-kappa B/metabolism , NF-kappa B/immunology , Fish Diseases/immunology , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/immunology , Adaptor Proteins, Vesicular Transport/chemistry , Adaptor Proteins, Vesicular Transport/metabolism , Signal Transduction/immunology , Gene Expression Regulation/immunology , Immunity, Innate/genetics , Sequence Alignment/veterinary , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Myeloid Differentiation Factor 88/immunology , Myeloid Differentiation Factor 88/chemistry , Gene Expression Profiling/veterinary , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology , Toll-Like Receptors/chemistry , Toll-Like Receptors/metabolism , Base Sequence
17.
Fish Shellfish Immunol ; 151: 109744, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38960107

ABSTRACT

MicroRNAs (miRNAs) have been demonstrated to act as crucial modulators with considerable impacts on the immune system. Cottonseed meal is often used as a protein source in aqua feed, cottonseed meal contains gossypol, which is harmful to animals. However, there is a lack of research on the role of miRNAs in fish exposed to gossypol stress. To determine the regulatory effects of miRNAs on gossypol toxicity, Cyprinus carpio were given to oral administration of 20 mg/kg gossypol for 7 days, and the gossypol concentration in the tissues was tested. Then, we detected spleen index, histology, immune enzyme activities of fish induced by gossypol. The results of miRNA sequencing revealed 8 differentially expressed miRNAs in gossypol group, and miR-214_L-1R+4 was found involved in immune response induced by gossypol. The potential targets of miR-214_L-1R+4 were predicted, and found a putative miR-214_L-1R+4 binding site in the 3'UTR of MyD88a. Furthermore, dual-luciferase reporter assays displayed miR-214_L-1R+4 decreased MyD88a expression through binding to the 3'UTR of MyD88a. Moreover, miR-214_L-1R+4 antagomir were intraperitoneally administered to C. carpio, down-regulated miR-214_L-1R+4 could increase MyD88a expression, as well as inflammatory cytokines and anti-inflammatory cytokines expression. These findings revealed that miR-214_L-1R+4 via the MyD88-dependent signaling pathway modulate the immune response to gossypol in C. carpio spleen.


Subject(s)
Carps , Fish Proteins , Gossypol , MicroRNAs , Myeloid Differentiation Factor 88 , Signal Transduction , Animals , Carps/immunology , Carps/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Gossypol/pharmacology , Gossypol/administration & dosage , Signal Transduction/drug effects , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Fish Proteins/genetics , Fish Proteins/immunology , Fish Proteins/metabolism , Immunity, Innate/drug effects , Immunity, Innate/genetics
18.
Nat Commun ; 15(1): 6067, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39025856

ABSTRACT

After recognizing its ligand lipopolysaccharide, Toll-like receptor 4 (TLR4) recruits adaptor proteins to the cell membrane, thereby initiating downstream signaling and triggering inflammation. Whether this recruitment of adaptor proteins is dependent solely on protein-protein interactions is unknown. Here, we report that the sphingolipid sphinganine physically interacts with the adaptor proteins MyD88 and TIRAP and promotes MyD88 recruitment in macrophages. Myeloid cell-specific deficiency in serine palmitoyltransferase long chain base subunit 2, which encodes the key enzyme catalyzing sphingolipid biosynthesis, decreases the membrane recruitment of MyD88 and inhibits inflammatory responses in in vitro bone marrow-derived macrophage and in vivo sepsis models. In a melanoma mouse model, serine palmitoyltransferase long chain base subunit 2 deficiency decreases anti-tumor myeloid cell responses and increases tumor growth. Therefore, sphinganine biosynthesis is required for the initiation of TLR4 signal transduction and serves as a checkpoint for macrophage pattern recognition in sepsis and melanoma mouse models.


Subject(s)
Macrophages , Melanoma , Myeloid Differentiation Factor 88 , Sepsis , Serine C-Palmitoyltransferase , Sphingosine , Toll-Like Receptor 4 , Animals , Toll-Like Receptor 4/metabolism , Sepsis/metabolism , Macrophages/metabolism , Myeloid Differentiation Factor 88/metabolism , Mice , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Melanoma/metabolism , Melanoma/pathology , Melanoma/genetics , Serine C-Palmitoyltransferase/metabolism , Serine C-Palmitoyltransferase/genetics , Humans , Signal Transduction , Disease Models, Animal , Inflammation/metabolism , Receptors, Interleukin-1/metabolism , Receptors, Interleukin-1/genetics , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Mice, Inbred C57BL , Mice, Knockout , HEK293 Cells , Lipopolysaccharides
19.
Nat Immunol ; 25(9): 1678-1691, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39060650

