Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Eur J Pharmacol ; 960: 176116, 2023 Dec 05.
Article in English | MEDLINE | ID: mdl-38059443

ABSTRACT

Cardiac fibrosis (CF) in response to persistent exogenous stimuli or myocardial injury results in cardiovascular diseases (CVDs). Protein tyrosine phosphatase 1B (PTP1B) can promote collagen deposition through regulating AMPK/TGF-ß/Smads signaling pathway, and PTP1B knockout improves cardiac dysfunction against overload-induced heart failure. Oleanolic acid (OA) has been proven to be an inhibitor of PTP1B, and its anti-cardiac remodeling effects have been validated in different mouse models. To improve the bioactivity of OA and to clarify whether OA derivatives with stronger inhibition of PTP1B activity have greater prevention of cardiac remodeling than OA, four new OA derivatives were synthesized and among them, we found that compound B had better effects than OA in inhibiting cardiac fibrosis both in vivo in the isoproterenol (ISO)-induced mouse cardiac fibrosis and in vitro in the TGF-ß/ISO-induced 3T3 cells. Combining with the results of molecular docking, surface plasmon resonance and PTP1B activity assay, we reported that OA and compound B directly bound to PTP1B and inhibited its activity, and that compound B showed comparable binding capability but stronger inhibitory effect on PTP1B activity than OA. Moreover, compound B presented much greater effects on AMPK activation and TGF-ß/Smads inhibition than OA. Taken together, OA derivative compound B more significantly alleviated cardiac fibrosis than OA through much greater inhibition of PTP1B activity and thus much stronger regulation of AMPK/TGF-ß/Smads signaling pathway.


Subject(s)
Oleanolic Acid , Transforming Growth Factor beta , Animals , Mice , Transforming Growth Factor beta/metabolism , Oleanolic Acid/pharmacology , Oleanolic Acid/therapeutic use , AMP-Activated Protein Kinases/metabolism , Signal Transduction , Molecular Docking Simulation , Fibrosis , Transforming Growth Factor beta1/metabolism
2.
Eur J Med Chem ; 260: 115723, 2023 Nov 15.
Article in English | MEDLINE | ID: mdl-37595545

ABSTRACT

N-acetylneuraminic acid (Neu5Ac) is a glycan receptor of viruses spread in many eukaryotic cells. The present work aimed to design, synthesis and biological evaluation of a panel of Neu5Ac derivatives based on a cyclodextrin (CD) scaffold for targeting influenza and coronavirus membrane proteins. The multivalent Neu5Ac glycoclusters efficiently inhibited chicken erythrocyte agglutination induced by intact influenza virus in a Neu5Ac density-dependent fashion. Compared with inhibition by Neu5Ac, the multivalent inhibitor with 21 Neu5Ac residues on the primary face of the ß-CD scaffold afforded 1788-fold higher binding affinity inhibition for influenza virus hemagglutinin with a dissociation constant (KD) of 3.87 × 10-7 M. It showed moderate binding affinity to influenza virus neuraminidase, but with only about one-thirtieth the potency of that with the HA protein. It also exhibited strong binding affinity to the spike protein of three human coronaviruses (severe acute respiratory syndrome coronavirus, Middle East respiratory syndrome coronavirus, and severe acute respiratory syndrome coronavirus 2), with KD values in the low micromolar range, which is about 10-time weaker than that of HA. Therefore, these multivalent sialylated CD derivatives have possible therapeutic application as broad-spectrum antiviral entry inhibitors for many viruses by targeting the Neu5Ac of host cells.


Subject(s)
COVID-19 , Cyclodextrins , HIV Fusion Inhibitors , Influenza, Human , Humans , Animals , N-Acetylneuraminic Acid , Antiviral Agents/pharmacology , Chickens
3.
J Med Chem ; 65(10): 7154-7169, 2022 05 26.
Article in English | MEDLINE | ID: mdl-35579113

ABSTRACT

Influenza hemagglutinin that drives viral entry into cells via the membrane fusion process is an up-and-coming antiviral drug target. Herein, we described for the first time the design, synthesis, and biological characteristics of a new class of pentacyclic triterpenoid-based proteolysis targeting chimeras (PROTACs) to enhance the degradation of hemagglutinin target. Among these PROTACs, V3 showed the best degradation effect on the hemagglutinin with a median degradation concentration of 1.44 µM in a ubiquitin and proteasome-dependent manner and broad-spectrum anti-influenza A virus activity but not affected the entry of influenza virus. Moreover, intravenous injection of V3 protected mice against influenza A virus-induced toxic effects. Further diazirine-containing photo-crosslinking mass spectrometric analysis of hemagglutinin complexes indicated crosslinking to Asn15, Thr31, and Asn27, a novel target of hemagglutinin. Taken together, our data revealed that oleanolic acid-based PROTACs could degrade hemagglutinin protein, providing a new direction toward the discovery of potential anti-influenza drugs.


Subject(s)
Influenza, Human , Triterpenes , Animals , Chimera/metabolism , Hemagglutinins , Humans , Influenza, Human/drug therapy , Intercellular Signaling Peptides and Proteins , Mice , Proteins/metabolism , Proteolysis , Triterpenes/chemistry
4.
Front Chem ; 10: 836955, 2022.
Article in English | MEDLINE | ID: mdl-35494649

ABSTRACT

In our continuing efforts toward the design of novel pentacyclic triterpene derivatives as potential anti-influenza virus entry inhibitors, a series of homogeneous heptavalent glycyrrhetinic acid derivatives based on ß-cyclodextrin scaffold were designed and synthesized by click chemistry. The structure was unambiguously characterized by NMR, IR, and MALDI-TOF-MS measurements. Seven conjugates showed sufficient inhibitory activity against influenza virus infection based on the cytopathic effect reduction assay with IC50 values in the micromolar range. The interactions of conjugate 37, the most potent compound (IC50 = 2.86 µM, CC50 > 100 µM), with the influenza virus were investigated using the hemagglutination inhibition assay. Moreover, the surface plasmon resonance assay further confirmed that compound 37 bound to the influenza HA protein specifically with a dissociation constant of 5.15 × 10-7 M. Our results suggest the promising role of ß-cyclodextrin as a scaffold for preparing a variety of multivalent compounds as influenza entry inhibitors.

5.
Oxid Med Cell Longev ; 2022: 7038834, 2022.
Article in English | MEDLINE | ID: mdl-35281460

ABSTRACT

In this study, sulfated polysaccharides extracted from Laminaria japonica were degraded by free radicals to obtain low molecular weight fucoidan (LMWF). The in vivo and in vitro effects of LMWF on bleomycin-treated pulmonary fibrosis mice and TGF-treated A549 cells, respectively, were evaluated, and the role of antioxidant activity was assessed. H&E, Masson's trichrome, and Sirius red staining results showed that bleomycin induced obvious pathological changes and collagen deposition in the lung tissue of mice. However, LMWF effectively inhibited collagen deposition, and based on immunohistochemistry analyses, LMWF can also inhibit the expression of fibrosis markers. At the same time, LMWF could regulate related antioxidant factors in the lung tissue of pulmonary fibrosis mice and reduce the pressure of oxidative stress. Moreover, LMWF could improve the morphology of cells induced with TGF, which confirmed that LMWF could inhibit fibrosis via antioxidant activity modulation.


Subject(s)
Antioxidants/therapeutic use , Polysaccharides/therapeutic use , Pulmonary Fibrosis/drug therapy , Animals , Antioxidants/pharmacology , Humans , Male , Mice , Molecular Weight , Polysaccharides/pharmacology , Pulmonary Fibrosis/mortality , Survival Analysis
6.
J Nanosci Nanotechnol ; 18(6): 3901-3907, 2018 Jun 01.
Article in English | MEDLINE | ID: mdl-29442725

ABSTRACT

MoS2, a kind of two-dimensional material with unique performances, has been widely used in many fields. However, an in-depth understanding of its toxicity is still needed, let alone its effects on the environmental microorganism. Herein, we used different methods, including metabolomics technology, to investigate the influence of bulk MoS2 (BMS) on yeast cells. The results indicated that high concentrations (1 mg/L and more) of BMS could destroy cell membrane and induce ROS accumulation. When exposed to a low concentration of BMS (0.1 mg/L), the intracellular concentrations of many metabolites (e.g., fumaric acid, lysine) increased. However, most of their concentrations descended significantly as the yeast cells were treated with BMS of high concentrations (1 mg/L and more). Metabolomics analysis further revealed that exposure to high concentrations of BMS could significantly affect some metabolic pathways such as amino acid and citrate cycle related metabolism. These findings will be beneficial for MoS2 toxicity assessment and further applications.


Subject(s)
Metabolomics , Nanoparticles , Saccharomyces cerevisiae/metabolism , Metabolic Networks and Pathways , Metabolome
7.
Chemosphere ; 198: 216-225, 2018 May.
Article in English | MEDLINE | ID: mdl-29421733

ABSTRACT

As a new rising star in the post-graphene two-dimensional materials (2DMs), molybdenum disulfide (MoS2) attracts increasing attentions and is widely applied. However, the chemical and toxicological interaction between MoS2 and other co-contaminants is still poorly understood. Nano-silver (N-Ag) is the most commonly used nanomaterial in commercial products and distributed widely in the environment. Herein, we investigated the effects of chitosan functionalized MoS2 (CS-MoS2) nanosheets, a water-dispersible form of MoS2, on the microbial toxicity of N-Ag. We found that the incorporation of CS-MoS2 nanosheets attenuated the oxidative stress induced by N-Ag on yeast cells, while caused more membrane stress. In addition, the inhibition of N-Ag on the metabolic activities of yeast cells could be attenuated by CS-MoS2 nanosheets as well. The coexistence of N-Ag and CS-MoS2 nanosheets mainly perturbed the amino acid-related metabolic pathways in yeast cells, and phosphoric acid was a potential nanotoxicity biomarker. We further found that CS-MoS2 nanosheets dramatically absorbed the Ag ion released from N-Ag, which might be responsible for its attenuation effect on the microbial toxicity of N-Ag. Our findings provide more new insights for the ecotoxicity evaluation of MoS2 and other 2DMs.


Subject(s)
Disulfides/chemistry , Metabolome/drug effects , Molybdenum/chemistry , Nanostructures/chemistry , Saccharomyces cerevisiae/drug effects , Silver/toxicity , Chitosan/chemistry , Drug Interactions , Metabolic Networks and Pathways/drug effects , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Saccharomyces cerevisiae/metabolism , Silver/chemistry , Surface Properties
8.
RSC Adv ; 8(32): 17826-17836, 2018 May 14.
Article in English | MEDLINE | ID: mdl-35542100

ABSTRACT

Molybdenum disulfide (MoS2), one of the next-generation two-dimensional materials (2DMs), has attracted increasing attention due to its unique physicochemical properties. However, the aquatic toxicity of dispersible MoS2 is still unknown. Herein, we synthesized chitosan functionalized MoS2 (CS-MoS2) micro-sheets with a satisfying water-dispersible performance. The average length and width of the as-prepared CS-MoS2 micro-sheets were 5.04 µm and 3.12 µm, respectively, and they had a pristine 2H polymorph. The toxicity of CS-MoS2 micro-sheets was assessed by investigating the organs, gills and liver of adult zebrafish. We found that exposure to high concentrations of CS-MoS2 micro-sheets (10 mg L-1 and 20 mg L-1) led to lamellar fusions in the gills, and significant localized lesions, such as peripheral nuclei and vacuole formation, in the liver. In addition, treatment with 20 mg L-1 CS-MoS2 micro-sheets suppressed gene expression of antioxidant enzymes (e.g., CAT and GPx1a gene) and induced the expression levels of the proinflammatory response and apoptosis (e.g., IL-1ß, IL-6, and AIF gene) in gill and liver tissues. Further, reactive oxygen species (ROS) were generated upon treatment with 20 mg L-1 CS-MoS2 micro-sheets in both organs. To the best of our knowledge, this is the first investigation of the aquatic toxicity of dispersible MoS2 in zebrafish, and further highlights the potential environmental risk of MoS2.

9.
ACS Biomater Sci Eng ; 3(12): 3261-3272, 2017 Dec 11.
Article in English | MEDLINE | ID: mdl-33445369

ABSTRACT

In postgraphene two-dimensional materials (2DMs), MoS2 has attracted increasing attention in the biomedical field due to its excellent physicochemical properties. However, the toxicity and biocompatibility evaluation of MoS2 is not fully addressed. Herein, chitosan functionalized MoS2 (CS-MoS2) nanosheets, which showed perfect dispersibility and stability performances, were synthesized and characterized. We found that CS-MoS2 nanosheets inhibited the viability of human dermal fibroblasts (HDFs) moderately while causing cell membrane instability, ROS generation, and DNA damage in a dosage-dependent manner. CS-MoS2 nanosheets did not induce significant changes in the cell morphologies, but they seemed to impair the cell division of HDFs. CS-MoS2 nanosheets (100 µg/mL) activated EGFR and induced reactive oxygen species, Smad, and IL-1, which in turn led to cell inflammation and apoptosis. Furthermore, HDFs showed cellular stress responses when they were exposed to low concentrations of CS-MoS2 nanosheets (25 and 100 µg/mL) because most of the intracellular metabolites such as amino acids were induced at 25 µg/mL but were inhibited at 100 µg/mL. Pyroglutamic acid, phosphoric acid, and inositol might be used as biomarkers for evaluating the toxicity of CS-MoS2 nanosheets. Additionally, 100 µg/mL CS-MoS2 nanosheets inhibited glutathione metabolism and induced the imbalance of cellular redox homeostasis. It further suppressed the tricarboxylic acid cycle and other metabolic pathways, causing insufficient supply of substrates and energy for HDFs. These findings will fuel the risk assessment of MoS2 and other 2DMs and guide the safe material design and 2DM applications.

SELECTION OF CITATIONS
SEARCH DETAIL
...