Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 639
Filter
1.
Drug Dev Res ; 85(5): e22228, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38952003

ABSTRACT

Chromone-based compounds have established cytotoxic, antiproliferative, antimetastatic, and antiangiogenic effects on various cancer cell types via modulating different molecular targets. Herein, 17 novel chromone-2-carboxamide derivatives were synthesized and evaluated for their in vitro anticancer activity against 15 human cancer cell lines. Among the tested cell lines, MDA-MB-231, the triple-negative breast cancer cell line, was found to be the most sensitive, where the N-(2-furylmethylene) (15) and the α-methylated N-benzyl (17) derivatives demonstrated the highest growth inhibition with GI50 values of 14.8 and 17.1 µM, respectively. In vitro mechanistic studies confirmed the significant roles of compounds 15 and 17 in the induction of apoptosis and suppression of EGFR, FGFR3, and VEGF protein levels in MDA-MB-231 cancer cells. Moreover, compound 15 exerted cell cycle arrest at both the G0-G1 and G2-M phases. The in vivo efficacy of compound 15 as an antitumor agent was further investigated in female mice bearing Solid Ehrlich Carcinoma. Notably, administration of compound 15 resulted in a marked decrease in both tumor weight and volume, accompanied by improvements in biochemical, hematological, histological, and immunohistochemical parameters that verified the repression of both angiogenesis and inflammation as additional Anticancer mechanisms. Moreover, the binding interactions of compounds 15 and 17 within the binding sites of all three target receptors (EGFR, FGFR3, and VEGF) were clearly illustrated using molecular docking.


Subject(s)
Antineoplastic Agents , Chromones , ErbB Receptors , Molecular Docking Simulation , Receptor, Fibroblast Growth Factor, Type 3 , Triple Negative Breast Neoplasms , Vascular Endothelial Growth Factor A , Humans , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Female , Animals , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Vascular Endothelial Growth Factor A/metabolism , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Receptor, Fibroblast Growth Factor, Type 3/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Mice , Chromones/pharmacology , Chromones/chemical synthesis , Chromones/chemistry , Chromones/therapeutic use , Drug Design , Apoptosis/drug effects , Cell Proliferation/drug effects
2.
Cell Biochem Biophys ; 82(3): 2275-2283, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38839699

ABSTRACT

Iguratimod (T-614) is a compound widely used as anti-rheumatic drug. This study investigated the effect and underlying mechanism of T-614 on experimental Sjögren's syndrome (ESS). ESS mice model was established by injection of submandibular gland protein. Mice were randomly divided into control, experimental Sjögren's syndrome (ESS), ESS + T-614 (10 mg/kg), ESS + T-614 (20 mg/kg), and ESS + T-614 (30 mg/kg) groups. Human submandibular gland (HSG) were cultured with 0, 0.5, 5, or 50 µg/ml T-614 in the absence or presence of interferon-α (IFN-α). Haematoxylin and eosin (H&E) and cytokine levels were used to detect immune cells activation in submandibular glands. Apoptosis in submandibular glands tissues and cells was determined by TUNEL and flow cytometry. Apoptosis and NLRP3 inflammasome-related proteins were detected by western blotting. T-614 treatment attenuated submandibular gland damage in ESS mice. T-614 administration inhibited submandibular gland cell apoptosis in ESS mice. Furthermore, T-614 blocked inflammatory factor levels and NLRP3 inflammasome activation in the submandibular glands. In vitro, results corroborated that T-614 could protect HSG cells from IFN-α-induced cell apoptosis and inflammation by inhibiting NLRP3 inflammasome activation. Our results expounded that T-614 alleviated ESS by inhibiting NLRP3 inflammasome activation.


Subject(s)
Apoptosis , Disease Models, Animal , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Sjogren's Syndrome , Submandibular Gland , Sulfonamides , Sjogren's Syndrome/drug therapy , Sjogren's Syndrome/metabolism , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Mice , Humans , Submandibular Gland/drug effects , Submandibular Gland/metabolism , Submandibular Gland/pathology , Inflammasomes/metabolism , Apoptosis/drug effects , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Chromones/pharmacology , Chromones/therapeutic use , Female , Cytokines/metabolism , Mice, Inbred C57BL
3.
J Microbiol Immunol Infect ; 57(4): 647-659, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38839542

