Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters








Publication year range
1.
Toxicol Lett ; 396: 19-27, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38642674

ABSTRACT

Ricin toxin (RT) is highly cytotoxic and can release a considerable amount of pro-inflammatory factors due to depurination, causing excessive inflammation that may aggravate the harm to the body. Pyroptosis, a type of gasdermin-mediated cell death, is a contributor to the exacerbation of inflammation. Accumulating evidence indicate that pyroptosis plays a significant role in the pathogen infection and tissue injury, suggesting a potential correlation between pyroptosis and RT-induced inflammation. Here, we aim to demonstrate this correlation and explore its molecular mechanisms. Results showed that RT triggers mouse alveolar macrophage MH-S cells pyroptosis by activating caspase-3 and cleaving Gasgermin E (GSDME). In contrast, inhibition of caspase-3 with Z-DEVD-FMK (inhibitor of caspase-3) or knockdown of GSDME attenuates this process, suggesting the essential role of caspase-3/GSDME-mediated pyroptosis in contributing to RT-induced inflammation. Collectively, our study enhances our understanding of a novel mechanism of ricin cytotoxicity, which may emerge as a potential target in immunotherapy to control the RT-induced inflammation.


Subject(s)
Caspase 3 , Inflammation , Pyroptosis , Ricin , Pyroptosis/drug effects , Ricin/toxicity , Animals , Mice , Caspase 3/metabolism , Inflammation/chemically induced , Inflammation/metabolism , Cell Line , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/metabolism , Gasdermins
3.
Toxicol Lett ; 383: 177-191, 2023 Jul 01.
Article in English | MEDLINE | ID: mdl-37392970

ABSTRACT

γ-bungarotoxin (γ-BGT) is an RGD motif-containing protein, derived from the venom of Bungarus multicinctus, leading to acute death in mice. These RGD motif-containing proteins from snake venom belonging to the disintegrin family can interfere with vascular endothelial homeostasis by directly binding cell surface integrins. Targeting integrins that generate vascular endothelial dysfunction may contribute to γ-BGT poisoning, however, the underlying mechanisms have not been investigated in detail. In this study, the results showed that γ-BGT played a role in -promoting the permeability of the vascular endothelial barrier. Depending on its selective binding to integrin α5 in vascular endothelium (VE), γ-BGT initiated downstream events, including focal adhesion kinase dephosphorylation and cytoskeleton remodeling, resulting in the intercellular junction interruption. Those alternations facilitated paracellular permeability of VE and barrier dysfunction. Proteomics profiling identified that as a downstream effector of the integrin α5 / FAK signaling pathway cyclin D1 partially mediated the cellular structural changes and barrier dysfunction. Furthermore, VE-released plasminogen activator urokinase and platelet-derived growth factor D could serve as potential diagnostic biomarkers for γ-BGT-induced vascular endothelial dysfunction. Our results indicate the mechanisms through which γ-BGT as a novel disintegrin directly interacts with the VE, with consequences for barrier dysfunction.


Subject(s)
Bungarotoxins , Endothelium, Vascular , Integrin alpha5 , Snake Venoms , Animals , Mice , Bungarotoxins/toxicity , Disintegrins/pharmacology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Integrin alpha5/metabolism , Integrins/metabolism , Oligopeptides , Snake Venoms/toxicity
4.
Toxicol Lett ; 383: 152-161, 2023 Jul 01.
Article in English | MEDLINE | ID: mdl-37390852

ABSTRACT

DNA-encoded monoclonal antibodies (DMAbs) and in vivo expression of antibody therapeutics presents an innovative alternative to conventional delivery methods. Therefore, in order to prevent the lethal dose of ricin toxin (RT) and to avoid human anti-mouse antibody (HAMA) reaction, we developed the human neutralizing antibody 4-4E against RT and constructed DMAb-4-4E. The human neutralizing antibody 4-4E could neutralize RT in vitro and in vivo, while the mice in RT group all died. Using intramuscular electroporation (IM EP), antibodies were rapidly expressed in vivo within 7 days and were enriched in intestine and gastrocnemius muscle mostly. Besides, we found that DMAbs have shown a broad protective efficacy of RT poisoning prophylaxis. Driven by plasmids for IgG expression, mice were survived and the blood glucose level of mice in DMAb-IgG group returned to normal at 72 h post RT challenge, and the RT group died within 48 h. Furthermore, hindrance of protein disulfide isomerase (PDI) and accumulation of RT in endosomes were found in IgG-protected cells, revealing the possible mechanism of neutralization details. These data support the further study of RT-neutralizing monoclonal antibodies (mAbs) in the development.


Subject(s)
Foodborne Diseases , Plant Poisoning , Poisoning , Ricin , Toxins, Biological , Animals , Mice , Humans , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing , Ricin/toxicity , Immunoglobulin G , Mice, Inbred BALB C , Poisoning/prevention & control
5.
Front Cell Infect Microbiol ; 13: 1155293, 2023.
Article in English | MEDLINE | ID: mdl-37207187

ABSTRACT

Introduction: The constantly mutating SARS-CoV-2 has been infected an increasing number of people, hence the safe and efficacious treatment are urgently needed to combat the COVID-19 pandemic. Currently, neutralizing antibodies (Nabs), targeting the receptor-binding domain (RBD) of the SARS-CoV-2 spike protein are potentially effective therapeutics against COVID-19. As a new form of antibody, bispecific single chain antibodies (BscAbs) can be easily expressed in E. coli and exhibits broad-spectrum antiviral activity. Methods: In this study, we constructed two BscAbs 16-29, 16-3022 and three single chain variable fragments (scFv) S1-16, S2-29 and S3022 as a comparison to explore their antiviral activity against SARS-CoV-2. The affinity of the five antibodies was characterized by ELISA and SPR and the neutralizing activity of them was analyzed using pseudovirus or authentic virus neutralization assay. Bioinformatics and competitive ELISA methods were used to identify different epitopes on RBD. Results: Our results revealed the potent neutralizing activity of two BscAbs 16-29 and 16-3022 against SARS-CoV-2 original strain and Omicron variant infection. In addition, we also found that SARS-CoV RBD-targeted scFv S3022 could play a synergistic role with other SARS-CoV-2 RBD-targeted antibodies to enhance neutralizing activity in the form of a BscAb or in cocktail therapies. Discussion: This innovative approach offers a promising avenue for the development of subsequent antibody therapies against SARSCoV-2. Combining the advantages of cocktails and single-molecule strategies, BscAb therapy has the potential to be developed as an effective immunotherapeutic for clinical use to mitigate the ongoing pandemic.


Subject(s)
COVID-19 , Single-Chain Antibodies , Humans , SARS-CoV-2/genetics , Escherichia coli , Pandemics , Antibodies, Monoclonal , Antibodies, Neutralizing , Single-Chain Antibodies/genetics , Antibodies, Viral/therapeutic use , Antiviral Agents
6.
Front Pharmacol ; 12: 767900, 2021.
Article in English | MEDLINE | ID: mdl-34744746

ABSTRACT

Increasing studies have concentrated on investigating circular RNAs (circRNAs) as pivotal regulators in the progression of numerous diseases and biological processes and abundant evidence shows that circRNAs are participated in the regulation of innate immune responses. Several studies showed that Ricin Toxin (RT) could induce inflammatory injury. There was no research on the particular functions and underlying mechanisms of circRNAs in RT-induced inflammation. In this study, RNA sequencing performed on RT-treated and normal RAW264.7 macrophage cells was used to investigated the differentially expressed circRNAs. Based on the dataset, the expression of circEpc1 (mmu_circ_0,000,842) was identified higher in RT-treated cells. Moreover, gain-and-loss function assays showed that circEpc1 function as a promoter in RT-induced inflammation in vivo and in vitro. Mechanistically, circEpc1 acted as a miR-5114 sponge to relieve the suppressive effect of miR-5114 on its target NOD2 and thereby activating NF-κB and MAPK signaling pathways. Our results illuminated a link between RT-induced inflammation and the circEpc1 regulatory loop and provided novel insight into the functions of circRNA in innate immune, which may emerge as a potential target in immunotherapy to control the RT-induced inflammatory injury.

7.
Toxicon ; 203: 129-138, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34673083

ABSTRACT

Ricin toxin (RT) is a ribosome-inactivating protein derived from the beans of the castor oil plant. Our previous studies have reported that RT can induce the production of inflammatory cytokines and cause inflammatory injury in RAW264.7 cells. In order to explore the various biological processes that long noncoding RNA (lncRNA), circular RNA (circRNA) and micro RNA (miRNA) as endogenous non-coding RNAs (ceRNAs) may participate in the pro-inflammatory mechanism, RT (20 ng/mL) treated and normal RAW264.7 cells were firstly sequenced by RNA-seq. By comparing the differentially expressed genes, we obtained 10 hub genes and enriched the inflammatory-related signaling pathways. Based on our results, we concluded a lncRNA/circRNA-miRNA-mRNA network. Finally, we verified the key genes and pathways by qRT-PCR, WB and ELISA. From the experiment results, an opening MAPK signaling pathway in TNF signaling pathway via TNFR2 was found involved in RT-induced inflammation. This work provides a reference for searching for ceRNA targets or therapeutic drugs in RT-induced inflammatory injury in the future.


Subject(s)
MicroRNAs , RNA, Long Noncoding , Ricin , Animals , Gene Regulatory Networks , Inflammation/chemically induced , Mice , MicroRNAs/genetics , RAW 264.7 Cells , RNA, Circular , RNA, Long Noncoding/genetics , RNA, Messenger , Ricin/toxicity
8.
Toxicol Lett ; 347: 67-77, 2021 Sep 01.
Article in English | MEDLINE | ID: mdl-33865919

ABSTRACT

Ricin toxin (RT) is one of the most lethal toxins derived from the seed of castor beans. In addition to its main toxic mechanism of inhibiting the synthesis of cellular proteins, RT can induce the production of inflammatory cytokines. MicroRNAs (miRNAs) play a key role in regulating both innate and adaptive immunity. To elucidate the regulation of miRNAs in RT-induced inflammation injury, the RNA high-throughput sequencing (RNA-Seq) technology was used to analyze the expression profile of miRNAs and mRNAs in RT-treated RAW264.7 cells. Results showed that a total of 323 mRNAs and 19 miRNAs differentially expressed after RT treated. Meanwhile, 713 miRNA-mRNA interaction pairs were identified by bioinformatics analysis. KEGG (Kyoto Encyclopedia of Genes and Genomes) pathway analysis showed that those interaction pairs were mainly involved in JAK-STAT, T cell receptor, and MAPK signaling pathways. Moreover, we further predicted and determined the targeting relationship between miR-155-3p and GAB2 through TargetScan and dual-luciferase reporter assay. Mechanically, overexpression of miR-155-3p can reduce the secretion of TNF-α in RAW264.7 cells, revealing a possible mechanism of miR-155-3p regulating RT-induced inflammatory injury. This study provides a new perspective for clarifying the mechanism of RT-induced inflammatory injury and reveals the potential role of miRNAs in innate immune regulation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Inflammation Mediators/metabolism , Inflammation/chemically induced , Macrophages/drug effects , MicroRNAs/metabolism , RNA, Messenger/metabolism , Ricin/toxicity , Transcriptome , Adaptor Proteins, Signal Transducing/genetics , Animals , Gene Expression Profiling , Gene Expression Regulation , Gene Regulatory Networks , Inflammation/genetics , Inflammation/metabolism , Macrophages/metabolism , Mice , MicroRNAs/genetics , RAW 264.7 Cells , RNA, Messenger/genetics , Signal Transduction
9.
Front Pharmacol ; 11: 526129, 2020.
Article in English | MEDLINE | ID: mdl-33013378

ABSTRACT

Ricin toxin binding subunit B (RTB) is a galactose-binding lectin protein derived from the beans of the castor oil plant (Ricinus communis). Our previous studies have reported a direct immunomodulatory effect of recombinant RTB, which stimulates RAW264.7 cells to produce cytokines including TNF-α. However, the role of RTB in innate immune response and its specific mechanism have not been reported in detail. In this work, the results showed that RTB treatment of macrophages significantly increased TLR4 protein levels. RTB also activated TLR4 downstream events, including MyD88, IRAK, and TRAF6, resulting in macrophage activation and TNF-α production. This process is reflected in the increase of IκB phosphorylation. TLR4 knockdown macrophages treated with RTB exhibited greatly reduced IκB phosphorylation and TNF-α secretion. Moreover, treatment with MyD88 inhibitor also suppressed TNF-α production. The docking of RT and TLR4 was simulated by computer, and the contact residues were concentrated on RTB. Our results suggest that recombinant RTB can activate mouse macrophages to secrete TNF-α through activation of NF-κB via the TLR4 signaling pathways.

10.
Biochem Pharmacol ; 173: 113673, 2020 03.
Article in English | MEDLINE | ID: mdl-31629709

ABSTRACT

Gastric cancer is one of the most lethal cancers with unmet clinical treatment and low 5-year survival rate. Schisantherin A is a major compound derived from Fructusschisandrae while its anti-tumor role remains nearly unknown. Here, we reported that schisantherin A had an anti-proliferation effect on gastric cancer cell lines MKN45 and SGC-7901. Schisantherin A induced cell cycle arrest at G2/M phase and cell apoptosis, and inhibited cell migration in gastric cancer MKN45 and SGC7901 cells. Meanwhile, upregulation of cleaved caspase-9, cleaved caspase-3 and cleaved PARP were accompanied with the loss of mitochondrial membrane potential (MMP). Moreover, schisantherin A induced ROS-dependent JNK phosphorylation with higher ROS production. The JNK inhibitor and ROS scavenger NAC rescued the cell apoptosis and cycle inhibition elicited by schisantherin A. Furthermore, the expression level of antioxidant factor Nrf2 was suppressed by schisantherin A. These findings suggest that schisantherin A possesses an anti-tumor activity via activation of ROS/JNK with Nrf2 inhibition, indicating that schisantherin A is a promising chemotherapeutic candidate for gastric cancer.


Subject(s)
Apoptosis/drug effects , Cyclooctanes/pharmacology , Dioxoles/pharmacology , Lignans/pharmacology , MAP Kinase Signaling System/drug effects , Reactive Oxygen Species/metabolism , Stomach Neoplasms/metabolism , Caspases/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cyclooctanes/chemistry , Dioxoles/chemistry , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Lignans/chemistry , Membrane Potential, Mitochondrial/drug effects , Molecular Structure , Phosphorylation/drug effects , Plant Extracts/chemistry , Plant Extracts/pharmacology , Schisandra/chemistry , Stomach Neoplasms/pathology
11.
Biol Res ; 52(1): 57, 2019 Nov 25.
Article in English | MEDLINE | ID: mdl-31767027

ABSTRACT

BACKGROUND: Gastric cancer is a common malignant tumor with high morbidity and mortality worldwide, which seriously affects human health. Gramicidin is a short peptide antibiotic which could be used for treating infection induced by bacteria or fungi. However, the anti-cancer effect of gramicidin on gastric cancer cells and its underlying mechanism remains largely unknown. RESULTS: Gastric cancer cells SGC-7901, BGC-823 and normal gastric mucosal cells GES-1 were treated with different concentrations of gramicidin respectively. The results of CCK-8 experiment revealed cellular toxicity of gramicidin to cancer cells while cell colony formation assay showed that gramicidin significantly inhibited the proliferation of gastric cancer cells, but had little effect on normal gastric mucosal cells. In addition, the wound healing assay showed that gramicidin inhibited the migration of SGC-7901 cell. Meanwhile, apoptosis and cell cycle analysis revealed that gramicidin induced cell apoptosis with G2/M cell cycle inhibition. Furthermore, western blot analysis demonstrated that gramicidin down-regulated the expression of cyclinD1 and Bcl-2 as well as the FoxO1 phosphorylation. CONCLUSIONS: The current study illustrated the anti-tumor activity of gramicidin on gastric cancer cells, providing a possibility for gramicidin to be applied in clinical practice for the treatment of gastric cancer.


Subject(s)
Apoptosis/drug effects , Cell Cycle/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Gramicidin/pharmacology , Stomach Neoplasms/pathology , Cell Line, Tumor , Cyclin D1/drug effects , Cyclin D1/metabolism , Down-Regulation , Forkhead Box Protein O1/drug effects , Forkhead Box Protein O1/metabolism , Humans , Phosphorylation , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism
13.
J Pharmacol Sci ; 139(4): 377-384, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30928090

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a progressive and lethal lung disease characterized by inflammation, multifocal fibrotic lesions and excessive collagen deposition with limited therapies. As a major bioactive compound in garlic, S-allyl-l-cysteine (SAC) is a neuroprotective drug candidate to prevent cognitive decline, however, its anti-pulmonary fibrotic activity remains unknown. Here, we investigated whether SAC could attenuate bleomycin (BLM)-induced pulmonary fibrosis and inflammation in mice. Our results showed that SAC dose-dependently reduced the infiltration of inflammatory cells, pulmonary lesions and collagen deposition in BLM treated mice with downregulated mRNA expression levels of fibrotic genes including alpha smooth muscle actin (α-SMA), fibronectin, collagen I and collagen III as well as the protein level of α-SMA. In addition, SAC could also reduce the mRNA expression of inflammatory mediators such as TNF-α and iNOS. Furthermore, higher phosphorylation of AKT and NF-κB p65 in IPF patient samples and murine samples was verified by immunohistochemistry while SAC could decrease the phosphorylation level of AKT and NF-κB p65 in mice stimulated with BLM. These findings, for the first time, indicate that SAC might mediate AKT/NF-κB signaling pathway to inhibit BLM-induced pulmonary fibrosis and support the potential role of SAC as an anti-pulmonary fibrosis agent.


Subject(s)
Bleomycin/adverse effects , Cysteine/analogs & derivatives , NF-kappa B/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/genetics , Signal Transduction/drug effects , Actins/genetics , Actins/metabolism , Animals , Collagen/genetics , Collagen/metabolism , Cysteine/pharmacology , Cysteine/therapeutic use , Dose-Response Relationship, Drug , Fibronectins/genetics , Fibronectins/metabolism , Gene Expression/drug effects , Inflammation , Inflammation Mediators/metabolism , Mice , Mice, Inbred C57BL , Phosphorylation/drug effects , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , RNA, Messenger/metabolism
14.
Pathol Res Pract ; 215(4): 761-765, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30819583

ABSTRACT

OBJECTIVES: To figure out that if there is a consistency relationship of the BRAFV600E mutation in matched-lymph node metastasis and original papillary thyroid carcinoma (PTC) specimen for the same patient. METHODS: We collected the specimen of thyroids and matched-lymph node metastases of PTCs and tested the BRAFV600E mutation status with amplification refractory mutation system (ARMS) PCR. RESULTS: 20 patients with PTC and metastasis lymph node were hired. In this cohort, 16 (80%) patients had the same BRAF genetic mutation status in thyroid and metastasis, and the other 4 (20%) had an inconsistent situation. CONCLUSIONS: Within our cohort, the data suggested that wild-type BRAFV600E oncogene in thyroid primary tumor does not rule out its mutation in lymph node metastasis, and vice versa.


Subject(s)
Lymphatic Metastasis/genetics , Proto-Oncogene Proteins B-raf/genetics , Thyroid Cancer, Papillary/genetics , Thyroid Neoplasms/genetics , Adult , DNA Mutational Analysis , Female , Humans , Lymph Nodes/pathology , Lymphatic Metastasis/pathology , Male , Middle Aged , Mutation , Thyroid Cancer, Papillary/pathology , Thyroid Neoplasms/pathology , Young Adult
15.
Acta Pharmacol Sin ; 40(8): 1040-1048, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30644422

ABSTRACT

Staphylococcus aureus (S. aureus) infection leads to a severe inflammatory response and causes acute lung injury (ALI), eventually threatening human life. Therefore, it is of importance to find an agent to inhibit inflammation and reduce ALI. Here, we found that costunolide, a sesquiterpene lactone, displays anti-inflammatory effects and ameliorates heat-killed S. aureus (HKSA)-induced pneumonia. Costunolide treatment attenuated HKSA-induced murine ALI in which pulmonary neutrophil infiltration was inhibited, lung edema was decreased, and the production of pro-inflammatory cytokines was significantly reduced. In addition, costunolide dose-dependently inhibited the generation of IL-6, TNF-α, IL-1ß, and keratinocyte-derived cytokine (KC), as well as the expression of iNOS, in HKSA-induced macrophages. Furthermore, costunolide attenuated the phosphorylation of p38 MAPK and cAMP response element-binding protein (CREB). Collectively, our findings suggested that costunolide is a promising agent for alleviating bacterial-induced ALI via the inhibition of the MAPK signaling pathways.


Subject(s)
Acute Lung Injury/drug therapy , Anti-Inflammatory Agents/therapeutic use , Macrophage Activation/drug effects , Sesquiterpenes/therapeutic use , Acute Lung Injury/microbiology , Animals , Cytokines/metabolism , Lung/microbiology , Lung/pathology , MAP Kinase Signaling System/drug effects , Male , Mice, Inbred C57BL , Neutrophil Infiltration/drug effects , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Pneumonia, Staphylococcal/drug therapy , Pneumonia, Staphylococcal/microbiology , Pulmonary Edema/drug therapy , Pulmonary Edema/microbiology , Staphylococcus aureus
16.
Ann Vasc Surg ; 57: 220-228, 2019 May.
Article in English | MEDLINE | ID: mdl-30684626

ABSTRACT

BACKGROUND: Previous studies have shown that Ginkgo biloba extract (GBE) dietary diminished salt-related elevation of blood pressure and ameliorated ischemic diseases. However, whether GBE could improve vascular elasticity to protect mesenteric arterioles of old rats is still elusive. In this study, we aimed to investigate the effects of GBE on vascular elasticity of old rats and its possible underlying mechanism. METHODS: Morphological changes of mesenteric arterioles were observed using hematoxylin and eosin and Verhoeff-Van Gieson staining, and diameters of mesenteric arterioles under various pressure were detected after GBE administration. In addition, phosphorylation level of Akt and FoxO3a proteins from mesenteric arterioles were detected. RESULTS: The results implicated that GBE treatment narrowed endothelial cell gap and increased the curvature of inner elastic membrane with reduced middle layer collagen fiber. Meanwhile, compared with young rats, old rats appeared to have lower vascular elasticity while GBE treatment at 50, 100, and 200 mg/kg dosage through intragastric administration per day for 3 weeks could effectively improve the vascular elasticity under different pressures in a dose-dependent manner. Furthermore, phosphorylation level of Akt and FoxO3a was also reduced in GBE-treated rats. CONCLUSIONS: This is the first report to indicate that GBE might exert protective effect on mesenteric arterioles of old rats via improving vascular elasticity and Akt/FoxO3a signaling pathway might be involved in this action.


Subject(s)
Arterioles/drug effects , Cardiovascular Agents/pharmacology , Forkhead Box Protein O3/metabolism , Mesentery/blood supply , Plant Extracts/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Vascular Stiffness/drug effects , Age Factors , Animals , Arterioles/enzymology , Arterioles/pathology , Arterioles/physiopathology , Dose-Response Relationship, Drug , Elasticity , Ginkgo biloba , Male , Phosphorylation , Rats, Sprague-Dawley , Signal Transduction/drug effects
17.
Acta Pharmacol Sin ; 40(1): 64-74, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30013035

ABSTRACT

Isoalantolactone (IAL) is a sesquiterpene lactone extracted from roots of Inula helenium L and has shown anti-inflammatory effects. In this study we investigated the therapeutic effects of IAL on acute lung injury (ALI) and elucidated the mechanisms underlying its anti-inflammation potential in vitro and in vivo. Treatment with lipopolysaccharide (LPS, 100 ng/mL) drastically stimulated production of inflammatory mediators such as NO, TNF-α, IL-1ß, and IL-6 in mouse bone marrow-derived macrophages (BMDMs), which was dose-dependently suppressed by pretreatment with IAL (2.5, 5, 10, 20 µM). We further revealed that IAL suppressed LPS-induced NF-κB, ERK, and Akt activation. Moreover, the downregulation of non-degradable K63-linked polyubiquitination of TRAF6, an upstream transcription factor of NF-κB, contributed to the anti-inflammatory effects of IAL. ALI was induced in mice by intratracheal injection of LPS (5 mg/kg). Administration of IAL (20 mg/kg, i.p.) significantly suppressed pulmonary pathological changes, neutrophil infiltration, pulmonary permeability, and pro-inflammatory cytokine expression. Our results demonstrate that IAL is a potential therapeutic reagent against inflammation and ALI.


Subject(s)
Acute Lung Injury/drug therapy , Anti-Inflammatory Agents/therapeutic use , Inflammation/drug therapy , Sesquiterpenes/therapeutic use , TNF Receptor-Associated Factor 6/metabolism , Ubiquitination/drug effects , Acute Lung Injury/chemically induced , Acute Lung Injury/pathology , Animals , Cytokines/metabolism , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins , Lipopolysaccharides , Lung/pathology , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , NF-kappa B p50 Subunit/metabolism , Signal Transduction/drug effects
18.
Biol. Res ; 52: 57-57, 2019. ilus, graf
Article in English | LILACS | ID: biblio-1505777

ABSTRACT

BACKGROUND: Gastric cancer is a common malignant tumor with high morbidity and mortality worldwide, which seriously affects human health. Gramicidin is a short peptide antibiotic which could be used for treating infection induced by bacteria or fungi. However, the anti-cancer effect of gramicidin on gastric cancer cells and its underlying mechanism remains largely unknown. RESULTS: Gastric cancer cells SGC-7901, BGC-823 and normal gastric mucosal cells GES-1 were treated with different concentrations of gramicidin respectively. The results of CCK-8 experiment revealed cellular toxicity of gramicidin to cancer cells while cell colony formation assay showed that gramicidin significantly inhibited the proliferation of gastric cancer cells, but had little effect on normal gastric mucosal cells. In addition, the wound healing assay showed that gramicidin inhibited the migration of SGC-7901 cell. Meanwhile, apoptosis and cell cycle analysis revealed that gramicidin induced cell apoptosis with G2/M cell cycle inhibition. Furthermore, western blot analysis demonstrated that gramicidin down-regulated the expression of cyclinD1 and Bcl-2 as well as the FoxO1 phosphorylation. CONCLUSIONS: The current study illustrated the anti-tumor activity of gramicidin on gastric cancer cells, providing a possibility for gramicidin to be applied in clinical practice for the treatment of gastric cancer.


Subject(s)
Humans , Stomach Neoplasms/pathology , Cell Cycle/drug effects , Cell Movement/drug effects , Apoptosis/drug effects , Cell Proliferation/drug effects , Gramicidin/pharmacology , Phosphorylation , Down-Regulation , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Cyclin D1/drug effects , Cyclin D1/metabolism , Cell Line, Tumor , Forkhead Box Protein O1/drug effects , Forkhead Box Protein O1/metabolism
19.
Biochem Pharmacol ; 155: 198-206, 2018 09.
Article in English | MEDLINE | ID: mdl-29991449

ABSTRACT

Asthma is one of the most common pulmonary diseases that threatens human life because of lack of effective medicines. Protostemonine (PSN), an active alkaloid extracted from the roots of Stemona sesslifolia, has anti-inflammatory effects on acute lung injury and acute liver failure. However, it has not been defined whether PSN alleviates asthmatic inflammation. Here, we reported that PSN inhibits pulmonary eosinophil infiltration, goblet cell hyperplasia, mucus secretion, IgE and Th2 cytokine (IL-4, IL-5, IL-13 and IL-33) production by using DRA (dust mites, ragweed and aspergillus)-induced murine asthma model. Moreover, PSN also attenuated the expression of Arginase-1 (Arg-1), Ym-1 and Fizz-1, markers of AAM (alternatively activated macrophage) polarization, in lung tissues. In addition, PSN attenuated IL-4-induced expression of Arg-1, Ym-1 and Fizz-1 in bone marrow derived macrophages (BMDMs). Treatment with PSN decreased IL-4-induced STAT6 phosphorylation, KLF4 and IRF4 expression in BMDMs. Collectively, our results indicated that PSN ameliorates AAM polarization and asthmatic inflammation and might be a potential agent for treating asthma.


Subject(s)
Ambrosia/adverse effects , Aspergillus fumigatus , Asthma/drug therapy , Macrophages/drug effects , Plant Extracts/therapeutic use , Pyroglyphidae , Animals , Asthma/chemically induced , Asthma/metabolism , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Kruppel-Like Factor 4 , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Plant Extracts/isolation & purification , Plant Extracts/pharmacology , Random Allocation , Stemonaceae
20.
Biochem Pharmacol ; 154: 183-192, 2018 08.
Article in English | MEDLINE | ID: mdl-29746822

ABSTRACT

Sepsis caused by Gram-negative bacteria is one of major causes for the progression of acute lung injury (ALI) with limited treatment and effective medicines. Tabersonine is an indole alkaloid mainly isolated from Catharanthus roseus, and a potential drug candidate for treatment of cancer and Alzheimer's disease (AD), however, its anti-inflammatory effect has not been revealed. In this study, we reported that tabersonine ameliorated lipopolysaccharides (LPS)-induced ALI in vivo and inhibited LPS-mediated macrophage activation in vitro. By using murine ALI model, we found that tabersonine significantly attenuated LPS-induced pathological injury in the lung. Tabersonine also inhibited LPS-mediated neutrophil infiltration, elevation of MPO activity and the production of TNF-α, IL-6 and IL-1ß. Furthermore, tabersonine inhibited LPS-induced the production of pro-inflammatory mediators such as iNOS, NO and cytokines by suppressing NF-κB and p38 MAPK/MK2 signaling cascades. Tabersonine reduced the K63-linked polyubiquitination of TRAF6. Taken together, these results suggested that tabersonine has anti-inflammatory activities in vitro and in vivo, and is a potential therapeutic candidate for the treatment of ALI/ARDS.


Subject(s)
Acute Lung Injury/prevention & control , Anti-Inflammatory Agents/therapeutic use , Indole Alkaloids/therapeutic use , Lipopolysaccharides/toxicity , Quinolines/therapeutic use , TNF Receptor-Associated Factor 6/antagonists & inhibitors , Ubiquitination/drug effects , Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Dose-Response Relationship, Drug , Indole Alkaloids/pharmacology , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Male , Mice , Mice, Inbred C57BL , Quinolines/pharmacology , Random Allocation , TNF Receptor-Associated Factor 6/metabolism , Ubiquitination/physiology
SELECTION OF CITATIONS
SEARCH DETAIL