ABSTRACT

Whole-exome sequencing of two unrelated kindreds with systemic autoimmune disease featuring antinuclear antibodies with IgG4 elevation uncovered an identical ultrarare heterozygous TNIP1Q333P variant segregating with disease. Mice with the orthologous Q346P variant developed antinuclear autoantibodies, salivary gland inflammation, elevated IgG2c, spontaneous germinal centers and expansion of age-associated B cells, plasma cells and follicular and extrafollicular helper T cells. B cell phenotypes were cell-autonomous and rescued by ablation of Toll-like receptor 7 (TLR7) or MyD88. The variant increased interferon-ß without altering nuclear factor kappa-light-chain-enhancer of activated B cells signaling, and impaired MyD88 and IRAK1 recruitment to autophagosomes. Additionally, the Q333P variant impaired TNIP1 localization to damaged mitochondria and mitophagosome formation. Damaged mitochondria were abundant in the salivary epithelial cells of Tnip1Q346P mice. These findings suggest that TNIP1-mediated autoimmunity may be a consequence of increased TLR7 signaling due to impaired recruitment of downstream signaling molecules and damaged mitochondria to autophagosomes and may thus respond to TLR7-targeted therapeutics.


Subject(s)
Autoimmune Diseases , DNA-Binding Proteins , Immunoglobulin G , Myeloid Differentiation Factor 88 , Toll-Like Receptor 7 , Animals , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Humans , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/immunology , Mice , Myeloid Differentiation Factor 88/metabolism , Myeloid Differentiation Factor 88/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Female , Male , Signal Transduction , Mitochondria/metabolism , Exome Sequencing , Antibodies, Antinuclear/immunology , B-Lymphocytes/immunology , Mice, Knockout , Mice, Inbred C57BL , Germinal Center/immunology , Pedigree , Salivary Glands/immunology , Salivary Glands/metabolism , Salivary Glands/pathology , Membrane Glycoproteins
20.
Gene ; 928: 148768, 2024 Nov 30.
Article in English | MEDLINE | ID: mdl-39013482

ABSTRACT

Although antiviral drugs can effectively inhibit hepatitis B virus (HBV) replication, the maintenance of chronic inflammation in the liver is still considered to be an important cause for the progression of HBV-related liver disease to liver fibrosis and advanced liver disease. As an endogenous inhibitory receptor of IL-1R and TLR signaling pathways, single immunoglobulin interleukin-1-related receptor (SIGIRR) has been proven to reduce inflammation in tissues to maintain system homeostasis. However, the relationship between SIGIRR expression and HBV replication and inflammatory pathway activation in hepatocytes remains unclear. In this study, hepatitis B virus X protein (HBx) upregulated MyD88 in liver cells, promoting NF-κB signaling and inflammatory factor production with LPS treatment, and the cell supernatant accelerated the activation and collagen secretion of hepatic stellate cells. However, SIGIRR overexpression suppressed HBx-mediated MyD88/NF-κB inflammatory signaling activation and inflammatory cytokine production induced by LPS in hepatocytes and HBV replication hepatocytes. Although we did not find any effect of SIGIRR on HBV replication in vitro, this study investigated the role of SIGIRR in blocking the proinflammatory function of HBx, which may provide a new idea for the treatment of chronic hepatitis B.


Subject(s)
Hepatitis B virus , Hepatocytes , Inflammation , Myeloid Differentiation Factor 88 , NF-kappa B , Receptors, Interleukin-1 , Signal Transduction , Trans-Activators , Viral Regulatory and Accessory Proteins , Hepatocytes/metabolism , Hepatocytes/virology , Humans , Receptors, Interleukin-1/metabolism , Receptors, Interleukin-1/genetics , Myeloid Differentiation Factor 88/metabolism , Myeloid Differentiation Factor 88/genetics , NF-kappa B/metabolism , Hepatitis B virus/physiology , Trans-Activators/genetics , Trans-Activators/metabolism , Inflammation/metabolism , Inflammation/genetics , Hepatitis B, Chronic/virology , Hepatitis B, Chronic/genetics , Hepatitis B, Chronic/metabolism , Virus Replication , Lipopolysaccharides , Hep G2 Cells , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/virology
SELECTION OF CITATIONS
SEARCH DETAIL