ABSTRACT

BACKGROUND: Co-therapy with albendazole and steroid is commonly used in patients with eosinophilic meningoencephalitis caused by Angiostrongylus cantonensis infections. However, anthelminthics often worsen symptoms, possibly due to the inflammatory reaction to antigens released by dying worms. Therefore, the present study was to investigate the curative effects and probable mechanisms of the platelet-derived growth factor receptor-beta (PDGFR-ß) inhibitor AG1296 (AG) and the phosphoinositide 3-kinase inhibitor (PI3K) LY294002 (LY) in A. cantonensis-induced neurovascular unit dysfunction and eosinophilic meningoencephalitis. METHODS: Western blots were used to detect matrix protein degradation and the expressions of PDGFR-ß/PI3K signaling pathway. The co-localization of PDGFR-ß and vascular smooth muscle cells (VSMCs), and metalloproteinase-9 (MMP-9) and VSMCs on the blood vessels were measured by confocal laser scanning immunofluorescence microscopy. Sandwich enzyme-linked immunosorbent assays were used to test S100B, interleukin (IL)-6, and transforming growth factor beta in the cerebrospinal fluid to determine their possible roles in mouse resistance to A. cantonensis. RESULTS: The results showed that AG and LY cotherapy decreased the MMP-9 activity and inflammatory reaction. Furthermore, S100B, IL-6 and eosinophil counts were reduced by inhibitor treatment. The localization of PDGFR-ß and MMP-9 was observed in VSMCs. Furthermore, we showed that the degradation of the neurovascular matrix and blood-brain barrier permeability were reduced in the mouse brain. CONCLUSIONS: These findings demonstrate the potential of PDGFR-ß inhibitor AG and PI3K inhibitor LY co-therapy as anti-A. cantonensis drug candidates through improved neurovascular unit dysfunction and reduced inflammatory response.


Subject(s)
Angiostrongylus cantonensis , Chromones , Meningoencephalitis , Morpholines , Strongylida Infections , Animals , Angiostrongylus cantonensis/drug effects , Meningoencephalitis/drug therapy , Meningoencephalitis/parasitology , Strongylida Infections/drug therapy , Mice , Chromones/pharmacology , Chromones/therapeutic use , Morpholines/pharmacology , Morpholines/therapeutic use , Signal Transduction/drug effects , Male , Receptor, Platelet-Derived Growth Factor beta/metabolism , Blood-Brain Barrier/drug effects , Disease Models, Animal , Phosphatidylinositol 3-Kinases/metabolism , Anthelmintics/therapeutic use , Anthelmintics/pharmacology , Matrix Metalloproteinase 9/metabolism , Eosinophilia/drug therapy , Drug Therapy, Combination , Sulfonamides
4.
Acta Pharmacol Sin ; 45(9): 1926-1936, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38744938

ABSTRACT

Primary Sjögren's syndrome (pSS) is a chronic inflammatory autoimmune disease with an unclear pathogenesis, and there is currently no approved drug for the treatment of this disease. Iguratimod, as a novel clinical anti-rheumatic drug in China and Japan, has shown remarkable efficacy in improving the symptoms of patients with pSS in clinical studies. In this study we investigated the mechanisms underlying the therapeutic effect of iguratimod in the treatment of pSS. Experimental Sjögren's syndrome (ESS) model was established in female mice by immunizing with salivary gland protein. After immunization, ESS mice were orally treated with iguratimod (10, 30, 100 mg·kg-1·d-1) or hydroxychloroquine (50 mg·kg-1·d-1) for 70 days. We showed that iguratimod administration dose-dependently increased saliva secretion, and ameliorated ESS development by predominantly inhibiting B cells activation and plasma cell differentiation. Iguratimod (30 and 100 mg·kg-1·d-1) was more effective than hydroxychloroquine (50 mg·kg-1·d-1). When the potential target of iguratimod was searched, we found that iguratimod bound to TEC kinase and promoted its degradation through the autophagy-lysosome pathway in BAFF-activated B cells, thereby directly inhibiting TEC-regulated B cells function, suggesting that the action mode of iguratimod on TEC was different from that of conventional kinase inhibitors. In addition, we found a crucial role of TEC overexpression in plasma cells of patients with pSS. Together, we demonstrate that iguratimod effectively ameliorates ESS via its unique suppression of TEC function, which will be helpful for its clinical application. Targeting TEC kinase, a new regulatory factor for B cells, may be a promising therapeutic option.


Subject(s)
Cell Differentiation , Chromones , Plasma Cells , Protein-Tyrosine Kinases , Sjogren's Syndrome , Sulfonamides , Animals , Sjogren's Syndrome/drug therapy , Female , Cell Differentiation/drug effects , Mice , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Plasma Cells/drug effects , Chromones/pharmacology , Chromones/therapeutic use , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Benzofurans/pharmacology , Benzofurans/therapeutic use , Hydroxychloroquine/pharmacology , Hydroxychloroquine/therapeutic use , Disease Models, Animal , Humans , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Antirheumatic Agents/pharmacology , Antirheumatic Agents/therapeutic use
5.
Front Immunol ; 15: 1347045, 2024.
Article in English | MEDLINE | ID: mdl-38756781

ABSTRACT

It is essential to understand the interactions and relationships between Mycobacterium tuberculosis (Mtb) and macrophages during the infection in order to design host-directed, immunomodulation-dependent therapeutics to control Mtb. We had reported previously that ornithine acetyltransferase (MtArgJ), a crucial enzyme of the arginine biosynthesis pathway of Mtb, is allosterically inhibited by pranlukast (PRK), which significantly reduces bacterial growth. The present investigation is centered on the immunomodulation in the host by PRK particularly the activation of the host's immune response to counteract bacterial survival and pathogenicity. Here, we show that PRK decreased the bacterial burden in the lungs by upregulating the population of pro-inflammatory interstitial macrophages (IMs) and reducing the population of Mtb susceptible alveolar macrophages (AMs), dendritic cells (DCs), and monocytes (MO). Additionally, we deduce that PRK causes the host macrophages to change their metabolic pathway from fatty acid metabolism to glycolytic metabolism around the log phage of bacterial multiplication. Further, we report that PRK reduced tissue injury by downregulating the Ly6C-positive population of monocytes. Interestingly, PRK treatment improved tissue repair and inflammation resolution by increasing the populations of arginase 1 (Arg-1) and Ym1+Ym2 (chitinase 3-like 3) positive macrophages. In summary, our study found that PRK is useful not only for reducing the tubercular burden but also for promoting the healing of the diseased tissue.


Subject(s)
Chromones , Disease Models, Animal , Mycobacterium tuberculosis , Animals , Mycobacterium tuberculosis/immunology , Mice , Chromones/pharmacology , Chromones/therapeutic use , Antitubercular Agents/therapeutic use , Antitubercular Agents/pharmacology , Tuberculosis/immunology , Tuberculosis/microbiology , Tuberculosis/drug therapy , Macrophages/immunology , Macrophages/microbiology , Macrophages/metabolism , Mice, Inbred C57BL , Female , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/microbiology , Tuberculosis, Pulmonary/drug therapy , Lung/microbiology , Lung/immunology , Lung/pathology
6.
Ren Fail ; 46(1): 2327498, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38666363

ABSTRACT

Iguratimod is a novel synthetic, small-molecule immunosuppressive agent used to treat rheumatoid arthritis. Through ongoing exploration of its role and mechanisms of action, iguratimod has been observed to have antifibrotic effects in the lung and skin; however, its effect on renal fibrosis remains unknown. This study aimed to investigate whether iguratimod could affect renal fibrosis progression. Three different concentrations of iguratimod (30 mg/kg/day, 10 mg/kg/day, and 3 mg/kg/day) were used to intervene in unilateral ureteral obstruction (UUO) model mice. Iguratimod at 10 mg/kg/day was observed to be effective in slowing UUO-mediated renal fibrosis. In addition, stimulating bone marrow-derived macrophages with IL-4 and/or iguratimod, or with TGF-ß and iguratimod or SRC inhibitors in vitro, suggested that iguratimod mitigates the progression of renal fibrosis in UUO mice, at least in part, by inhibiting the IL-4/STAT6 signaling pathway to attenuate renal M2 macrophage infiltration, as well as by impeding SRC activation to reduce macrophage-myofibroblast transition. These findings reveal the potential of iguratimod as a treatment for renal disease.


Subject(s)
Disease Models, Animal , Fibrosis , Interleukin-4 , Macrophages , STAT6 Transcription Factor , Sulfonamides , Ureteral Obstruction , Animals , Ureteral Obstruction/complications , Mice , Macrophages/drug effects , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Interleukin-4/metabolism , STAT6 Transcription Factor/metabolism , Male , Myofibroblasts/drug effects , Chromones/pharmacology , Chromones/therapeutic use , Kidney/pathology , Signal Transduction/drug effects , Transforming Growth Factor beta/metabolism , Kidney Diseases/etiology , Kidney Diseases/prevention & control , Kidney Diseases/pathology , Kidney Diseases/drug therapy , Mice, Inbred C57BL , Immunosuppressive Agents/pharmacology
7.
Curr Top Med Chem ; 23(9): 713-735, 2023.
Article in English | MEDLINE | ID: mdl-36786146

ABSTRACT

Tropical, vector-borne, and neglected diseases with a limited number of medication therapies include Leishmaniasis, Malaria, Chagas and Human African Trypanosomiasis (HAT). Chromones are a large class of heterocyclic compounds with significant applications. This heterocycle has long aroused the interest of scientists and the general public from biosynthetic and synthetic points of view owing to its interesting pharmacological activities. Chromones and their hybrids and isomeric forms proved to be an exciting scaffold to investigate these diseases. The in vitro activities of Chromone, Chromane, and a panel of other related benzopyran class compounds against Trypanosoma brucei rhodesiense, Trypanosoma brucei gambiense, Trypanosoma cruzi, and numerous Leishmanial and Malarial species were investigated in our previous studies. The current article briefly describes the neglected diseases and the current treatment. This review aims to attempt to find better alternatives by scrutinizing natural and synthetic derivatives for which chromones and their analogues were discovered to be a new and highly effective scaffold for the treatment of neglected diseases, including compounds with dual activity or activity against multiple parasites. Additionally, the efficacy of other new scaffolds was also thoroughly examined. This article also discusses prospects for identifying more unique targets for the disease, focusing on flavonoids as drug molecules that are less cytotoxic and high antiprotozoal potential. It also emphasizes the changes that can be made while searching for potential therapies-comparing existing treatments against protozoal diseases and the advantages of the newer chromone analogues over them. Finally, the structure- activity relationship at each atom of the chromone has also been highlighted.


Subject(s)
Antiprotozoal Agents , Malaria , Trypanosomiasis, African , Animals , Humans , Neglected Diseases/drug therapy , Retrospective Studies , Trypanosomiasis, African/drug therapy , Antiprotozoal Agents/pharmacology , Antiprotozoal Agents/therapeutic use , Malaria/drug therapy , Chromones/pharmacology , Chromones/therapeutic use
8.
Funct Integr Genomics ; 23(1): 35, 2023 Jan 11.
Article in English | MEDLINE | ID: mdl-36629976

ABSTRACT

Rohitukine is a chromone alkaloid and precursor of potent anticancer drugs flavopiridol, P-276-00, and 2,6-dichloro-styryl derivative (11d) (IIIM-290). The metabolite is reported to possess anticancer, anti-inflammatory, antiadipogenic, immunomodulatory, gastroprotective, anti-implantation, antidyslipidemic, anti-arthritic, and anti-fertility properties. However, the physiological role of rohitukine in plant system is yet to be explored. Here, we studied the effect of rohitukine isolated from Dysoxylum gotadhora on Arabidopsis thaliana. The A. thaliana plants grown on a medium fortified with different rohitukine concentrations showed a significant effect on the growth and development. The root growth of A. thaliana seedlings showed considerable inhibition when grown on medium containing 1.0 mM of rohitukine. Transcriptomic analysis indicated the expression of 895 and 932 genes in control and treated samples respectively at a cut-off of FPKM ≥ 1 and P-value < 0.05. Gene ontology (GO) analysis revealed the upregulation of genes related to photosynthesis, membrane transport, antioxidation, xenobiotic degradation, and some transcription factors (TFs) in response to rohitukine. Conversely, rohitukine downregulated several genes including RNA helicases and those involved in nitrogen compound metabolism. The RNA-seq result was also validated by real-time qRT-PCR analysis. In light of these results, we discuss (i) likely ecological importance of rohitukine in parent plant as well as (ii) comparison between responses to rohitukine treatment in plants and mammals.


Subject(s)
Alkaloids , Antineoplastic Agents , Arabidopsis , Animals , Arabidopsis/genetics , Antineoplastic Agents/pharmacology , Chromones/pharmacology , Chromones/therapeutic use , Alkaloids/pharmacology , Gene Expression Profiling , Transcriptome , Gene Expression Regulation, Plant , Mammals
9.
ACS Chem Neurosci ; 13(23): 3488-3501, 2022 12 07.
Article in English | MEDLINE | ID: mdl-36383455

ABSTRACT

Based on a multitarget strategy, a series of novel chromanone-1-benzyl-1,2,3,6-tetrahydropyridin hybrids were identified for the potential treatment of Alzheimer's disease (AD). Biological evaluation demonstrated that these hybrids exhibited significant inhibitory activities toward acetylcholinesterase (AChE) and monoamine oxidase B (MAO-B). The optimal compound C10 possessed excellent dual AChE/MAO-B inhibition both in terms of potency and equilibrium (AChE: IC50 = 0.58 ± 0.05 µM; MAO-B: IC50 = 0.41 ± 0.04 µM). Further molecular modeling and kinetic investigations revealed that compound C10 was a dual-binding inhibitor bound to both the catalytic anionic site and peripheral anionic site of AChE. In addition, compound C10 exhibited low neurotoxicity and potently inhibited AChE enzymatic activity. Furthermore, compound C10 more effectively protected against mitochondrial dysfunction and oxidation than donepezil, strongly inhibited AChE-induced amyloid aggregation, and moderately reduced glutaraldehyde-induced phosphorylation of tau protein in SH-SY5Y cells. Moreover, compound C10 displayed largely enhanced improvements in cognitive behaviors and spatial memory in a scopolamine-induced AD mice model with better efficacy than donepezil. Overall, the multifunctional profiles of compound C10 suggest that it deserves further investigation as a promising lead for the prospective treatment of AD.


Subject(s)
Alzheimer Disease , Cholinesterase Inhibitors , Chromones , Monoamine Oxidase Inhibitors , Animals , Humans , Mice , Acetylcholinesterase/metabolism , Alzheimer Disease/drug therapy , Cell Line, Tumor , Chromones/chemical synthesis , Chromones/pharmacology , Chromones/therapeutic use , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/pharmacology , Cholinesterase Inhibitors/therapeutic use , Monoamine Oxidase Inhibitors/chemical synthesis , Monoamine Oxidase Inhibitors/pharmacology , Monoamine Oxidase Inhibitors/therapeutic use , Drug Design
10.
Molecules ; 26(24)2021 Dec 16.
Article in English | MEDLINE | ID: mdl-34946728

ABSTRACT

Chromone glycosides comprise an important group of secondary metabolites. They are widely distributed in plants and, to a lesser extent, in fungi and bacteria. Significant biological activities, including antiviral, anti-inflammatory, antitumor, antimicrobial, etc., have been discovered for chromone glycosides, suggesting their potential as drug leads. This review compiles 192 naturally occurring chromone glycosides along with their sources, classification, biological activities, and spectroscopic features. Detailed biosynthetic pathways and chemotaxonomic studies are also described. Extensive spectroscopic features for this class of compounds have been thoroughly discussed, and detailed 13C-NMR data of compounds 1-192, have been added, except for those that have no reported 13C-NMR data.


Subject(s)
Anti-Infective Agents , Anti-Inflammatory Agents , Antineoplastic Agents , Chromones , Glycosides , Animals , Anti-Infective Agents/chemistry , Anti-Infective Agents/metabolism , Anti-Infective Agents/therapeutic use , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/metabolism , Anti-Inflammatory Agents/therapeutic use , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/therapeutic use , Chromones/chemistry , Chromones/metabolism , Chromones/therapeutic use , Glycosides/biosynthesis , Glycosides/chemistry , Glycosides/therapeutic use , Humans , Magnetic Resonance Spectroscopy , Molecular Structure
11.
Eur J Pharmacol ; 913: 174636, 2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34801529

ABSTRACT

PURPOSE: In the context of well-known inhibitory effects of Farrerol on the invasion of lung squamous cell carcinoma cells, the unexplored effect and regulatory mechanism of Farrerol on laryngeal squamous cell carcinoma (LSCC) emerged as the target in this study. METHODS: After treatment with Farrerol alone, or together with MitoTempo, the viability, apoptosis, cell cycle distribution, migration, and invasion of LSCC cells were measured using MTT, flow cytometry, wound-healing, and transwell assays, respectively. Meanwhile, the levels of cytochrome C (Cyt C), Cleaved caspase-3/9, Cyclin D1, E-cadherin, N-cadherin, and Vimentin in LSCC cells were evaluated by Western blot; the reactive oxygen species (ROS) formation intensity and the disruption of mitochondrial membrane potential (MMP) of LSCC cells were assessed using flow cytometry; and the effect of Farrerol on xenograft tumor formation was evaluated in animal experiment. RESULTS: Farrerol (10, 20, 50 µM) inhibited the viability, proliferation, cell cycle progression, migration and invasion, but promoted apoptosis, ROS formation intensity and disruption of MMP of LSCC cells. Moreover, Farrerol up-regulated Cyt C (in the cytoplasm), Cleaved caspase-3/9 and E-cadherin levels, but down-regulated Cyclin D1, N-cadherin and Vimentin levels in LSCC cells. Additionally, we uncovered that MitoTempo reversed the promoting effects of Farrerol on ROS formation intensity, apoptosis, and Cyt C and Cleaved caspase-3/9 levels in LSCC cells, while improving the disruption of MMP in Farrerol-treated LSCC cells. Also, Farrerol lessened the volume and weight of mice tumors. CONCLUSIONS: Farrerol suppressed the migration, invasion, and induced the apoptosis of LSCC cells via the mitochondria-mediated pathway.


Subject(s)
Chromones/pharmacology , Laryngeal Neoplasms/drug therapy , Mitochondria/drug effects , Squamous Cell Carcinoma of Head and Neck/drug therapy , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Chromones/therapeutic use , Gene Expression Regulation, Neoplastic/drug effects , Humans , Laryngeal Neoplasms/genetics , Laryngeal Neoplasms/pathology , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/metabolism , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/prevention & control , Organophosphorus Compounds/pharmacology , Piperidines/pharmacology , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/pathology , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
12.
Int J Mol Sci ; 22(20)2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34681783

ABSTRACT

Despite comprehensive therapy and extensive research, glioblastoma (GBM) still represents the most aggressive brain tumor in adults. Glioma stem cells (GSCs) are thought to play a major role in tumor progression and resistance of GBM cells to radiochemotherapy. The PIM1 kinase has become a focus in cancer research. We have previously demonstrated that PIM1 is involved in survival of GBM cells and in GBM growth in a mouse model. However, little is known about the importance of PIM1 in cancer stem cells. Here, we report on the role of PIM1 in GBM stem cell behavior and killing. PIM1 inhibition negatively regulates the protein expression of the stem cell markers CD133 and Nestin in GBM cells (LN-18, U-87 MG). In contrast, CD44 and the astrocytic differentiation marker GFAP were up-regulated. Furthermore, PIM1 expression was increased in neurospheres as a model of GBM stem-like cells. Treatment of neurospheres with PIM1 inhibitors (TCS PIM1-1, Quercetagetin, and LY294002) diminished the cell viability associated with reduced DNA synthesis rate, increased caspase 3 activity, decreased PCNA protein expression, and reduced neurosphere formation. Our results indicate that PIM1 affects the glioblastoma stem cell behavior, and its inhibition kills glioblastoma stem-like cells, pointing to PIM1 targeting as a potential anti-glioblastoma therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/pathology , Glioblastoma/pathology , Neoplastic Stem Cells/drug effects , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Survival/drug effects , Cell Survival/genetics , Chromones/pharmacology , Chromones/therapeutic use , Drug Screening Assays, Antitumor , Flavones/pharmacology , Flavones/therapeutic use , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Morpholines/pharmacology , Morpholines/therapeutic use , Neoplastic Stem Cells/pathology , Proto-Oncogene Proteins c-pim-1/genetics , Tumor Cells, Cultured
14.
Eur Rev Med Pharmacol Sci ; 25(14): 4687-4692, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34337716

ABSTRACT

OBJECTIVE: Iguratimod is a new kind of synthetic small molecule disease modified anti-rheumatic drug with good efficacy for rheumatoid arthritis (RA) treatment; meanwhile, it exhibits potency to alleviate alveolar inflammation and pulmonary fibrosis. However, its application in RA interstitial lung disease (ILD) patients is seldomly reported. Thus, the current study aimed to investigate the efficacy and safety of iguratimod plus glucocorticoid/cyclophosphamide vs. glucocorticoid/cyclophosphamide in treating RA-ILD patients. PATIENTS AND METHODS: Totally 101 RA-ILD patients underwent glucocorticoid/cyclophosphamide (Control group: n=61) or iguratimod plus glucocorticoid/cyclophosphamide (Iguratimod group: n=40) treatment were analyzed. General inflammation, disease activity, serum disease marker levels, high resolution lung computed tomography (HRCT) score, lung function indexes were evaluated within 24-week (W) treatment. RESULTS: No difference of baseline demographic or disease-related features was observed between Iguratimod group and Control group. Iguratimod group showed lower levels of CRP and ESR at W4, W12 and W24; as well as decreased DAS28 score, rheumatoid factor and anti-cyclic citrullinate peptide antibody levels at W12 and W24 compared to Control group. HRCT score showed no difference between Iguratimod group and Control group at any time points. As to lung function indexes, forced vital capacity percent predicted [FVC (% predicted)], carbon monoxide diffusion capacity percent predicted [DLCO (%predicted)] and 6-minute-walk distance (6MWD) were all higher in Iguratimod group compared with Control group at W4, W12 and W24. Besides, no difference in adverse events was discovered between these two groups. CONCLUSIONS: Iguratimod attenuates general inflammation, disease activity, and improves lung function in RA-ILD patients.


Subject(s)
Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , Chromones/therapeutic use , Lung Diseases, Interstitial/drug therapy , Sulfonamides/therapeutic use , Adult , Aged , Antirheumatic Agents/administration & dosage , Arthritis, Rheumatoid/diagnosis , Chromones/administration & dosage , Female , Humans , Lung Diseases, Interstitial/diagnosis , Male , Middle Aged , Respiratory Function Tests , Sulfonamides/administration & dosage , Tomography, X-Ray Computed , Young Adult
15.
Cancer Med ; 10(18): 6428-6441, 2021 09.
Article in English | MEDLINE | ID: mdl-34337875

ABSTRACT

INTRODUCTION: The AURKA gene encodes a protein kinase involved in cell cycle regulation and plays an oncogenic role in many cancers. The main objective of this study is to analyze AURKA expression in 13 common cancers and its role in prognostic and drug resistance. METHOD: Using the cancer genome atlas (TCGA) as well as CCLE and GDSC data, the level of AURKA gene expression and its role in prognosis and its association with drug resistance were evaluated, respectively. In addition, the expression level of AURKA was assessed in colorectal cancer (CRC) and gastric cancer (GC) samples. Besides, using Gene Expression Omnibus (GEO) data, drugs that could affect the expression level of this gene were also identified. RESULTS: The results indicated that the expression level of AURKA gene in 13 common cancers increased significantly compared to normal samples or it survived poorly (HR >1, p < 0.01) in lung, prostate, kidney, bladder, and uterine cancers. Also, the gene expression data showed increased expression in CRC and GC samples compared to normal ones. The level of AURKA was significantly associated with the resistance to SB 505124, NU-7441, and irinotecan drugs (p < 0.01). Eventually, GEO data showed that JQ1, actinomycin D1, and camptothecin could reduce the expression of AURKA gene in different cancer cell lines (logFC < 1, p < 0.01). CONCLUSION: Increased expression of AURKA is observed in prevalent cancers and associated with poor prognostic and the development of drug resistance. In addition, some chemotherapy drugs can reduce the expression of this gene.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Aurora Kinase A/metabolism , Biomarkers, Tumor/metabolism , Drug Resistance, Neoplasm/genetics , Neoplasms/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Aurora Kinase A/analysis , Aurora Kinase A/antagonists & inhibitors , Azepines/pharmacology , Azepines/therapeutic use , Benzodioxoles/pharmacology , Benzodioxoles/therapeutic use , Biomarkers, Tumor/analysis , Biomarkers, Tumor/antagonists & inhibitors , Camptothecin/pharmacology , Camptothecin/therapeutic use , Cell Line, Tumor , Chromones/pharmacology , Chromones/therapeutic use , Dactinomycin/analogs & derivatives , Dactinomycin/pharmacology , Dactinomycin/therapeutic use , Datasets as Topic , Drug Resistance, Neoplasm/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Imidazoles/pharmacology , Imidazoles/therapeutic use , Irinotecan/pharmacology , Irinotecan/therapeutic use , Kaplan-Meier Estimate , Male , Middle Aged , Morpholines/pharmacology , Morpholines/therapeutic use , Neoplasms/genetics , Neoplasms/mortality , Neoplasms/pathology , Prognosis , Pyridines/pharmacology , Pyridines/therapeutic use , Triazoles/pharmacology , Triazoles/therapeutic use
16.
Cell Tissue Res ; 386(1): 17-28, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34236519

ABSTRACT

Gliomas are aggressive brain tumors with very high resistance to chemotherapy throughout the overexpression of multiple intracellular survival pathways. Therefore, the aim of the present study was to investigate for the first time the anticancer activity of LY294002, phosphatidylinositol 3-kinase (PI3K) inhibitor and sorafenib, and rapidly accelerated fibrosarcoma kinase (Raf) inhibitor in the elimination of human glioma cells by programmed cell death. MOGGCCM (anaplastic astrocytoma, III) and T98G (glioblastoma multiforme, IV) cell lines incubated with LY294002 and/or sorafenib were used in the experiments. Simultaneous treatment with both drugs was more effective in the elimination of cancer cells on the way of apoptosis with no significant necrotic effect than single application. It was correlated with decreasing the mitochondrial membrane potential and activation of caspase 3 and 9. The expression of Raf and PI3K was also inhibited. Blocking of those kinases expression by specific siRNA revealed significant apoptosis induction, exceeding the level observed after LY294002 and sorafenib treatment in non-transfected lines but only in MOGGCCM cells. Our results indicated that combination of LY294002 and sorafenib was very efficient in apoptosis induction in glioma cells. Anaplastic astrocytoma cells turned out to be more sensitive for apoptosis induction than glioblastoma multiforme after blocking PI3K and Raf expression with siRNA.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Chromones/therapeutic use , Morpholines/therapeutic use , Sorafenib/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Chromones/pharmacology , Glioma/drug therapy , Glioma/mortality , Humans , Morpholines/pharmacology , Sorafenib/pharmacology , Survival Analysis , Transfection
17.
J Orthop Surg Res ; 16(1): 457, 2021 Jul 16.
Article in English | MEDLINE | ID: mdl-34271950

ABSTRACT

BACKGROUND: This study aims to evaluate the efficacy and safety of the iguratimod (IGU) as monotherapy or combined therapy in patients with rheumatoid arthritis (RA) by using meta-analysis. METHODS: We searched Medline, EMBASE, Cochrane library, CNKI, Wanfang medical network from initial to 30 June, 2020, for randomized clinical trials (RCTs). Two authors independently screened the studies via reading the title, abstract, and full text. The risk of bias in individual studies was assessed using the Cochrane Risk of Bias tool. STATA 12.0 was used for pooled analysis of all included studies. RESULTS: A total of 23 RCTs were included in this analysis. Meta-analysis showed that patients in the IGU monotherapy or combined therapy group had significantly higher ACR20 (OR = 1.97, 95% CI 1.29 to 3.00, P = 0.002), lower DAS28-CRP (SMD = -3.49, 95% CI -5.40 to -1.58, P < 0.001) and DAS28-ESR (SMD = -2.61, 95% CI -3.64 to -1.57, P < 0.001), as well as shorter duration of morning stiffness (SMD = -2.06, 95% CI -2.86 to -1.25, P < 0.001) and lower HAQ score (SMD = -0.91, 95% CI -1.61 to -0.21, P = 0.011), than those received other disease-modifying antirheumatic drugs (DMARDs) monotherapy (primarily comprising methotrexate). For the safety profile, IGU monotherapy had similar risks for gastrointestinal reactions (P = 0.070), leucopenia (P = 0.309), increment in transaminase (P = 0.321), increase of ALT (P = 0.051), and liver damage (P = 0.182) to methotrexate monotherapy, and IGU combined with other DMARDs therapy did not increase the risks of these AEs (P > 0.05). CONCLUSIONS: Our evidence suggests that IGU is effective and tolerant as monotherapy or combined therapy especially with methotrexate in patients with active RA. IGU may be regarded as a potential alternative to methotrexate, and a preferable choice when combined with other DMARDs for the treatment of RA.


Subject(s)
Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , Chromones/therapeutic use , Sulfonamides/therapeutic use , Drug Therapy, Combination , Humans , Randomized Controlled Trials as Topic
18.
Inflammation ; 44(5): 1895-1907, 2021 Oct.
Article in English | MEDLINE | ID: mdl-33860870

ABSTRACT

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


Subject(s)
Asthma/metabolism , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Toll-Like Receptor 4/metabolism , Animals , Anti-Asthmatic Agents/pharmacology , Anti-Asthmatic Agents/therapeutic use , Asthma/chemically induced , Asthma/drug therapy , Asthma/pathology , Chromones/pharmacology , Chromones/therapeutic use , Female , Metformin/pharmacology , Metformin/therapeutic use , Mice , Mice, Inbred BALB C , Morpholines/pharmacology , Morpholines/therapeutic use , Myeloid Differentiation Factor 88/antagonists & inhibitors , NF-kappa B/antagonists & inhibitors , Naphthyridines/pharmacology , Naphthyridines/therapeutic use , Ovalbumin/toxicity , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Phosphoinositide-3 Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/physiology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Toll-Like Receptor 4/antagonists & inhibitors
19.
Int Immunopharmacol ; 96: 107563, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33812258

ABSTRACT

OBJECTIVE: Iguratimod, an anti-rheumatic drug, has been widely used in the treatment of rheumatoid arthritis, but is still at an investigative stage for treatment of systemic lupus erythematosus (SLE). We examined the therapeutic effects of iguratimod and the mechanism underlying the efficacy in murine lupus model. METHODS: Pristane-induced lupus model of BALB/c mice (PI mice) were treated with iguratimod and mycophenolate mofetil. Proteinuria, anti-dsDNA antibodies and immunoglobulins production were measured. Renal pathology was evaluated. The percentage of Th17 and Treg cells in spleen and the expression of cytokines and mRNAs related to Th17 and Treg cells was analyzed. RESULTS: Iguratimod attenuated the severity of nephritis in PI mice in a dose-dependent manner. Proteinuria was continuously decreased and pathology of glomerulonephritis and tubulonephritis was significantly reduced along with reduction of glomerular immune complex deposition. Also, serum anti-dsDNA and total IgG and IgM levels were reduced by iguratimod in mice. It is worth mentioning that the efficacy of the 30 mg/kg/d iguratimod dose is comparable to, or even better than, 100 mg kg/d of mycophenolate mofetil. Furthermore, the percentage of Th17 cells was found decreased and the percentage of Treg cells increased. ROR-γt mRNA and serum cytokines (IL-17A and IL-22) of Th17 cells decreased accordingly. By contrast, Foxp3 mRNA and cytokines (TGF-ß and IL-10) of Treg cells increased. CONCLUSION: Iguratimod ameliorates nephritis of SLE and modulates the Th17/Treg ratio in murine nephritis of SLE, suggesting that Iguratimod could be an effective drug in treatment of SLE.


Subject(s)
Arthritis, Rheumatoid/drug therapy , Chromones/therapeutic use , Immunosuppressive Agents/therapeutic use , Lupus Erythematosus, Systemic/drug therapy , Lupus Nephritis/drug therapy , Sulfonamides/therapeutic use , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Animals , Antibodies, Antinuclear/blood , Cytokines/metabolism , Disease Models, Animal , Forkhead Transcription Factors/metabolism , Humans , Immunoglobulins/blood , Mice , Mice, Inbred BALB C , Proteinuria , Terpenes/administration & dosage
20.
Int Immunopharmacol ; 97: 107596, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33892300

ABSTRACT

OBJECTIVE: Our study aimed to investigate the effect of Iguratimod (IGU) on bleomycin (BLM)-induced interstitial lung disease (ILD). METHODS: The pulmonary fibrosis model group mice were developed by intratracheal injection of BLM. Mice were divided into two groups at random: (1) Control group (BLM group) - endotracheal BLM (BLM, 3.5 mg/kg, Kayaku, Japan) plus an intraperitoneal injection of normal saline, and (2) BLM + IGU group - intratracheal BLM (same as the control group) + IGU intraperitoneal injection (50 mg/kg/d). The alveolar lavage fluid, histopathology/immunohistochemistry, imaging, and other tests were performed on days 7, 14, 21, and 28 after injection. RESULTS: Lung function, including Compliance (Crs),Tissue damping (G), Static compliance (Cst), Inspiratory capacity (IC), Elastance (Ers), Tissue elastance (H) and Respiratory system resistance (Rrs) in mice, was improved by IGU. IGU reduced BLM-induced changes in pulmonary fibrosis and pulmonary inflammation, as shown in histological examination.Collagen production and inflammatory damage in the lungs caused by BLM were also reduced by IGU. IGU reduced the expression of immunoglobulin IgG and type I collagen in BLM-induced pulmonary fibrosis mice by inhibiting the production of B cells and immunoglobulin, and also delayed the deterioration of imaging changes. CONCLUSION: IGU inhibits immunoglobulin secretion by B cells to relieve pulmonary inflammation and fibrosis. IGU also plays a protective role in the lung in ILD.


Subject(s)
B-Lymphocytes/drug effects , Chromones/pharmacology , Lung Diseases, Interstitial/drug therapy , Sulfonamides/pharmacology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Bleomycin/adverse effects , Chromones/therapeutic use , Disease Models, Animal , Humans , Immunoglobulins/metabolism , Lung/drug effects , Lung/immunology , Lung/pathology , Lung Diseases, Interstitial/chemically induced , Lung Diseases, Interstitial/immunology , Lung Diseases, Interstitial/pathology , Mice , Sulfonamides/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL