Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62.935
Filtrar
1.
Sci Rep ; 14(1): 12827, 2024 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-38834834

RESUMO

Gut microbiota plays a crucial role in gastrointestinal tumors. Additionally, gut microbes influence the progression of esophageal cancer. However, the major bacterial genera that affect the invasion and metastasis of esophageal cancer remain unknown, and the underlying mechanisms remain unclear. Here, we investigated the gut flora and metabolites of patients with esophageal squamous cell carcinoma and found abundant Bacteroides and increased secretion and entry of the surface antigen lipopolysaccharide (LPS) into the blood, causing inflammatory changes in the body. We confirmed these results in a mouse model of 4NQO-induced esophageal carcinoma in situ and further identified epithelial-mesenchymal transition (EMT) occurrence and TLR4/Myd88/NF-κB pathway activation in mouse esophageal tumors. Additionally, in vitro experiments revealed that LPS from Bacteroides fragile promoted esophageal cancer cell proliferation, migration, and invasion, and induced EMT by activating the TLR4/Myd88/NF-κB pathway. These results reveal that Bacteroides are closely associated with esophageal cancer progression through a higher inflammatory response level and signaling pathway activation that are both common to inflammation and tumors induced by LPS, providing a new biological target for esophageal cancer prevention or treatment.


Assuntos
Transição Epitelial-Mesenquimal , Neoplasias Esofágicas , Carcinoma de Células Escamosas do Esôfago , Lipopolissacarídeos , Fator 88 de Diferenciação Mieloide , NF-kappa B , Transdução de Sinais , Receptor 4 Toll-Like , Receptor 4 Toll-Like/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Animais , NF-kappa B/metabolismo , Humanos , Neoplasias Esofágicas/patologia , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/microbiologia , Camundongos , Carcinoma de Células Escamosas do Esôfago/metabolismo , Carcinoma de Células Escamosas do Esôfago/patologia , Carcinoma de Células Escamosas do Esôfago/microbiologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Linhagem Celular Tumoral , Invasividade Neoplásica , Inflamação/metabolismo , Inflamação/patologia , Bacteroidetes , Microbioma Gastrointestinal , Movimento Celular/efeitos dos fármacos , Masculino , Metástase Neoplásica , Proliferação de Células , Feminino
2.
Mol Biol Rep ; 51(1): 709, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38824265

RESUMO

BACKGROUND: Cystatin is a protease inhibitor that also regulates genes expression linked to inflammation and plays a role in defense and regulation. METHODS AND RESULTS: Cystatin 10 (Smcys10) was cloned from Scophthalmus maximus and encodes a 145 amino acid polypeptide. The results of qRT-PCR showed that Smcys10 exhibited tissue-specific expression patterns, and its expression was significantly higher in the skin than in other tissues. The expression level of Smcys10 was significantly different in the skin, gill, head kidney, spleen and macrophages after Vibrio anguillarum infection, indicating that Smcys10 may play an important role in resistance to V. anguillarum infection. The recombinant Smcys10 protein showed binding and agglutinating activity in a Ca2+-dependent manner against bacteria. rSmcys10 treatment upregulated the expression of IL-10, TNF-α and TGF-ß in macrophages of turbot and hindered the release of lactate dehydrogenase (LDH) from macrophages after V. anguillarum infection, which confirmed that rSmcys10 reduced the damage to macrophages by V. anguillarum. The NF-κB pathway was suppressed by Smcys10, as demonstrated by dual-luciferase analysis. CONCLUSIONS: These results indicated that Smcys10 is involved in the host antibacterial immune response.


Assuntos
Cistatinas , Doenças dos Peixes , Proteínas de Peixes , Linguados , Macrófagos , Vibrio , Animais , Linguados/imunologia , Linguados/genética , Linguados/metabolismo , Vibrio/patogenicidade , Cistatinas/genética , Cistatinas/metabolismo , Proteínas de Peixes/genética , Proteínas de Peixes/metabolismo , Proteínas de Peixes/imunologia , Macrófagos/metabolismo , Macrófagos/imunologia , Doenças dos Peixes/imunologia , Doenças dos Peixes/genética , Doenças dos Peixes/microbiologia , Vibrioses/imunologia , Vibrioses/veterinária , Vibrioses/genética , NF-kappa B/metabolismo , Clonagem Molecular/métodos , Regulação da Expressão Gênica
3.
Pak J Pharm Sci ; 37(2(Special)): 463-473, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38822551

RESUMO

Solanum lyratum Thunb., a traditional Chinese herbal medicine, has a promising background. However, the anti-inflammatory effects of its component steroid alkaloid have not been explored. In this study, animal and cell experiments were performed to investigate the anti-inflammatory effects and mechanism of action of Solanum lyratum Thunb steroid alkaloid (SLTSA), in order to provide evidence for its potential utilization. SLTSA effectively inhibited ear swelling and acute abdominal inflammation of mice. We observed concentration-dependent inhibition of pro-inflammatory cytokines by SLTSA, as confirmed by the ELISA and RT-qPCR results. Flow cytometry, immunofluorescence and RT-qPCR analyses revealed that SLTSA suppressed TLR4 expression. Western blot results indicated that SLTSA inhibited the activation of the TLR4/MyD88/NF-κB signaling pathway. Our study demonstrated that SLTSA possesses anti-inflammatory properties.


Assuntos
Alcaloides , Anti-Inflamatórios , Transdução de Sinais , Solanum , Animais , Solanum/química , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/isolamento & purificação , Camundongos , Alcaloides/farmacologia , Alcaloides/isolamento & purificação , Transdução de Sinais/efeitos dos fármacos , NF-kappa B/metabolismo , Receptor 4 Toll-Like/metabolismo , Citocinas/metabolismo , Células RAW 264.7 , Fator 88 de Diferenciação Mieloide/metabolismo , Masculino
4.
J Nanobiotechnology ; 22(1): 314, 2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38840113

RESUMO

Osteoporosis is the most common bone metabolic disease that affects the health of middle-aged and elderly people, which is hallmarked by imbalanced bone remodeling and a deteriorating immune microenvironment. Magnesium and calcium are pivotal matrix components that participate in the bone formation process, especially in the immune microenvironment regulation and bone remodeling stages. Nevertheless, how to potently deliver magnesium and calcium to bone tissue remains a challenge. Here, we have constructed a multifunctional nanoplatform composed of calcium-based upconversion nanoparticles and magnesium organic frameworks (CM-NH2-PAA-Ald, denoted as CMPA), which features bone-targeting and pH-responsive properties, effectively regulating the inflammatory microenvironment and promoting the coordination of osteogenic functions for treating osteoporosis. The nanoplatform can efficaciously target bone tissue and gradually degrade in response to the acidic microenvironment of osteoporosis to release magnesium and calcium ions. This study validates that CMPA possessing favorable biocompatibility can suppress inflammation and facilitate osteogenesis to treat osteoporosis. Importantly, high-throughput sequencing results demonstrate that the nanoplatform exerts a good inflammatory regulation effect through inhibition of the nuclear factor kappa-B signaling pathway, thereby normalizing the osteoporotic microenvironment. This collaborative therapeutic strategy that focuses on improving bone microenvironment and promoting osteogenesis provides new insight for the treatment of metabolic diseases such as osteoporosis.


Assuntos
Cálcio , Magnésio , Nanopartículas , Osteogênese , Osteoporose , Osteogênese/efeitos dos fármacos , Osteoporose/tratamento farmacológico , Magnésio/farmacologia , Magnésio/química , Cálcio/metabolismo , Animais , Nanopartículas/química , Camundongos , Inflamação/tratamento farmacológico , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/metabolismo , Humanos , Microambiente Celular/efeitos dos fármacos , Feminino , NF-kappa B/metabolismo
5.
J Transl Med ; 22(1): 535, 2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38840216

RESUMO

BACKGROUND: Inflammation and endothelial barrier dysfunction are the major pathophysiological changes in acute respiratory distress syndrome (ARDS). Sphingosine-1-phosphate receptor 3 (S1PR3), a G protein-coupled receptor, has been found to mediate inflammation and endothelial cell (EC) integrity. However, the function of S1PR3 in ARDS has not been fully elucidated. METHODS: We used a murine lipopolysaccharide (LPS)-induced ARDS model and an LPS- stimulated ECs model to investigate the role of S1PR3 in anti-inflammatory effects and endothelial barrier protection during ARDS. RESULTS: We found that S1PR3 expression was increased in the lung tissues of mice with LPS-induced ARDS. TY-52156, a selective S1PR3 inhibitor, effectively attenuated LPS-induced inflammation by suppressing the expression of proinflammatory cytokines and restored the endothelial barrier by repairing adherens junctions and reducing vascular leakage. S1PR3 inhibition was achieved by an adeno-associated virus in vivo and a small interfering RNA in vitro. Both the in vivo and in vitro studies demonstrated that pharmacological or genetic inhibition of S1PR3 protected against ARDS by inhibiting the NF-κB pathway and improving mitochondrial oxidative phosphorylation. CONCLUSIONS: S1PR3 inhibition protects against LPS-induced ARDS via suppression of pulmonary inflammation and promotion of the endothelial barrier by inhibiting NF-κB and improving mitochondrial oxidative phosphorylation, indicating that S1PR3 is a potential therapeutic target for ARDS.


Assuntos
Lipopolissacarídeos , Camundongos Endogâmicos C57BL , Mitocôndrias , NF-kappa B , Fosforilação Oxidativa , Síndrome do Desconforto Respiratório , Receptores de Esfingosina-1-Fosfato , Animais , Síndrome do Desconforto Respiratório/induzido quimicamente , Síndrome do Desconforto Respiratório/metabolismo , Síndrome do Desconforto Respiratório/patologia , Receptores de Esfingosina-1-Fosfato/metabolismo , Receptores de Esfingosina-1-Fosfato/antagonistas & inibidores , NF-kappa B/metabolismo , Mitocôndrias/metabolismo , Mitocôndrias/efeitos dos fármacos , Fosforilação Oxidativa/efeitos dos fármacos , Masculino , Receptores de Lisoesfingolipídeo/metabolismo , Receptores de Lisoesfingolipídeo/antagonistas & inibidores , Humanos , Pulmão/patologia , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Camundongos , Inflamação/patologia , Células Endoteliais/metabolismo , Células Endoteliais/efeitos dos fármacos , Substâncias Protetoras/farmacologia , Citocinas/metabolismo
6.
J Cell Mol Med ; 28(11): e18476, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38842136

RESUMO

Osteoarthritis (OA) is a complicated disease that involves apoptosis and mitophagy. MST1 is a pro-apoptotic factor. Hence, decreasing its expression plays an anti-apoptotic effect. This study aims to investigate the protective effect of MST1 inhibition on OA and the underlying processes. Immunofluorescence (IF) was used to detect MST1 expression in cartilage tissue. Western Blot, ELISA and IF were used to analyse the expression of inflammation, extracellular matrix (ECM) degradation, apoptosis and mitophagy-associated proteins. MST1 expression in chondrocytes was inhibited using siRNA and shRNA in vitro and in vivo. Haematoxylin-Eosin, Safranin O-Fast Green and alcian blue staining were used to evaluate the therapeutic effect of inhibiting MST1. This study discovered that the expression of MST1 was higher in OA patients. Inhibition of MST1 reduced inflammation, ECM degradation and apoptosis and enhanced mitophagy in vitro. MST1 inhibition slows OA progression in vivo. Inhibiting MST1 suppressed apoptosis, inflammation and ECM degradation via promoting Parkin-mediated mitophagy and the Nrf2-NF-κB axis. The results suggest that MST1 is a possible therapeutic target for the treatment of osteoarthritis as its inhibition delays the progression of OA through the Nrf2-NF-κB axis and mitophagy.


Assuntos
Apoptose , Condrócitos , Progressão da Doença , Mitofagia , Fator 2 Relacionado a NF-E2 , NF-kappa B , Osteoartrite , Transdução de Sinais , Ubiquitina-Proteína Ligases , Osteoartrite/metabolismo , Osteoartrite/patologia , Osteoartrite/genética , Mitofagia/genética , Fator 2 Relacionado a NF-E2/metabolismo , Fator 2 Relacionado a NF-E2/genética , NF-kappa B/metabolismo , Humanos , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina-Proteína Ligases/genética , Animais , Condrócitos/metabolismo , Condrócitos/patologia , Apoptose/genética , Masculino , Camundongos , Matriz Extracelular/metabolismo , Técnicas de Silenciamento de Genes , Inflamação/patologia , Inflamação/metabolismo , Inflamação/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Peptídeos e Proteínas de Sinalização Intracelular
7.
Crit Rev Eukaryot Gene Expr ; 34(5): 45-57, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38842203

RESUMO

Inflammation-mediated dysfunction of cardiomyocytes is the main cause of diabetic cardiomyopathy (DCM). The present study aimed to investigate the roles of siah E3 ubiquitin protein ligase 1 (SIAH1) in DCM. The online dataset GSE4172 was used to analyze the differentially expressed genes in myocardial inflammation of DCM patients. RT-qPCR was conducted to detect mRNA levels. Enzyme-Linked Immunosorbent Assay (ELISA) was performed to detect cytokine release. Western blot was used to detect protein expression. Lactate dehydrogenase (LDH) assay was used to determine cytotoxicity. In vitro ubiquitination assay was applied to determine the ubiquitination of nuclear factor kappa B inhibitor alpha (1κВ-α). Terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assay was used to detect the death of cardiomyocytes. Flow cytometry was applied for determining cardiomyocyte pyroptosis. The results showed that SIAH1 was overexpressed in human inflammatory cardiomyopathy. High expression of SIAH1 was associated with inflammatory response. SIAH1 was also overexpressed lipopolysaccharide (LPS)-induced inflammatory cardiomyopathy model in vitro. However, SIAH1 knockdown suppressed the inflammatory-related pyroptosis of cardiomyocytes. SIAH1 promoted the ubiquitination of 1κВ-α and activated nuclear factor kappa В (NF-κВ) signaling, which promoted the pyroptosis of cardiomyocytes. In conclusion, SIAH1 exacerbated the progression of human inflammatory cardiomyopathy via inducing the ubiquitination of 1κВ-α and activation of NF-κВ signaling. Therefore, SIAHI/IκB-α/NF-κB signaling may be a potential target for human inflammatory cardiomyopathy.


Assuntos
Cardiomiopatias Diabéticas , Miócitos Cardíacos , NF-kappa B , Piroptose , Transdução de Sinais , Ubiquitina-Proteína Ligases , Cardiomiopatias Diabéticas/metabolismo , Cardiomiopatias Diabéticas/patologia , Cardiomiopatias Diabéticas/genética , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Humanos , NF-kappa B/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina-Proteína Ligases/genética , Inibidor de NF-kappaB alfa/metabolismo , Inibidor de NF-kappaB alfa/genética , Ubiquitinação , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Inflamação/metabolismo , Inflamação/patologia , Inflamação/genética
9.
Inhal Toxicol ; 36(4): 275-281, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38836332

RESUMO

Multiwalled carbon nanotubes (MWCNTs) have numerous applications in the field of carbon nanomaterials. However, the associated toxicity concerns have increased significantly because of their widespread use. The inhalation of MWCNTs can lead to nanoparticle deposition in the lung tissue, causing inflammation and health risks. In this study, celastrol, a natural plant medicine with potent anti-inflammatory properties, effectively reduced the number of inflammatory cells, including white blood cells, neutrophils, and lymphocytes, and levels of inflammatory cytokines, such as IL-1ß, IL-6, and TNF-α, in mice lungs exposed to MWCNTs. Moreover, celastrol inhibited the activation of the NF-κB-signaling pathway. This study confirmed these findings by demonstrating comparable reductions in inflammation upon exposure to MWCNTs in mice with the deletion of NF-κB (P50-/-). These results indicate the utility of celastrol as a promising pharmacological agent for preventing MWCNT-induced lung tissue inflammation.


Assuntos
Camundongos Endogâmicos C57BL , NF-kappa B , Nanotubos de Carbono , Triterpenos Pentacíclicos , Pneumonia , Transdução de Sinais , Triterpenos , Animais , Triterpenos Pentacíclicos/farmacologia , Nanotubos de Carbono/toxicidade , Transdução de Sinais/efeitos dos fármacos , Triterpenos/farmacologia , Pneumonia/induzido quimicamente , Pneumonia/tratamento farmacológico , Pneumonia/prevenção & controle , Pneumonia/metabolismo , NF-kappa B/metabolismo , Masculino , Pulmão/efeitos dos fármacos , Pulmão/patologia , Pulmão/metabolismo , Anti-Inflamatórios/farmacologia , Citocinas/metabolismo , Camundongos , Camundongos Knockout , Líquido da Lavagem Broncoalveolar/citologia , Líquido da Lavagem Broncoalveolar/química
10.
Cell Mol Biol (Noisy-le-grand) ; 70(6): 211-216, 2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38836660

RESUMO

This study investigated the regulatory impact of Toll-like receptor 4 (TLR4) gene on glioma cell proliferation and apoptosis, elucidating the molecular mechanisms underlying TLR4-induced growth inhibition in vivo. U-87MG-Sh and U-87MG-NC cells, with silenced TLR4 and negative control plasmid respectively, were established. Eighteen nude mice, divided into transfection, negative control, and blank control groups, were inoculated with corresponding cells. Over four weeks, the transfection group exhibited significantly reduced tumor growth rates, smaller mass and volume, and lower growth activity compared to controls. Histological analysis revealed sparse tumor cells, increased fibrous connective tissue, and slower angiogenesis in the transfection group. Flow cytometry demonstrated a lower proliferation index and increased G0/1 cell count in the transfection group. mRNA levels of TLR4, NF-κB, and CyclinD1 were significantly lower in the transfection group. TLR4 silencing correlated with U-87MG cell proliferation regulation, growth inhibition, NF-κB and CyclinD1 modulation, and induction of cell cycle arrest and apoptosis. These findings suggest TLR4 as a potential gene therapy target for glioma.


Assuntos
Apoptose , Proliferação de Células , Ciclina D1 , Inativação Gênica , Glioma , Camundongos Nus , NF-kappa B , Receptor 4 Toll-Like , Animais , Receptor 4 Toll-Like/metabolismo , Receptor 4 Toll-Like/genética , Glioma/patologia , Glioma/genética , Glioma/metabolismo , Proliferação de Células/genética , Linhagem Celular Tumoral , Apoptose/genética , Humanos , NF-kappa B/metabolismo , Ciclina D1/metabolismo , Ciclina D1/genética , Camundongos , Pontos de Checagem do Ciclo Celular/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Regulação Neoplásica da Expressão Gênica , Camundongos Endogâmicos BALB C
11.
Cell Mol Biol (Noisy-le-grand) ; 70(6): 224-232, 2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38836655

RESUMO

Asthenospermia is a predominant cause of male infertility, and antioxidant supplements can be effective in treating asthenospermia. We demonstrate the antioxidant potential of traditional Chinese medicine, the Yishenhuoxue (YSHX) formula, in treating polyglycosides of Tripterygium wilfordii (GTW)-induced asthenospermia in rats. Fifty male rats were randomly divided into the normal, model, and treatment groups. HE staining was used to evaluate the improvement of spermatogenic function of rats, and TBA reaction, qRT-PCR, Western Blot and other methods were used to determine the changes of oxidative stress indicators and to evaluate the improvement of antioxidant capacity of rats by YSHX. Comparison with the model group showed significant improvement in pathological damage caused by GTW to seminiferous tubules. MDA and NO content in rat testes decreased, especially in middle- and high-dosage groups. No significant changes were observed in SOD and CAT activity or mRNA expression. GSH-Px activity and GSH mRNA expression were significantly higher in the low-dosage group than in the model group. Compared to the model group, GR activity was significantly lower in the middle and high dosage groups, while the mRNA expression was higher. The PKC-beta level increased, while p-ERK1/2, NF-κB, and the ratio of p-ERK1/2*(ERK1/2)-1 decreased significantly in the treatment groups. Therefore, YSHX can alleviate GTW-induced testicular damage, enhance GSH-Px activity, regulate GSH redox cycling, and mitigate oxidative stress injury. Furthermore, YSHX can promote PKC-beta expression and inhibit the phosphorylation of ERK1/2 and NF-κB. Using YSHX may be an effective way to increase sperm motility via the PKC-ERK1/2-NF-ĸB axis.


Assuntos
Antioxidantes , Astenozoospermia , Medicamentos de Ervas Chinesas , Estresse Oxidativo , Ratos Sprague-Dawley , Animais , Masculino , Antioxidantes/farmacologia , Medicamentos de Ervas Chinesas/farmacologia , Medicamentos de Ervas Chinesas/uso terapêutico , Astenozoospermia/tratamento farmacológico , Astenozoospermia/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Ratos , NF-kappa B/metabolismo , Testículo/efeitos dos fármacos , Testículo/metabolismo , Tripterygium/química , Malondialdeído/metabolismo , Superóxido Dismutase/metabolismo
12.
Front Immunol ; 15: 1361606, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38846937

RESUMO

Introduction: Pathological changes in the articular cartilage (AC) and synovium are major manifestations of osteoarthritis (OA) and are strongly associated with pain and functional limitations. Exosome-derived microRNAs (miRNAs) are crucial regulatory factors in intercellular communication and can influence the progression of OA by participating in the degradation of chondrocytes and the phenotypic transformation in the polarization of synovial macrophages. However, the specific relationships and pathways of action of exosomal miRNAs in the pathological progression of OA in both cartilage and synovium remain unclear. Methods: This study evaluates the effects of fibroblast-like synoviocyte (FLS)-derived exosomes (FLS-Exos), influenced by miR-146a, on AC degradation and synovial macrophage polarization. We investigated the targeted relationship between miR-146a and TRAF6, both in vivo and in vitro, along with the involvement of the NF-κB signaling pathway. Results: The expression of miR-146a in the synovial exosomes of OA rats was significantly higher than in healthy rats. In vitro, the upregulation of miR-146a reduced chondrocyte apoptosis, whereas its downregulation had the opposite effect. In vivo, exosomes derived from miR-146a-overexpressing FLSs (miR-146a-FLS-Exos) reduced AC injury and chondrocyte apoptosis in OA. Furthermore, synovial proliferation was reduced, and the polarization of synovial macrophages shifted from M1 to M2. Mechanistically, the expression of TRAF6 was inhibited by targeting miR-146a, thereby modulating the Toll-like receptor 4/TRAF6/NF-κB pathway in the innate immune response. Discussion: These findings suggest that miR-146a, mediated through FLS-Exos, may alleviate OA progression by modulating cartilage degradation and macrophage polarization, implicating the NF-κB pathway in the innate immune response. These insights highlight the therapeutic potential of miR-146a as a protective agent in OA, underscoring the importance of exosomal miRNAs in the pathogenesis and potential treatment of the disease.


Assuntos
Exossomos , Macrófagos , MicroRNAs , Osteoartrite , Sinoviócitos , Fator 6 Associado a Receptor de TNF , MicroRNAs/genética , Animais , Exossomos/metabolismo , Osteoartrite/metabolismo , Osteoartrite/patologia , Osteoartrite/imunologia , Ratos , Macrófagos/imunologia , Macrófagos/metabolismo , Sinoviócitos/metabolismo , Sinoviócitos/patologia , Masculino , Fator 6 Associado a Receptor de TNF/metabolismo , Fator 6 Associado a Receptor de TNF/genética , Cartilagem Articular/metabolismo , Cartilagem Articular/patologia , Condrócitos/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais , Ratos Sprague-Dawley , Fibroblastos/metabolismo , Membrana Sinovial/metabolismo , Membrana Sinovial/patologia , Membrana Sinovial/imunologia , Células Cultivadas , Apoptose , Receptor 4 Toll-Like/metabolismo , Receptor 4 Toll-Like/genética , Ativação de Macrófagos
13.
Front Immunol ; 15: 1352404, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38846950

RESUMO

Background: CD2v, a critical outer envelope glycoprotein of the African swine fever virus (ASFV), plays a central role in the hemadsorption phenomenon during ASFV infection and is recognized as an essential immunoprotective protein. Monoclonal antibodies (mAbs) targeting CD2v have demonstrated promise in both diagnosing and combating African swine fever (ASF). The objective of this study was to develop specific monoclonal antibodies against CD2v. Methods: In this investigation, Recombinant CD2v was expressed in eukaryotic cells, and murine mAbs were generated through meticulous screening and hybridoma cloning. Various techniques, including indirect enzyme-linked immunosorbent assay (ELISA), western blotting, immunofluorescence assay (IFA), and bio-layer interferometry (BLI), were employed to characterize the mAbs. Epitope mapping was conducted using truncation mutants and epitope peptide mapping. Results: An optimal antibody pair for a highly sensitive sandwich ELISA was identified, and the antigenic structures recognized by the mAbs were elucidated. Two linear epitopes highly conserved in ASFV genotype II strains, particularly in Chinese endemic strains, were identified, along with a unique glycosylated epitope. Three mAbs, 2B25, 3G25, and 8G1, effectively blocked CD2v-induced NF-κB activation. Conclusions: This study provides valuable insights into the antigenic structure of ASFV CD2v. The mAbs obtained in this study hold great potential for use in the development of ASF diagnostic strategies, and the identified epitopes may contribute to vaccine development against ASFV.


Assuntos
Vírus da Febre Suína Africana , Febre Suína Africana , Anticorpos Monoclonais , Mapeamento de Epitopos , NF-kappa B , Animais , Vírus da Febre Suína Africana/imunologia , NF-kappa B/metabolismo , NF-kappa B/imunologia , Suínos , Camundongos , Febre Suína Africana/imunologia , Febre Suína Africana/virologia , Anticorpos Monoclonais/imunologia , Proteínas do Envelope Viral/imunologia , Epitopos/imunologia , Anticorpos Antivirais/imunologia , Camundongos Endogâmicos BALB C
14.
J Environ Sci (China) ; 145: 1-12, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-38844310

RESUMO

The potential association between colorectal cancer (CRC) and environmental pollutants is worrisome. Previous studies have found that some perfluoroalkyl acids, including perfluorooctane sulfonate (PFOS), induced colorectal tumors in experimental animals and promoted the migration of and invasion by CRC cells in vitro, but the underlying mechanism is unclear. Here, we investigated the effects of PFOS on the proliferation and migration of CRC cells and the potential mechanisms involving activating the PI3K/Akt-NF-κB signal pathway and epithelial-mesenchymal transition (EMT). It was found that PFOS promoted the growth and migration of HCT116 cells at non-cytotoxic concentrations and increased the mRNA expression of the migration-related angiogenic cytokines vascular endothelial growth factor (VEGF) and interleukin-8 (IL-8). In a mechanistic investigation, the up-stream signal pathway PI3K/Akt-NF-κB was activated by PFOS, and the process was suppressed by LY294002 (PI3K/Akt inhibitor) and BAY11-7082 (NF-κB inhibitor) respectively, leading to less proliferation of HCT116 cells. Furthermore, matrix metalloproteinases (MMP) and EMT-related markers were up-regulated after PFOS exposure, and were also suppressed respectively by LY294002 and BAY11-7082. Moreover, the up-regulation of EMT markers was suppressed by a MMP inhibitor GM6001. Taken together, our results indicated that PFOS promotes colorectal cancer cell migration and proliferation by activating the PI3K/Akt-NF-κB signal pathway and epithelial-mesenchymal transition. This could be a potential toxicological mechanism of PFOS-induced malignant development of colorectal cancer.


Assuntos
Ácidos Alcanossulfônicos , Movimento Celular , Neoplasias Colorretais , Transição Epitelial-Mesenquimal , Fluorocarbonos , Fluorocarbonos/toxicidade , Ácidos Alcanossulfônicos/toxicidade , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Neoplasias Colorretais/patologia , Humanos , Movimento Celular/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Poluentes Ambientais/toxicidade , Células HCT116 , Proteínas Proto-Oncogênicas c-akt/metabolismo , NF-kappa B/metabolismo , Proliferação de Células/efeitos dos fármacos , Linhagem Celular Tumoral
15.
Drug Dev Res ; 85(4): e22219, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38845211

RESUMO

Sepsis is a life-threatening organ dysfunction that endangers patient lives and is caused by an imbalance in the host defense against infection. Sepsis continues to be a significant cause of morbidity and mortality in critically sick patients. Oxymatrine (OMT), a quinolizidine alkaloid derived from the traditional Chinese herb Sophora flavescens Aiton, has been shown to have anti-inflammatory effects on a number of inflammatory illnesses according to research. In this study, we aimed to evaluate the therapeutic effects of OMT on sepsis and explore the underlying mechanisms. We differentiated THP-1 cells into THP-1 macrophages and studied the anti-inflammatory mechanism of OMT in a lipopolysaccharide (LPS)-induced THP-1 macrophage sepsis model. Activation of the receptor for advanced glycation end products (RAGE), as well as NF-κB, was assessed by Western blot analysis and immunofluorescence staining. ELISA was used to measure the levels of inflammatory factors. We found that OMT significantly inhibited HMGB1-mediated RAGE/NF-κB activation and downstream inflammatory cytokine production in response to LPS stimulation. Finally, an in vivo experiment was performed on septic mice to further study the effect of OMT on injured organs. The animal experiments showed that OMT significantly inhibited HMGB1-mediated RAGE/NF-κB activation, protected against the inflammatory response and organ injury induced by CLP, and prolonged the survival rate of septic mice. Herein, we provide evidence that OMT exerts a significant therapeutic effect on sepsis by inhibiting the HMGB1/RAGE/NF-κB signaling pathway.


Assuntos
Alcaloides , Proteína HMGB1 , Inflamação , Lipopolissacarídeos , NF-kappa B , Quinolizinas , Receptor para Produtos Finais de Glicação Avançada , Sepse , Transdução de Sinais , Alcaloides/farmacologia , Alcaloides/uso terapêutico , Quinolizinas/farmacologia , Quinolizinas/uso terapêutico , Animais , Sepse/tratamento farmacológico , Sepse/complicações , Sepse/metabolismo , NF-kappa B/metabolismo , Proteína HMGB1/metabolismo , Proteína HMGB1/antagonistas & inibidores , Humanos , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Transdução de Sinais/efeitos dos fármacos , Camundongos , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Masculino , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Células THP-1 , Camundongos Endogâmicos C57BL , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Matrinas
16.
Sci Adv ; 10(23): eadl6083, 2024 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-38838151

RESUMO

Hepatocellular carcinoma (HCC) acquires an immunosuppressive microenvironment, leading to unbeneficial therapeutic outcomes. Hyaluronan-mediated motility receptor (HMMR) plays a crucial role in tumor progression. Here, we found that aberrant expression of HMMR could be a predictive biomarker for the immune suppressive microenvironment of HCC, but the mechanism remains unclear. We established an HMMR-/- liver cancer mouse model to elucidate the HMMR-mediated mechanism of the dysregulated "don't eat me" signal. HMMR knockout inhibited liver cancer growth and induced phagocytosis. HMMRhigh liver cancer cells escaped from phagocytosis via sustaining CD47 signaling. Patients with HMMRhighCD47high expression showed a worse prognosis than those with HMMRlowCD47low expression. HMMR formed a complex with FAK/SRC in the cytoplasm to activate NF-κB signaling, which could be independent of membrane interaction with CD44. Notably, targeting HMMR could enhance anti-PD-1 treatment efficiency by recruiting CD8+ T cells. Overall, our data revealed a regulatory mechanism of the "don't eat me" signal and knockdown of HMMR for enhancing anti-PD-1 treatment.


Assuntos
Antígeno CD47 , Carcinoma Hepatocelular , Receptores de Hialuronatos , Neoplasias Hepáticas , Fagócitos , Fagocitose , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/genética , Animais , Humanos , Camundongos , Receptores de Hialuronatos/metabolismo , Receptores de Hialuronatos/genética , Fagócitos/metabolismo , Fagócitos/imunologia , Antígeno CD47/metabolismo , Antígeno CD47/genética , Linhagem Celular Tumoral , Transdução de Sinais , Microambiente Tumoral/imunologia , Evasão da Resposta Imune , NF-kappa B/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Camundongos Knockout , Quinase 1 de Adesão Focal/metabolismo , Quinase 1 de Adesão Focal/genética , Evasão Tumoral
17.
Sci Rep ; 14(1): 12917, 2024 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-38839811

RESUMO

Allii Macrostemonis Bulbus (AMB) is a traditional Chinese medicine with medicinal and food homology. AMB has various biological activities, including anti-coagulation, lipid-lowering, anti-tumor, and antioxidant effects. Saponins from Allium macrostemonis Bulbus (SAMB), the predominant beneficial compounds, also exhibited lipid-lowering and anti-inflammatory properties. However, the effect of SAMB on atherosclerosis and the underlying mechanisms are still unclear. This study aimed to elucidate the pharmacological impact of SAMB on atherosclerosis. In apolipoprotein E deficiency (ApoE-/-) mice with high-fat diet feeding, oral SAMB administration significantly attenuated inflammation and atherosclerosis plaque formation. The in vitro experiments demonstrated that SAMB effectively suppressed oxidized-LDL-induced foam cell formation by down-regulating CD36 expression, thereby inhibiting lipid endocytosis in bone marrow-derived macrophages. Additionally, SAMB effectively blocked LPS-induced inflammatory response in bone marrow-derived macrophages potentially through modulating the NF-κB/NLRP3 pathway. In conclusion, SAMB exhibits a potential anti-atherosclerotic effect by inhibiting macrophage foam cell formation and inflammation. These findings provide novel insights into potential preventive and therapeutic strategies for the clinical management of atherosclerosis.


Assuntos
Aterosclerose , Células Espumosas , Inflamação , Saponinas , Animais , Células Espumosas/efeitos dos fármacos , Células Espumosas/metabolismo , Aterosclerose/tratamento farmacológico , Aterosclerose/patologia , Aterosclerose/metabolismo , Aterosclerose/prevenção & controle , Saponinas/farmacologia , Camundongos , Inflamação/tratamento farmacológico , Inflamação/patologia , Allium/química , Masculino , Apolipoproteínas E/deficiência , Dieta Hiperlipídica/efeitos adversos , NF-kappa B/metabolismo , Camundongos Endogâmicos C57BL , Lipoproteínas LDL/metabolismo
18.
Sci Rep ; 14(1): 12935, 2024 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-38839973

RESUMO

The inhibition of tumor necrosis factor (TNF)-α trimer formation renders it inactive for binding to its receptors, thus mitigating the vicious cycle of inflammation. We designed a peptide (PIYLGGVFQ) that simulates a sequence strand of human TNFα monomer using a series of in silico methods, such as active site finding (Acsite), protein-protein interaction (PPI), docking studies (GOLD and Flex-X) followed by molecular dynamics (MD) simulation studies. The MD studies confirmed the intermolecular interaction of the peptide with the TNFα. Fluorescence-activated cell sorting and fluorescence microscopy revealed that the peptide effectively inhibited the binding of TNF to the cell surface receptors. The cell culture assays showed that the peptide significantly inhibited the TNFα-mediated cell death. In addition, the nuclear translocation of the nuclear factor kappa B (NFκB) was significantly suppressed in the peptide-treated A549 cells, as observed in immunofluorescence and gel mobility-shift assays. Furthermore, the peptide protected against joint damage in the collagen-induced arthritis (CIA) mouse model, as revealed in the micro focal-CT scans. In conclusion, this TNFα antagonist would be helpful for the prevention and repair of inflammatory bone destruction and subsequent loss in the mouse model of CIA as well as human rheumatoid arthritis (RA) patients. This calls upon further clinical investigation to utilize its potential effect as an antiarthritic drug.


Assuntos
Peptídeos , Fator de Necrose Tumoral alfa , Humanos , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Animais , Camundongos , Peptídeos/farmacologia , Peptídeos/química , Artrite Experimental/tratamento farmacológico , Artrite Experimental/metabolismo , Artrite Experimental/patologia , Simulação de Acoplamento Molecular , Células A549 , Simulação de Dinâmica Molecular , NF-kappa B/metabolismo , NF-kappa B/antagonistas & inibidores , Masculino , Antirreumáticos/farmacologia , Antirreumáticos/química , Antirreumáticos/uso terapêutico , Ligação Proteica , Modelos Animais de Doenças
19.
Mol Med ; 30(1): 76, 2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38840067

RESUMO

BACKGROUND: Advanced glycation end product-modified low-density lipoprotein (AGE-LDL) is related to inflammation and the development of atherosclerosis. Additionally, it has been demonstrated that receptor for advanced glycation end products (RAGE) has a role in the condition known as calcific aortic valve disease (CAVD). Here, we hypothesized that the AGE-LDL/RAGE axis could also be involved in the pathophysiological mechanism of CAVD. METHODS: Human aortic valve interstitial cells (HAVICs) were stimulated with AGE-LDL following pre-treatment with or without interleukin 37 (IL-37). Low-density lipoprotein receptor deletion (Ldlr-/-) hamsters were randomly allocated to chow diet (CD) group and high carbohydrate and high fat diet (HCHFD) group. RESULTS: AGE-LDL levels were significantly elevated in patients with CAVD and in a hamster model of aortic valve calcification. Our in vitro data further demonstrated that AGE-LDL augmented the expression of intercellular cell adhesion molecule-1 (ICAM-1), interleukin-6 (IL-6) and alkaline phosphatase (ALP) in a dose-dependent manner through NF-κB activation, which was attenuated by nuclear factor kappa-B (NF-κB) inhibitor Bay11-7082. The expression of RAGE was augmented in calcified aortic valves, and knockdown of RAGE in HAVICs attenuated the AGE-LDL-induced inflammatory and osteogenic responses as well as NF-κB activation. IL-37 suppressed inflammatory and osteogenic responses and NF-κB activation in HAVICs. The vivo experiment also demonstrate that supplementation with IL-37 inhibited valvular inflammatory response and thereby suppressed valvular osteogenic activities. CONCLUSIONS: AGE-LDL promoted inflammatory responses and osteogenic differentiation through RAGE/NF-κB pathway in vitro and aortic valve lesions in vivo. IL-37 suppressed the AGE-LDL-induced inflammatory and osteogenic responses in vitro and attenuated aortic valve lesions in a hamster model of CAVD.


Assuntos
Estenose da Valva Aórtica , Valva Aórtica , Calcinose , Produtos Finais de Glicação Avançada , Lipoproteínas LDL , NF-kappa B , Osteogênese , Receptor para Produtos Finais de Glicação Avançada , Transdução de Sinais , Animais , Valva Aórtica/metabolismo , Valva Aórtica/patologia , Produtos Finais de Glicação Avançada/metabolismo , NF-kappa B/metabolismo , Humanos , Calcinose/metabolismo , Calcinose/patologia , Calcinose/genética , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Receptor para Produtos Finais de Glicação Avançada/genética , Estenose da Valva Aórtica/metabolismo , Estenose da Valva Aórtica/etiologia , Estenose da Valva Aórtica/patologia , Cricetinae , Osteogênese/efeitos dos fármacos , Masculino , Lipoproteínas LDL/metabolismo , Modelos Animais de Doenças , Feminino , Pessoa de Meia-Idade , Proteínas Glicadas
20.
Invest Ophthalmol Vis Sci ; 65(6): 2, 2024 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-38829670

RESUMO

Purpose: The purpose of this study was to investigate the involvement of the TLR4/NF-κB/NLRP3 signaling pathway and its underlying mechanism in diabetic dry eye. Methods: Two models of diabetic dry eye were established in high glucose-induced human corneal epithelial (HCE-T) cells and streptozotocin (STZ)-induced C57BL/6 mice, and the TLR4 inhibitor fosfenopril (FOS) was utilized to suppress the TLR4/NF-κB/NLRP3 signaling pathway. The expression changes in TLR4, NF-κB, NLRP3, and IL-1ß, and other factors were detected by Western blot and RT‒qPCR, the wound healing rate was evaluated by cell scratch assay, and the symptoms of diabetic mice were evaluated by corneal sodium fluorescein staining and tear secretion assay. Results: In the diabetic dry eye model, the transcript levels of TLR4, NF-κB, NLRP3, and IL-1ß were raised, and further application of FOS, a TLR4 inhibitor, downregulated the levels of these pathway factors. In addition, FOS was found to be effective in increasing the wound healing rate of high glucose-induced HCE-T cells, increasing tear production, and decreasing corneal fluorescence staining scores in diabetic mice, as measured by cell scratch assay, corneal sodium fluorescein staining assay, and tear production. Conclusions: The current study found that the TLR4/NF-κB/NLRP3 signaling pathway regulates diabetic dry eye in an in vitro and in vivo model, and that FOS reduces the signs of dry eye in diabetic mice, providing a new treatment option for diabetic dry eye.


Assuntos
Diabetes Mellitus Experimental , Síndromes do Olho Seco , Camundongos Endogâmicos C57BL , NF-kappa B , Proteína 3 que Contém Domínio de Pirina da Família NLR , Transdução de Sinais , Receptor 4 Toll-Like , Animais , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/antagonistas & inibidores , Receptor 4 Toll-Like/metabolismo , Receptor 4 Toll-Like/antagonistas & inibidores , Camundongos , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/metabolismo , NF-kappa B/metabolismo , NF-kappa B/antagonistas & inibidores , Síndromes do Olho Seco/tratamento farmacológico , Síndromes do Olho Seco/metabolismo , Epitélio Corneano/efeitos dos fármacos , Epitélio Corneano/metabolismo , Humanos , Masculino , Western Blotting , Modelos Animais de Doenças , Células Cultivadas , Lágrimas/metabolismo , Reação em Cadeia da Polimerase em Tempo Real
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA