Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
J Chem Inf Model ; 64(8): 3047-3058, 2024 04 22.
Article in English | MEDLINE | ID: mdl-38520328

ABSTRACT

Covalent drugs exhibit advantages in that noncovalent drugs cannot match, and covalent docking is an important method for screening covalent lead compounds. However, it is difficult for covalent docking to screen covalent compounds on a large scale because covalent docking requires determination of the covalent reaction type of the compound. Here, we propose to use deep learning of a lateral interactions spiking neural network to construct a covalent lead compound screening model to quickly screen covalent lead compounds. We used the 3CL protease (3CL Pro) of SARS-CoV-2 as the screen target and constructed two classification models based on LISNN to predict the covalent binding and inhibitory activity of compounds. The two classification models were trained on the covalent complex data set targeting cysteine (Cys) and the compound inhibitory activity data set targeting 3CL Pro, respected, with good prediction accuracy (ACC > 0.9). We then screened the screening compound library with 6 covalent binding screening models and 12 inhibitory activity screening models. We tested the inhibitory activity of the 32 compounds, and the best compound inhibited SARS-CoV-2 3CL Pro with an IC50 value of 369.5 nM. Further assay implied that dithiothreitol can affect the inhibitory activity of the compound to 3CL Pro, indicating that the compound may covalently bind 3CL Pro. The selectivity test showed that the compound had good target selectivity to 3CL Pro over cathepsin L. These correlation assays can prove the rationality of the covalent lead compound screening model. Finally, covalent docking was performed to demonstrate the binding conformation of the compound with 3CL Pro. The source code can be obtained from the GitHub repository (https://github.com/guzh970630/Screen_Covalent_Compound_by_LISNN).


Subject(s)
Coronavirus 3C Proteases , Molecular Docking Simulation , Neural Networks, Computer , SARS-CoV-2 , Coronavirus 3C Proteases/metabolism , Coronavirus 3C Proteases/antagonists & inhibitors , Coronavirus 3C Proteases/chemistry , SARS-CoV-2/enzymology , SARS-CoV-2/drug effects , Humans , Drug Discovery , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Antiviral Agents/metabolism , Protease Inhibitors/pharmacology , Protease Inhibitors/chemistry , Protease Inhibitors/metabolism , COVID-19 Drug Treatment , Deep Learning , Protein Binding , COVID-19/virology
2.
Bioorg Chem ; 82: 58-67, 2019 02.
Article in English | MEDLINE | ID: mdl-30268974

ABSTRACT

Ribosomal protein S1 (RpsA) has been identified as a novel target of pyrazinoic acid (POA), which is the active form of pyrazinamide (PZA), in vivo. RpsA plays a crucial role in trans-translation, which is widespread in microbes. In our investigation, we first described the discovery of promising RpsA antagonists for drug-resistant mycobacterium (MtRpsAd438A) and M. smegmatis, as well as wild-type M. tuberculosis. These antagonists were discovered via structure/ligand-based virtual screening approaches. A total of 21 targeted compounds were selected by virtual screening, combined scores, affinity, similarities and rules for potential as drugs. Next, the affinities of these compounds for three targeted proteins were tested in vitro by applying various technologies, including fluorescence quenching titration (FQT), saturation transfer difference (STD), and chemical shift perturbation (CSP) assays. The results showed that seven compounds had a high affinity for the targeted proteins. Our discovery set the stage for discovering new chemical entities (NCEs) for PZA-resistant tuberculosis and providing key residues for rational drug design to target RpsA.


Subject(s)
Antitubercular Agents/pharmacology , Azoles/pharmacology , Bacterial Proteins/antagonists & inhibitors , Heterocyclic Compounds, 2-Ring/pharmacology , Ribosomal Proteins/antagonists & inhibitors , Antitubercular Agents/chemistry , Azoles/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Binding Sites , Drug Evaluation, Preclinical , Heterocyclic Compounds, 2-Ring/chemistry , Microbial Sensitivity Tests , Molecular Docking Simulation , Molecular Dynamics Simulation , Mutation , Mycobacterium smegmatis/drug effects , Mycobacterium tuberculosis/drug effects , Ribosomal Proteins/chemistry , Ribosomal Proteins/genetics , Software
3.
Molecules ; 24(10)2019 May 20.
Article in English | MEDLINE | ID: mdl-31137573

ABSTRACT

The programmed cell death ligand protein 1 (PD-L1) is a member of the B7 protein family and consists of 290 amino acid residues. The blockade of the PD-1/PD-L1 immune checkpoint pathway is effective in tumor treatment. Results: Two pharmacophore models were generated based on peptides and small molecules. Hypo 1A consists of one hydrogen bond donor, one hydrogen bond acceptor, two hydrophobic points and one aromatic ring point. Hypo 1B consists of one hydrogen bond donor, three hydrophobic points and one positive ionizable point. Conclusions: The pharmacophore model consisting of a hydrogen bond donor, hydrophobic points and a positive ionizable point may be helpful for designing small-molecule inhibitors targeting PD-L1.


Subject(s)
Peptides/pharmacology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Humans , Inhibitory Concentration 50 , Molecular Docking Simulation , Programmed Cell Death 1 Receptor/metabolism , ROC Curve , Reproducibility of Results
4.
J Enzyme Inhib Med Chem ; 32(1): 624-631, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28260395

ABSTRACT

Fibrotic diseases have become a major cause of death in the developed world. AdipoR1 agonists are potent inhibitors of fibrotic responses. Here, we focused on the in silico identification of novel AdipoR1 peptide agonists. A homology model was constructed to predict the 3D structure of AdipoR1. By docking to known active peptides, the putative active site of the model was further explored. A virtual screening study was then carried out with a set of manually designed peptides using molecular docking. Peptides with high docking scores were then evaluated for their anti-fibrotic properties. The data indicated that the novel peptide Pep70 significantly inhibited the proliferation of hepatic stellate cells (HSC) and NIH-3T3 cells (18.33% and 27.80%) and resulted in favouring cell-cycle arrest through increasing the accumulation of cells in the G0/G1 phase by 17.08% and 15.86%, thereby reducing the cell population in the G2/M phase by 11.25% and 15.95%, respectively. Additionally, Pep70 exhibited the most marked suppression on the expression of α-smooth muscle actin (α-SMA), collagen type I alpha1 (COL1A1) and TGF-ß1. Therefore, the peptide Pep70 was ultimately identified as an inhibitor of fibrotic responses and as a potential AdipoR1 agonist.


Subject(s)
Oligopeptides/chemistry , Peptides/pharmacology , Receptors, Adiponectin/agonists , Amino Acid Sequence , Animals , Binding Sites/drug effects , Cell Proliferation/drug effects , Crystallography, X-Ray , Mice , Models, Molecular , Molecular Docking Simulation , NIH 3T3 Cells , Oligopeptides/genetics , Peptides/chemical synthesis , Peptides/chemistry , Rats , Sequence Homology, Amino Acid , Transforming Growth Factor beta1/agonists , Transforming Growth Factor beta1/metabolism
5.
Bioorg Med Chem Lett ; 25(11): 2345-52, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25937012

ABSTRACT

Caseinolytic protein proteases (ClpP) are large oligomeric protein complexes that contribute to cell homeostasis as well as virulence regulation in bacteria. Inhibitors of ClpP can significantly attenuate the capability to produce virulence factors of the bacteria. In this work, we developed a workflow to expand the chemical space of potential ClpP inhibitors based on a set of ß-lactones. In our workflow, an artificial pharmacophore model was generated based on HipHop and HYPOGEN method. A de novo compound library based on molecular fingerprints was constructed and virtually screened by the pharmacophore model. The results were further investigated by molecular docking study. The workflow successfully achieved potential ClpP inhibitors. It could be applied to design more novel potential ClpP inhibitors and provide theoretical basis for the further optimization of the hit compounds.


Subject(s)
Lactones/pharmacology , Protease Inhibitors/pharmacology , Drug Design , Lactones/chemistry , Models, Chemical , Models, Molecular , Molecular Structure , Protease Inhibitors/chemistry , Protease Inhibitors/metabolism , Protein Conformation , Quantitative Structure-Activity Relationship
6.
Acta Biochim Biophys Sin (Shanghai) ; 47(10): 842-50, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26350098

ABSTRACT

The cellular prion protein (PrP(C)) is a kind of cell-surface Cu(2+)-binding glycoprotein. The oligomerization of PrP(C) is highly related to transmissible spongiform encephalopathies (TSEs). Cu(2+) plays a vital role in the oligomerization of PrP(C), and participates in the pathogenic process of TSE diseases. It is expected that Cu(2+)-binding has different effects on the oligomerization of TSE-sensitive human PrP(C) (HuPrP(C)) and TSE-resistant rabbit PrP(C) (RaPrP(C)). However, the details of the distinct effects remain unclear. In the present study, we measured the interactions of Cu(2+) with HuPrP(C) (91-230) and RaPrP(C) (91-228) by isothermal titration calorimetry, and compared the effects of Cu(2+)-binding on the oligomerization of both PrPs. The measured dissociation constants (Kd) of Cu(2+) were 11.1 ± 2.1 µM for HuPrP(C) and 21.1 ± 3.1 µM for RaPrP(C). Cu(2+)-binding promoted the oligomerization of HuPrP(C) more significantly than that of RaPrP(C). The far-ultraviolet circular dichroism spectroscopy experiments showed that Cu(2+)-binding induced more significant secondary structure change and increased more ß-sheet content for HuPrP(C) compared with RaPrP(C). Moreover, the urea-induced unfolding transition experiments indicated that Cu(2+)-binding decreased the conformational stability of HuPrP(C) more distinctly than that of RaPrP(C). These results suggest that RaPrP(C) possesses a low susceptibility to Cu(2+), potentially weakening the risk of Cu(2+)-induced TSE diseases. Our work sheds light on the Cu(2+)-promoted oligomerization of PrP(C), and may be helpful for further understanding the TSE-resistance of rabbits.


Subject(s)
Copper/chemistry , PrPC Proteins/chemistry , Animals , Binding Sites , Computer Simulation , Dimerization , Humans , Models, Chemical , Models, Molecular , Protein Binding , Protein Conformation , Rabbits , Species Specificity
7.
Future Med Chem ; 16(9): 887-903, 2024.
Article in English | MEDLINE | ID: mdl-38618977

ABSTRACT

Background: The epidemic caused by SARS-CoV-2 swept the world in 2019. The 3C-like protease (3CLpro) of SARS-CoV-2 plays a key role in viral replication, and its inhibition could inhibit viral replication. Materials & methods: The virtual screen based on receptor-ligand pharmacophore models and molecular docking were conducted to obtain the novel scaffolds of the 3CLpro. The molecular dynamics simulation was also carried out. All compounds were synthesized and evaluated in biochemical assays. Results: The compound C2 could inhibit 3CLpro with a 72% inhibitory rate at 10 µM. The covalent docking showed that C2 could form a covalent bond with the Cys145 in 3CLpro. Conclusion: C2 could be a potent lead compound of 3CLpro inhibitors against SARS-CoV-2.


[Box: see text].


Subject(s)
Antiviral Agents , Coronavirus 3C Proteases , Drug Design , Molecular Docking Simulation , Molecular Dynamics Simulation , SARS-CoV-2 , Coronavirus 3C Proteases/antagonists & inhibitors , Coronavirus 3C Proteases/metabolism , SARS-CoV-2/drug effects , Antiviral Agents/pharmacology , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Humans , Protease Inhibitors/pharmacology , Protease Inhibitors/chemical synthesis , Protease Inhibitors/chemistry , COVID-19 Drug Treatment , Structure-Activity Relationship
8.
Expert Opin Ther Pat ; 34(3): 99-126, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38648107

ABSTRACT

INTRODUCTION: The TGF-ß signaling pathway is a complex network that plays a crucial role in regulating essential biological functions and is implicated in the onset and progression of multiple diseases. This review highlights the recent advancements in developing inhibitors targeting the TGF-ß signaling pathway and their potential therapeutic applications in various diseases. AREA COVERED: The review discusses patents on active molecules related to the TGF-ß signaling pathway, focusing on three strategies: TGF-ß activity inhibition, blocking TGF-ß receptor binding, and disruption of the signaling pathway using small molecule inhibitors. Combination therapies and the development of fusion proteins targeting multiple pathways are also explored. The literature search was conducted using the Cortellis Drug Discovery Intelligence database, covering patents from 2021 onwards. EXPERT OPINION: The development of drugs targeting the TGF-ß signaling pathway has made significant progress in recent years. However, addressing challenges such as specificity, systemic toxicity, and patient selection is crucial for their successful clinical application. Targeting the TGF-ß signaling pathway holds promise as a promising approach for the treatment of various diseases.


Subject(s)
Drug Development , Molecular Targeted Therapy , Patents as Topic , Receptors, Transforming Growth Factor beta , Signal Transduction , Transforming Growth Factor beta , Humans , Signal Transduction/drug effects , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/antagonists & inhibitors , Animals , Receptors, Transforming Growth Factor beta/metabolism , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Drug Discovery
9.
Bioorg Med Chem Lett ; 23(14): 4166-71, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23743285

ABSTRACT

As increasing drug-resistance poses an emerging threat to public health, the development of novel antibacterial agents is critical. We developed a workflow consisting of various methods for de novo design. In the workflow, 2D-QSAR model based on molecular fingerprints was constructed to extract the bioactive molecular fingerprints from a data set of DNA-gyrase inhibitors with new structure and mechanism. These fingerprints were converted into molecular fragments which were recombined to generate compound library. The new compound library was virtually screened by LigandFit and Gold docking, and the results were further investigated by pharmacophore validation and binding mode analysis. The workflow successfully achieved a potential DNA-gyrase inhibitor. It could be applied to design more novel potential DNA-gyrase inhibitors and provide theoretical basis for further optimization of the hit compounds.


Subject(s)
Anti-Bacterial Agents/chemistry , DNA Gyrase/chemistry , Drug Design , Topoisomerase II Inhibitors/chemistry , Anti-Bacterial Agents/chemical synthesis , Binding Sites , DNA Gyrase/metabolism , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Molecular Docking Simulation , Protein Structure, Quaternary , Quantitative Structure-Activity Relationship , Topoisomerase II Inhibitors/chemical synthesis
10.
Front Chem ; 11: 1292869, 2023.
Article in English | MEDLINE | ID: mdl-37927570

ABSTRACT

Identifying compound-protein interaction plays a vital role in drug discovery. Artificial intelligence (AI), especially machine learning (ML) and deep learning (DL) algorithms, are playing increasingly important roles in compound-protein interaction (CPI) prediction. However, ML relies on learning from large sample data. And the CPI for specific target often has a small amount of data available. To overcome the dilemma, we propose a virtual screening model, in which word2vec is used as an embedding tool to generate low-dimensional vectors of SMILES of compounds and amino acid sequences of proteins, and the modified multi-grained cascade forest based gcForest is used as the classifier. This proposed method is capable of constructing a model from raw data, adjusting model complexity according to the scale of datasets, especially for small scale datasets, and is robust with few hyper-parameters and without over-fitting. We found that the proposed model is superior to other CPI prediction models and performs well on the constructed challenging dataset. We finally predicted 2 new inhibitors for clusters of differentiation 47(CD47) which has few known inhibitors. The IC50s of enzyme activities of these 2 new small molecular inhibitors targeting CD47-SIRPα interaction are 3.57 and 4.79 µM respectively. These results fully demonstrate the competence of this concise but efficient tool for CPI prediction.

11.
J Pept Sci ; 18(6): 413-7, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22517776

ABSTRACT

We have previously shown that a recombinant human PTH fragment, Pro-Pro-[Arg11] hPTH (1-34)-Pro-Pro-Asp (hPTH'), could be a potentially better and more cost-effective therapeutic agent than PTH (1-34) on osteoporosis. In this report, we characterized the solution conformations of hPTH' by NMR spectroscopy and modeled the interactions between the hPTH' and the PTH receptor. By comparing it with PTH (1-34) structures and their respective interactions with the PTH receptor, we identified two segments of helix extending from Ile5 to Met8 and from Glu22 to Gln29 with a divided kink between the two helixes around Arg20. Mutated arginine makes hPTH' fill the receptor cavity better as well as forms hydrogen bonds with Val193. Understanding the ligand receptor interactions will help us design small molecules to better mimic the activities of PTH.


Subject(s)
Parathyroid Hormone/chemistry , Peptide Fragments/chemistry , Receptor, Parathyroid Hormone, Type 1/chemistry , Amino Acid Sequence , Humans , Models, Molecular , Protein Conformation
12.
Expert Opin Ther Pat ; 32(10): 1097-1122, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36175357

ABSTRACT

INTRODUCTION: Fibrosis is a disease that damages organs and even causes death. Because of the complicated pathogenesis, the development of drugs for fibrosis is challenging. In the lysophosphatidic acid receptor type 1 (LPA1) signaling pathway, LPA1 and its downstream Rho-associated coiled-coil forming protein kinase (ROCK) are related to the process of fibrosis. Targeting LPA1 signaling pathway is a potential strategy for the treatment of fibrosis. AREA COVERED: This review describes the process of fibrosis mediated by the LPA1 signaling pathway and then summarizes LPA1 antagonist patents reported since 2010 and ROCK inhibitor patents since 2017 according to their scaffolds based on the Cortellis Drug Discovery Intelligence database. Information on LPA1 antagonists entering clinical trials is integrated. EXPERT OPINION: Over the past decade, a large number of antagonists targeting the LPA1 signaling pathway have been patented for fibrosis therapy. A limited number of compounds have entered clinical trials. Different companies and research groups have used different scaffolds when designing compounds for fibrosis therapy. Therefore, LPA1 and ROCK are competitive targets for the development of new therapies for fibrosis to provide a potential treatment method for fibrosis in the future.


Subject(s)
Receptors, Lysophosphatidic Acid , rho-Associated Kinases , Humans , Receptors, Lysophosphatidic Acid/metabolism , Patents as Topic , Fibrosis , Signal Transduction , Lysophospholipids/metabolism
13.
Curr Med Chem ; 28(10): 2033-2047, 2021.
Article in English | MEDLINE | ID: mdl-32452320

ABSTRACT

Virtual screening is an important means for lead compound discovery. The scoring function is the key to selecting hit compounds. Many scoring functions are currently available; however, there are no all-purpose scoring functions because different scoring functions tend to have conflicting results. Recently, neural networks, especially convolutional neural networks, have constantly been penetrating drug design and most CNN-based virtual screening methods are superior to traditional docking methods, such as Dock and AutoDock. CNNbased virtual screening is expected to improve the previous model of overreliance on computational chemical screening. Utilizing the powerful learning ability of neural networks provides us with a new method for evaluating compounds. We review the latest progress of CNN-based virtual screening and propose prospects.


Subject(s)
Drug Design , Neural Networks, Computer , Humans , Ligands
14.
Expert Opin Ther Pat ; 31(8): 723-743, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33645365

ABSTRACT

INTRODUCTION: Fibrosis is a serious disease that occurs in many organs, such as kidney, liver and lung. The deterioration of these organs ultimately leads to death. Due to the complex mechanisms of fibrosis, research and development of antifibrotic drugs is difficult. One solution is to focus on core pathways, one of which is the TGF-ß signaling pathway. In virtually every type of fibrosis, TGF-ß signaling is recognized as a critical pathway. AREA COVERED: This review discusses patents on active molecules related to the TGF-ß signaling. Molecules targeting components related to the activation of TGF-ß are introduced. Several strategies preventing signal propagation from active TGF-ß to downstream targets are also introduced, including TGF-ß antibodies, TGF-ß ligand traps, and inhibitors of TGF-ß receptor kinases. Finally, molecules affecting downstream targets in both canonical and noncanonical TGF-ß signaling pathways are described. EXPERT OPINION: Since the approval of pirfenidone, targeting TGF-ß signaling has been anticipated as an effective therapy for fibrosis. The potential of this therapy has been further supported by emerging patents on the TGF-ß signaling. This pathway can be entirely inhibited, from the activation of TGF-ß to downstream signaling. Inhibiting TGF-ß signaling is expected to provide more effective treatments for fibrosis.


Subject(s)
Drug Development , Fibrosis/drug therapy , Transforming Growth Factor beta/antagonists & inhibitors , Animals , Fibrosis/pathology , Humans , Molecular Targeted Therapy , Patents as Topic , Signal Transduction/drug effects
15.
Comput Struct Biotechnol J ; 19: 5494-5503, 2021.
Article in English | MEDLINE | ID: mdl-34712395

ABSTRACT

Cluster of differentiation 47 (CD47)/signal regulatory protein alpha (SIRPα) is a negative innate immune checkpoint signaling pathway that restrains immunosurveillance and immune clearance, and thus has aroused wide interest in cancer immunotherapy. Blockade of the CD47/SIRPα signaling pathway shows remarkable antitumor effects in clinical trials. Currently, all inhibitors targeting CD47/SIRPα in clinical trials are biomacromolecules. The poor permeability and undesirable oral bioavailability of biomacromolecules have caused researchers to develop small-molecule CD47/SIRPα pathway inhibitors. This review will summarize the recent advances in CD47/SIRPα interactions, including crystal structures, peptides and small molecule inhibitors. In particular, we have employed computer-aided drug discovery (CADD) approaches to analyze all the published crystal structures and docking results of small molecule inhibitors of CD47/SIRPα, providing insight into the key interaction information to facilitate future development of small molecule CD47/SIRPα inhibitors.

16.
J Cheminform ; 12(1): 42, 2020 Jun 08.
Article in English | MEDLINE | ID: mdl-33430983

ABSTRACT

With the rise of artificial intelligence (AI) in drug discovery, de novo molecular generation provides new ways to explore chemical space. However, because de novo molecular generation methods rely on abundant known molecules, generated molecules may have a problem of novelty. Novelty is important in highly competitive areas of medicinal chemistry, such as the discovery of kinase inhibitors. In this study, de novo molecular generation based on recurrent neural networks was applied to discover a new chemical space of kinase inhibitors. During the application, the practicality was evaluated, and new inspiration was found. With the successful discovery of one potent Pim1 inhibitor and two lead compounds that inhibit CDK4, AI-based molecular generation shows potentials in drug discovery and development.

17.
Future Med Chem ; 12(2): 127-145, 2020 01.
Article in English | MEDLINE | ID: mdl-31718293

ABSTRACT

Aim: CDK4/6 have critical roles in the early stage of the cell cycle. CDK2 acts later in the cell cycle and has a considerably broader range of protein substrates, some of which are essential for normal cell proliferation. Therefore, increasing the selectivity of cyclin-dependent kinase (CDK) inhibitors is critical. Methodology: In this study, we construct a versatile, specific CDK4 pharmacophore model that not only matches well with 8119 of the reported 9349 CDK4/6 inhibitors but also differentiates from the CDK2 pharmacophore. Results & Conclusion: we demonstrate the activity and selectivity determinants of CDK4/6 selective inhibitors based on the CDK4 pharmacophore model. Finally, we propose the future optimization strategy for CDK4/6 selective inhibitors, providing a theoretical basis for further research and development of CDK4/6 selective inhibitors.


Subject(s)
Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Drug Development , Enzyme Inhibitors/pharmacology , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/metabolism , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Models, Molecular
18.
J Ethnopharmacol ; 261: 112978, 2020 Oct 28.
Article in English | MEDLINE | ID: mdl-32442586

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Huiyang Shengji formula (HSF) is a compound Chinese herbal medicine prescription, and has long been used for treating chronic non-healing wounds. AIM OF THE STUDY: The purpose of this study was to provide new insight into molecular mechanisms of healing effects of the HSF treatments. MATERIALS AND METHODS: We established a rat diabetic skin ulcer (DSU) model, and assessed healing effects of four HSF treatments on DSUs by calculating wound healing rates and immunohistochemical detection of the expressions of angiogenesis-related factors in the model rats (Mod) relative to normal rats (Nor), including Huiyang extract (HE), Shengji extract (SE), Huiyang Shengji extract (HSE) and HSE associated with acupuncture (Ac-HSE). We then performed NMR-based metabolomic analyses on skin tissues of the Nor, Mod, HSE-treated, Ac-HSE-treated rats to address metabolic mechanisms underlying these effects. RESULTS: These treatments up-regulated expressions of two angiogenesis-related factors VEGF and CD31, and improved efficacy of healing DSUs, in which HSE and Ac-HSE exhibited the most significant effects. Compared with Mod, HSE and Ac-HSE groups shared four characteristic metabolites (lactate, histidine, succinate and acetate) and four significantly altered metabolic pathways with Nor. Both HSE and Ac-HSE treatments could partly reverse the metabolically disordered pathological state of DSUs to the normal state. They might improve wound healing through promoting glucose metabolism, BCAAs metabolism, and enhancing antioxidant capacity and angiogenesis in DSU tissues. Ac-HSE significantly enhanced wound healing rates compared to HSE, potentially owing to significant capacities of enhancing anti-oxidation and angiogenesis and interfering three more metabolic pathways. CONCLUSIONS: This work provides a mechanistic understanding of the healing effects of the HSE and Ac-HSE treatments on DSUs, is of benefit to improvements of the HSF treatments for clinically healing chronic non-healing wounds.


Subject(s)
Acupuncture Therapy , Diabetic Angiopathies/therapy , Drugs, Chinese Herbal/pharmacology , Magnetic Resonance Spectroscopy , Metabolomics , Skin Ulcer/therapy , Skin/drug effects , Wound Healing/drug effects , Wounds and Injuries/therapy , Animals , Diabetes Mellitus, Experimental/chemically induced , Diabetic Angiopathies/metabolism , Diabetic Angiopathies/pathology , Disease Models, Animal , Energy Metabolism/drug effects , Male , Neovascularization, Physiologic/drug effects , Rats, Sprague-Dawley , Signal Transduction , Skin/injuries , Skin/metabolism , Skin/pathology , Skin Ulcer/metabolism , Skin Ulcer/pathology , Streptozocin , Wounds and Injuries/metabolism , Wounds and Injuries/pathology
19.
Eur J Med Chem ; 196: 112317, 2020 Jun 15.
Article in English | MEDLINE | ID: mdl-32311606

ABSTRACT

The emergence of antibiotic-resistant Mycobacterium Tuberculosis (Mtb) infections compels new treatment strategies, of which targeting trans-translation is promising. During the trans-translation process, the ribosomal protein S1 (RpsA) plays a key role, and the Ala438 mutant is related to pyrazinamide (PZA) resistance, which shows its effects after being hydrolysed to pyrazinoic acid (POA). In this study, based on the structure of the RpsA C-terminal domain (RpsA-CTD) and POA complex, new compounds were designed. After being synthesized, the compounds were tested in vitro with saturation transfer difference (STD), fluorescence quenching titration (FQT) and chemical shift perturbation (CSP) experiments. Finally, six of the 17 new compounds have high affinity for both RpsA-CTD and its Ala438 deletion mutant. The active compounds provide new choices for targeting trans-translation in Mtb, and the analysis of the structure-activity relationships will be helpful for further structural modifications based on derivatives of 2-((hypoxanthine-2-yl)thio)acetic acid and 2-((5-hydroxylflavone-7-yl)oxy)acetamide.


Subject(s)
Acetamides/pharmacology , Anti-Bacterial Agents/pharmacology , Hypoxanthine/pharmacology , Mycobacterium tuberculosis/drug effects , Ribosomal Proteins/antagonists & inhibitors , Tuberculosis, Multidrug-Resistant/drug therapy , Acetamides/chemical synthesis , Acetamides/chemistry , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Drug Discovery , Hypoxanthine/chemical synthesis , Hypoxanthine/chemistry , Microbial Sensitivity Tests , Molecular Docking Simulation , Molecular Structure , Ribosomal Proteins/metabolism , Tuberculosis, Multidrug-Resistant/metabolism
20.
Aging (Albany NY) ; 12(4): 3626-3646, 2020 02 17.
Article in English | MEDLINE | ID: mdl-32074082

ABSTRACT

Cellular senescence is a physiological process reacting to stimuli, in which cells enter a state of irreversible growth arrest in response to adverse consequences associated with metabolic disorders. Molecular mechanisms underlying the progression of cellular senescence remain unclear. Here, we established a replicative senescence model of human umbilical vein endothelial cells (HUVEC) from passage 3 (P3) to 18 (P18), and performed biochemical characterizations and NMR-based metabolomic analyses. The cellular senescence degree advanced as the cells were sequentially passaged in vitro, and cellular metabolic profiles were gradually altered. Totally, 8, 16, 21 and 19 significant metabolites were primarily changed in the P6, P10, P14 and P18 cells compared with the P3 cells, respectively. These metabolites were mainly involved in 14 significantly altered metabolic pathways. Furthermore, we observed taurine retarded oxidative damage resulting from senescence. In the case of energy deficiency, HUVECs metabolized neutral amino acids to replenish energy, thus increased glutamine, aspartate and asparagine at the early stages of cellular senescence but decreased them at the later stages. Our results indicate that cellular replicative senescence is closely associated with promoted oxidative stress, impaired energy metabolism and blocked protein synthesis. This work may provide mechanistic understanding of the progression of cellular senescence.


Subject(s)
Amino Acids/metabolism , Cellular Senescence/physiology , Human Umbilical Vein Endothelial Cells/metabolism , Metabolome , Humans , Magnetic Resonance Spectroscopy , Metabolomics , Oxidative Stress/physiology
SELECTION OF CITATIONS
SEARCH DETAIL