Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Int J Biol Macromol ; 273(Pt 2): 132892, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38878921

ABSTRACT

TASK-3 generates a background K+ conductance which when inhibited by acidification depolarizes membrane potential and increases cell excitability. These channels sense pH by protonation of histidine residue H98, but recent evidence revealed that several other amino acid residues also contribute to TASK-3 pH sensitivity, suggesting that the pH sensitivity is determined by an intermolecular network. Here we use electrophysiology and molecular modeling to characterize the nature and requisite role(s) of multiple amino acids in pH sensing by TASK-3. Our results suggest that the pH sensor H98 and consequently pH sensitivity is influenced by remote amino acids that function as a hydrogen-bonding network to modulate ionic conductivity. Among the residues in the network, E30 and K79 are the most important for passing external signals near residue S31 to H98. The hydrogen-bond network plays a key role in selectivity or pH sensing in mTASK-3, and E30 and S31 in the network can modulate the conductive properties (E30) or reverse the pH sensitivity and selectivity of the channel (S31). Molecular dynamics simulations and pK1/2 calculation revealed that double mutants involving H98 + S31 primarily regulate the structure stability of the pore selectivity filter and pore loop regions, further strengthen the stability of the cradle suspension system, and alter the ionization state of E30 and K79, thereby preventing pore conformational change that normally occurs in response to varying extracellular pH. These results demonstrate that crucial residues in the hydrogen-bond network can remotely tune the pH sensing of mTASK-3 and may be a potential allosteric regulatory site for therapeutic molecule development.


Subject(s)
Hydrogen Bonding , Molecular Dynamics Simulation , Potassium Channels, Tandem Pore Domain , Hydrogen-Ion Concentration , Potassium Channels, Tandem Pore Domain/chemistry , Potassium Channels, Tandem Pore Domain/metabolism , Potassium Channels, Tandem Pore Domain/genetics , Humans , Mutation , Animals
2.
Signal Transduct Target Ther ; 9(1): 144, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38853183

ABSTRACT

Respiratory syncytial virus (RSV) is the major cause of bronchiolitis and pneumonia in young children and the elderly. There are currently no approved RSV-specific therapeutic small molecules available. Using high-throughput antiviral screening, we identified an oral drug, the prenylation inhibitor lonafarnib, which showed potent inhibition of the RSV fusion process. Lonafarnib exhibited antiviral activity against both the RSV A and B genotypes and showed low cytotoxicity in HEp-2 and human primary bronchial epithelial cells (HBEC). Time-of-addition and pseudovirus assays demonstrated that lonafarnib inhibits RSV entry, but has farnesyltransferase-independent antiviral efficacy. Cryo-electron microscopy revealed that lonafarnib binds to a triple-symmetric pocket within the central cavity of the RSV F metastable pre-fusion conformation. Mutants at the RSV F sites interacting with lonafarnib showed resistance to lonafarnib but remained fully sensitive to the neutralizing monoclonal antibody palivizumab. Furthermore, lonafarnib dose-dependently reduced the replication of RSV in BALB/c mice. Collectively, lonafarnib could be a potential fusion inhibitor for RSV infection.


Subject(s)
Pyridines , Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Human , Viral Fusion Proteins , Humans , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus Infections/genetics , Pyridines/pharmacology , Mice , Animals , Respiratory Syncytial Virus, Human/drug effects , Respiratory Syncytial Virus, Human/genetics , Viral Fusion Proteins/genetics , Viral Fusion Proteins/antagonists & inhibitors , Farnesyltranstransferase/antagonists & inhibitors , Farnesyltranstransferase/genetics , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Piperidines/pharmacology , Piperidines/chemistry , Mice, Inbred BALB C , Protein Conformation , Dibenzocycloheptenes
3.
Molecules ; 27(23)2022 Nov 28.
Article in English | MEDLINE | ID: mdl-36500386

ABSTRACT

TWIK-related acid-sensitive K+ (TASK) channels, including TASK-1, TASK-3, and TASK-5, are important members of the two-pore domain potassium (K2P) channel family. TASK-5 is not functionally expressed in the recombinant system. TASK channels are very sensitive to changes in extracellular pH and are active during all membrane potential periods. They are similar to other K2P channels in that they can create and use background-leaked potassium currents to stabilize resting membrane conductance and repolarize the action potential of excitable cells. TASK channels are expressed in both the nervous system and peripheral tissues, including excitable and non-excitable cells, and are widely engaged in pathophysiological phenomena, such as respiratory stimulation, pulmonary hypertension, arrhythmia, aldosterone secretion, cancers, anesthesia, neurological disorders, glucose homeostasis, and visual sensitivity. Therefore, they are important targets for innovative drug development. In this review, we emphasized the recent advances in our understanding of the biophysical properties, gating profiles, and biological roles of TASK channels. Given the different localization ranges and biologically relevant functions of TASK-1 and TASK-3 channels, the development of compounds that selectively target TASK-1 and TASK-3 channels is also summarized based on data reported in the literature.


Subject(s)
Potassium Channels, Tandem Pore Domain , Humans , Potassium Channels, Tandem Pore Domain/genetics , Potassium Channels, Tandem Pore Domain/chemistry , Membrane Potentials , Potassium/metabolism , Action Potentials , Arrhythmias, Cardiac
4.
iScience ; 25(7): 104591, 2022 Jul 15.
Article in English | MEDLINE | ID: mdl-35789855

ABSTRACT

Skp1 overexpression promotes tumor growth, whereas reduced Skp1 activity is also linked with genomic instability and neoplastic transformation. This highlights the need to gain better understanding of Skp1 biology in cancer settings. To this context, potent and cellularly active small-molecule Skp1 inhibitors may be of great value. Using a hypothesis-driven, structure-guided approach, we herein identify Z0933M as a potent Skp1 inhibitor with KD ∼0.054 µM. Z0933M occupies a hydrophobic hotspot (P1) - encompassing an aromatic cage of two phenylalanines (F101 and F139) - alongside C-terminal extension of Skp1 and, thus, hampers its ability to interact with F-box proteins, a prerequisite step to constitute intact and active SCF E3 ligase(s) complexes. In cellulo, Z0933M disrupted SCF E3 ligase(s) functioning, recapitulated previously reported effects of Skp1-reduced activity, and elicited cell death by a p53-dependent mechanism. We propose Z0933M as valuable tool for future efforts toward probing Skp1 cancer biology, with implications for cancer therapy.

5.
Contrast Media Mol Imaging ; 2021: 7081995, 2021.
Article in English | MEDLINE | ID: mdl-34949972

ABSTRACT

The aim of this study was to explore the application effect of intelligent incubator in neonatal care. We selected the period from January 1, 2018, to December 31, 2020, where there were 100 full-term and premature babies born in a hospital and transferred to the neonatal intensive care unit (NICU) within 1 hour after birth. Before the improved heat preservation, 100 full-term infants in the control group and 100 full-term infants in the intervention group of the intelligent warming box were formed into a full-term infant group for a comparative study. Statistics and comparison of the two groups of term infants and premature infants admitted to the hospital were carried out to assess body temperature and the changes in the incidence of each system. The research found that on comparison of admission body temperature between the control group and the intervention group, with the intervention group in the intelligent heating box, the incidence of hypothermia was significantly lower than that of the control group (95% vs. 37% of full-term infants; 98% vs. 49% of premature babies; there is a statistical significance (P < 0.05)). The intelligent heating box can reduce the fluctuation of the newborn's body temperature, keep the internal environment of newborns stable, and provide suitable conditions for the rapid growth of newborns, suitable for clinical promotion and application.


Subject(s)
Heating , Hypothermia , Humans , Infant , Infant, Newborn , Infant, Premature , Intensive Care Units, Neonatal
6.
Cancer Sci ; 112(8): 3218-3232, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34080260

ABSTRACT

Skp2 is overexpressed in multiple cancers and plays a critical role in tumor development through ubiquitin/proteasome-dependent degradation of its substrate proteins. Drugs targeting Skp2 have exhibited promising anticancer activity. Here, we identified a plant-derived Skp2 inhibitor, betulinic acid (BA), via high-throughput structure-based virtual screening of a phytochemical library. BA significantly inhibited the proliferation and migration of non-small cell lung cancer (NSCLC) through targeting Skp2-SCF E3 ligase both in vitro and in vivo. Mechanistically, BA binding to Skp2, especially forming H-bonds with residue Lys145, decreases its stability by disrupting Skp1-Skp2 interactions, thereby inhibiting the Skp2-SCF E3 ligase and promoting the accumulation of its substrates; that is, E-cadherin and p27. In both subcutaneous and orthotopic xenografts, BA significantly inhibited the proliferation and metastasis of NSCLC through targeting Skp2-SCF E3 ligase and upregulating p27 and E-cadherin protein levels. Taken together, BA can be considered a valuable therapeutic candidate to inhibit metastasis of NSCLC.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Enzyme Inhibitors/administration & dosage , Lung Neoplasms/drug therapy , Pentacyclic Triterpenes/administration & dosage , S-Phase Kinase-Associated Proteins/metabolism , A549 Cells , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Binding Sites , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Early Detection of Cancer , Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays , Humans , Lung Neoplasms/metabolism , Male , Mice , Pentacyclic Triterpenes/pharmacology , Protein Binding/drug effects , S-Phase Kinase-Associated Proteins/chemistry , Xenograft Model Antitumor Assays , Betulinic Acid
7.
Biochem Pharmacol ; 190: 114593, 2021 08.
Article in English | MEDLINE | ID: mdl-33964282

ABSTRACT

Aberrant activation of the Hedgehog (Hh) pathway is implicated in the pathogenesis and development of multiple cancers, especially Hh-driven medulloblastoma (MB). Smoothened (SMO) is a promising therapeutic target of the Hh pathway in clinical cancer treatment. However, SMO mutations frequently occur, which leads to drug resistance and tumor relapse. Novel inhibitors that target both the wild-type and mutant SMO are in high demand. In this study, we identified a novel Hh pathway inhibitor, pseudolaric acid B (PAB), which significantly inhibited the expression of Gli1 and its transcriptional target genes, such as cyclin D1 and N-myc, thus inhibiting the proliferation of DAOY and Ptch1+/- primary MB cells. Mechanistically, PAB can potentially bind to the extracellular entrance of the heptahelical transmembrane domain (TMD) of SMO, based on molecular docking and the BODIPY-cyclopamine binding assay. Further, PAB also efficiently blocked ciliogenesis, demonstrating the inhibitory effects of PAB on the Hh pathway at multiple levels. Thus, PAB may overcome drug-resistance induced by SMO mutations, which frequently occurs in clinical setting. PAB markedly suppressed tumor growth in the subcutaneous allografts of Ptch1+/- MB cells. Together, our results identified PAB as a potent Hh pathway inhibitor to treat Hh-dependent MB, especially cases resistant to SMO antagonists.


Subject(s)
Cerebellar Neoplasms/drug therapy , Diterpenes/pharmacology , Drugs, Chinese Herbal/therapeutic use , Hedgehog Proteins/antagonists & inhibitors , Medulloblastoma/drug therapy , Signal Transduction/drug effects , A549 Cells , Animals , Cerebellar Neoplasms/metabolism , Cerebellar Neoplasms/pathology , Diterpenes/chemistry , Diterpenes/therapeutic use , Dose-Response Relationship, Drug , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/pharmacology , HEK293 Cells , HeLa Cells , Hedgehog Proteins/chemistry , Hedgehog Proteins/metabolism , Humans , Male , Medulloblastoma/metabolism , Medulloblastoma/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Mice, Transgenic , NIH 3T3 Cells , Protein Structure, Secondary , Signal Transduction/physiology , Xenograft Model Antitumor Assays/methods
8.
Int J Oncol ; 56(5): 1274-1283, 2020 05.
Article in English | MEDLINE | ID: mdl-32319553

ABSTRACT

RNA­binding protein Musashi­2 (MSI2) serves as a regulator of numerous pivotal biological processes associated with cancer initiation, development and resistance to treatment, and may represent a promising drug target. However, whether MSI2 inhibition is of value in antitumor treatment remains to be determined. The present study demonstrated that MSI2 was upregulated in non­small cell lung cancer (NSCLC) and was inversely associated with the clinical outcome of the patients. Molecular docking analysis demonstrated that the small compound largazole binds to and may be a potential inhibitor of MSI2. Largazole markedly decreased the protein and mRNA levels of MSI2 and suppressed its downstream mammalian target of rapamycin signaling pathway. Largazole also inhibited the proliferation and induced apoptosis of NSCLC and chronic myeloid leukemia (CML) cells (including bone marrow mononuclear cells harvested from CML patients). These results indicate that MSI2 is an emerging therapeutic target for NSCLC and CML, and the MSI2 inhibitor largazole may hold promise as a treatment for these malignancies.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Depsipeptides/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Lung Neoplasms/genetics , RNA-Binding Proteins/genetics , Thiazoles/pharmacology , Adult , Aged , Animals , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Depsipeptides/chemistry , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Lung Neoplasms/drug therapy , Male , Mice , Middle Aged , Models, Molecular , Molecular Docking Simulation , Protein Conformation , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/chemistry , Thiazoles/chemistry , Up-Regulation/drug effects
9.
RSC Adv ; 10(62): 37871-37882, 2020 Oct 12.
Article in English | MEDLINE | ID: mdl-35515171

ABSTRACT

Biological remediation of toxic oxygen-containing anions such as nitrate that are common in the environment is of great significance. Therefore, it is necessary to understand the specific role of nitrate and nitrite reductase in the bioremediation process. Bacillus cereus LJ01, which was isolated from traditional Chinese soybean paste, effectively degraded nitrite (such as NaNO2) at 0-15 mmol L-1 in LB medium. Moreover, the nitrite-degrading active substance (ASDN) was isolated and purified from B. cereus LJ01. The nitrite-degrading activity of nitrite reductase (named LJ01-NiR) was 4004.89 U mg-1. The gene encoding the assimilation of nitrite reductase in B. cereus LJ01 was cloned and overexpressed in E. coli. The purified recombinant LJ01-NiR has a wide range of activities under temperature (20-60 °C), pH (6.5-8.0) and metal ions (Fe3+, Fe2+, Cu2+, Mn2+, and Al3+). Kinetic parameters of LJ01-NiR, including the values of K m and V max were 1.38 mM and 2.00 µmol g-1 min-1, respectively. The results showed that LJ01-NiR could degrade nitrite with or without an electron donor. In addition, sequence analysis revealed that LJ01-NiR was a ferredoxin-dependent nitrite reductase given the presence of conserved [Fe4-S4] cluster and heme-binding domain. The nitrite ion binds to the LJ01-NiR active site by forming three hydrogen bonds with the residues ASN72, ALA133 and ASN140. Due to its high nitrite-degrading activity, LJ01-NiR could potentially be used for environmental pollution treatment.

10.
Phytomedicine ; 66: 153111, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31790902

ABSTRACT

BACKGROUND: Hyperuricemia (HUA) is an important risk factor for renal diseases and contributes to renal fibrosis. It has been proved that phloretin has antioxidant and anti-inflammatory properties and could inhibit uric acid (UA) uptake in vitro. However, whether phloretin has a renal protective role in vivo remains unknown. PURPOSE: This study aims to evaluate the therapeutic effect of phloretin on HUA-induced renal injury in mice and to reveal its underlying mechanism. METHODS: Mice were induced hyperuricemic by oral gavage of adenine/potassium oxonate. The effects of phloretin on renal function, fibrosis, oxidative stress, inflammation, and UA metabolism in HUA mice were evaluated. The effect of phloretin on NLRP3 pathway was analyzed in human renal tubular cell lines (HK-2). RESULTS: HUA mice showed renal dysfunction with increased renal fibrosis, inflammation and mitochondrial stress. By contrast, phloretin reduced the level of serum blood urea nitrogen (BUN), urinary albumin to creatinine ratio (UACR), tubular necrosis, extracellular matrix (ECM) deposition and interstitial fibroblasts in HUA mice. The renal infiltration of inflammatory cells, cytokines such as NOD-like receptor family pyrin domain containing 3 (NLRP3) and interleukin-1ß (IL-1ß) release, mitochondrial reactive oxygen species (ROS) and morphological lesions in HUA mice also decreased. Furthermore, phloretin partly inhibited renal glucose transporter 9 (GLUT9) and promoted urinary UA excretion in HUA mice. In vitro, phloretin suppressed the NLPR3 pathway under LPS or UA stimulation in HK-2 cells. CONCLUSIONS: Phloretin could effectively attenuate UA-induced renal injury via co-inhibiting NLRP3 and UA reabsorption, and thus it might be a potential therapy to hyperuricemia-related renal diseases.


Subject(s)
Fibrosis/drug therapy , Hyperuricemia/drug therapy , Inflammation/drug therapy , Kidney Diseases/drug therapy , Phloretin/pharmacology , Uric Acid/metabolism , Animals , Cell Line , Cytokines/metabolism , Humans , Inflammasomes/drug effects , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Oxidative Stress , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
11.
ACS Med Chem Lett ; 10(6): 966-971, 2019 Jun 13.
Article in English | MEDLINE | ID: mdl-31223456

ABSTRACT

Malaria is caused by infection from the Plasmodium parasite and kills hundreds of thousands of people every year. Emergence of new drug resistant strains of Plasmodium demands identification of new drugs with novel chemotypes and mechanisms of action. As a follow up to our evaluation of 4-aryl-N-benzylpyrrolidine-3-carboxamides as novel pyrrolidine-based antimalarial agents, we describe herein the structure-activity relationships of the reversed amide homologues 2-aryl-N-(4-arylpyrrolidin-3-yl)acetamides. Unlike their carboxamide homologues, acetamide pyrrolidines do not require a third chiral center to be potent inhibitors of P. falciparum and have good pharmacokinetic properties and improved oral efficacy in a mouse model of malaria. Compound (-)-32a (CWHM-1552) has an in vitro IC50 of 51 nM in the P. falciparum 3D7 assay and an in vivo ED90 of <10 mg/kg/day and ED99 of 30 mg/kg/day in a murine P. chabaudi model. Remarkably, the absolute stereochemical preference for this acetamide series (3S,4R) is opposite of that determined for the homologous carboxamide series. Lead compounds for this class have modest affinities for the hERG channel and inhibit CYP 3A4. Additional optimization is needed in order to eliminate these undesired properties from this otherwise promising series of antimalarial compounds.

12.
Life Sci ; 227: 175-186, 2019 Jun 15.
Article in English | MEDLINE | ID: mdl-31026453

ABSTRACT

The tumor microenvironment comprises stromal and tumor cells which interact with each other through complex cross-talks that are mediated by a variety of growth factors, cytokines, and chemokines. The chemokine ligand 13 (CXCL13) and its chemokine receptor 5 (CXCR5) are among the key chemotactic factors which play crucial roles in deriving cancer cell biology. CXCL13/CXCR5 signaling axis makes pivotal contributions to the development and progression of several human cancers. In this review, we discuss how CXCL13/CXCR5 signaling modulates cancer cell ability to grow, proliferate, invade, and metastasize. Furthermore, we also discuss the preliminary evidence on context-dependent functioning of this axis within the tumor-immune microenvironment, thus, highlighting its potential dichotomy with respect to anticancer immunity and cancer immune-evasion mechanisms. At the end, we briefly shed light on the therapeutic potential or implications of targeting CXCL13/CXCR5 axis within the tumor microenvironment.


Subject(s)
Chemokine CXCL13/metabolism , Neoplasms/physiopathology , Receptors, CXCR5/metabolism , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cell Proliferation , Chemokine CXCL13/physiology , Disease Progression , Humans , Neoplasms/metabolism , Receptors, CXCR5/physiology , Signal Transduction , Tumor Microenvironment
13.
J Med Chem ; 62(7): 3503-3512, 2019 04 11.
Article in English | MEDLINE | ID: mdl-30856324

ABSTRACT

Identification of novel chemotypes with antimalarial efficacy is imperative to combat the rise of Plasmodium species resistant to current antimalarial drugs. We have used a hybrid target-phenotype approach to identify and evaluate novel chemotypes for malaria. In our search for drug-like aspartic protease inhibitors in publicly available phenotypic antimalarial databases, we identified GNF-Pf-4691, a 4-aryl- N-benzylpyrrolidine-3-carboxamide, as having a structure reminiscent of known inhibitors of aspartic proteases. Extensive profiling of the two terminal aryl rings revealed a structure-activity relationship in which relatively few substituents are tolerated at the benzylic position, but the 3-aryl position tolerates a range of hydrophobic groups and some heterocycles. Out of this effort, we identified (+)-54b (CWHM-1008) as a lead compound. 54b has EC50 values of 46 and 21 nM against drug-sensitive Plasmodium falciparum 3D7 and drug-resistant Dd2 strains, respectively. Furthermore, 54b has a long half-life in mice (4.4 h) and is orally efficacious in a mouse model of malaria (qd; ED99 ∼ 30 mg/kg/day). Thus, the 4-aryl- N-benzylpyrrolidine-3-carboxamide chemotype is a promising novel chemotype for malaria drug discovery.


Subject(s)
Antimalarials/pharmacology , Pyrrolidines/pharmacology , Administration, Oral , Animals , Antimalarials/administration & dosage , Antimalarials/chemistry , Biological Availability , Disease Models, Animal , Drug Evaluation, Preclinical , Malaria/drug therapy , Mice , Microsomes, Liver/drug effects , Pyrrolidines/administration & dosage , Pyrrolidines/chemistry , Structure-Activity Relationship
14.
Onco Targets Ther ; 12: 9639-9650, 2019.
Article in English | MEDLINE | ID: mdl-32009799

ABSTRACT

BACKGROUND: Membrane-associated guanylate kinase (MAGUK) with inverted orientation protein 1 (MAGI1) is a novel member of the MAGUK family with a vital role in tumor progression related to invasion and metastasis. However, the function of MAGI1 in glioma is currently unknown. We therefore analyzed the expression of MAGI1 protein in human glioma samples, glioma cell lines and glioma stem cells (GSCs), and explored its effects on glioma cell proliferation and apoptosis. METHODS: MAGI1 expression in glioma tissues was examined by Western blotting and real-time polymerase chain reaction and its relationships with clinical pathological features were analyzed. The effects of MAGI1 knockdown on the proliferation of glioma cell lines and GSCs were detected by CCK8 and colony-formation assays, and apoptosis was assessed by flow cytometry. We also investigated the effects of MAGI1 silencing on protein expression levels of epithelial-mesenchymal transition biomarkers, as well as ß-catenin, cyclin D1, PTEN and phospho-Akt by Western blotting. RESULTS: MAGI1 was significantly downregulated in glioma tissues and its expression was related to cancer progression. Silencing of MAGI1 in both glioma cell lines and GSCs enhanced proliferation and inhibited apoptosis. MAGI1 knockdown also significantly increased the expression levels of N-cadherin, vimentin, ß-catenin, cyclin D1 and phospho-Akt and reduced the expression of E-cadherin and PTEN. CONCLUSIONS: Our results indicated that MAGI1 might play a vital role in glioma progression and may represent a potential therapeutic target for the treatment of glioma.

15.
Eur J Med Chem ; 152: 542-559, 2018 May 25.
Article in English | MEDLINE | ID: mdl-29758518

ABSTRACT

The bromodomain and extra-terminal proteins (BET) have emerged as promising therapeutic targets for the treatment of castration-resistant prostate cancer (CRPC). We report the design, synthesis and evaluation of a new series of benzoxazinone-containing 3,5-dimethylisoxazole derivatives as selective BET inhibitors. One of the new compounds, (R)-12 (Y02234), binds to BRD4(1) with a Kd value of 110 nM and blocks bromodomain and acetyl lysine interactions with an IC50 value of 100 nM. It also exhibits selectivity for BET over non-BET bromodomain proteins and demonstrates reasonable anti-proliferation and colony formation inhibition effect in prostate cancer cell lines such as 22Rv1 and C4-2B. The BRD4 inhibitor (R)-12 also significantly suppresses the expression of ERG, Myc and AR target gene PSA at the mRNA level in prostate cancer cells. Treatment with (R)-12 significantly suppresses the tumor growth of prostate cancer (TGI = 70%) in a 22Rv1-derived xenograft model. These data suggest that compound (R)-12 is a promising lead compound for the development of a new class of therapeutics for the treatment of CRPC.


Subject(s)
Antineoplastic Agents/pharmacology , Benzoxazines/pharmacology , Isoxazoles/pharmacology , Prostatic Neoplasms, Castration-Resistant/drug therapy , Proteins/antagonists & inhibitors , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Benzoxazines/chemistry , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Isoxazoles/chemistry , Male , Molecular Structure , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Proteins/metabolism , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Tumor Cells, Cultured
16.
Org Lett ; 20(7): 1837-1840, 2018 04 06.
Article in English | MEDLINE | ID: mdl-29537285

ABSTRACT

A highly efficient synthesis of planar chiral pyrido[3,4- b] ferrocenes by a palladium-catalyzed enantioselective isocyanide insertion/desymmetric C(sp2)-H bond activation reaction was developed. Various planar chiral pyridoferrocenes were obtained in high yields with good to excellent enantioselectivity under mild conditions (up to 99% yield, 99% ee), enabled by a unique SPINOL-derived phosphoramidite ligand.

17.
J Med Chem ; 60(10): 4135-4146, 2017 05 25.
Article in English | MEDLINE | ID: mdl-28475316

ABSTRACT

In this report, we disclose the design and synthesis of a series of pentafluorosulfanyl (SF5) benzopyran derivatives as novel COX-2 inhibitors with improved pharmacokinetic and pharmacodynamic properties. The pentafluorosulfanyl compounds showed both potency and selectivity for COX-2 and demonstrated efficacy in several murine models of inflammation and pain. More interestingly, one of the compounds, R,S-3a, revealed exceptional efficacy in the adjuvant induced arthritis (AIA) model, achieving an ED50 as low as 0.094 mg/kg. In addition, the pharmacokinetics of compound R,S-3a in rat revealed a half-life in excess of 12 h and plasma drug concentrations well above its IC90 for up to 40 h. When R,S-3a was dosed just two times a week in the AIA model, efficacy was still maintained. Overall, drug R,S-3a and other analogues are suitable candidates that merit further investigation for the treatment of inflammation and pain as well as other diseases where COX-2 and PGE2 play a role in their etiology.


Subject(s)
Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/therapeutic use , Arthritis, Experimental/drug therapy , Benzopyrans/chemistry , Benzopyrans/therapeutic use , Cyclooxygenase 2 Inhibitors/chemistry , Cyclooxygenase 2 Inhibitors/therapeutic use , Inflammation/drug therapy , Animals , Anti-Inflammatory Agents/blood , Anti-Inflammatory Agents/pharmacology , Arthritis, Experimental/enzymology , Benzopyrans/blood , Benzopyrans/pharmacology , Cyclooxygenase 2 Inhibitors/blood , Cyclooxygenase 2 Inhibitors/pharmacology , Humans , Hyperalgesia/drug therapy , Hyperalgesia/enzymology , Inflammation/enzymology , Male , Mice , Models, Molecular , Rats, Inbred Lew , Rats, Sprague-Dawley
18.
Biochem Biophys Res Commun ; 489(3): 346-352, 2017 07 29.
Article in English | MEDLINE | ID: mdl-28549582

ABSTRACT

SPRY domain-containing SOCS box protein 2 (SPSB2) is a negative regulator of inducible nitric oxide synthase (iNOS) that modulates the lifetime of iNOS and thus the levels of nitric oxide (NO) production. Inhibitors that can disrupt the endogenous SPSB2-iNOS interaction and augment NO production have potential as novel antimicrobial and anticancer drugs. In this study, we have designed a cyclic peptide (cR8), containing an RGD motif and the SPSB2 binding motif (DINNNV). ITC and chemical shift perturbation showed that cR8 binds to the iNOS binding site on SPSB2 with a Kd of 671 nM, and saturation transfer difference NMR showed that cR8 binds to αvß3 integrin-expressing cells. Moreover, we determined the crystal structure of SPSB2 in complex with cR8, at a resolution of 1.34 Å. cR8 forms extensive hydrogen bonding with SPSB2 residues, but loss of an intramolecular hydrogen bond that is present in SPSB2-bound iNOS peptide may destabilize the bound conformation of cR8 and lead to a gentle reduction in SPSB2 binding affinity. These results serve as a useful basis for designing site-directed SPSB2 inhibitors in the future.


Subject(s)
Drug Design , Nitric Oxide Synthase Type II/metabolism , Oligopeptides/pharmacology , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology , Suppressor of Cytokine Signaling Proteins/chemistry , Suppressor of Cytokine Signaling Proteins/metabolism , Crystallization , Crystallography , Humans , Models, Molecular , Molecular Conformation , Oligopeptides/chemistry , Peptides, Cyclic/chemical synthesis , Protein Binding/drug effects , Structure-Activity Relationship
19.
Oncotarget ; 8(2): 2681-2693, 2017 Jan 10.
Article in English | MEDLINE | ID: mdl-27835873

ABSTRACT

The S phase kinase-associated protein 1 (Skp1), an adaptor protein of the Skp1-Cul1-F-box protein complex, binds the ubiquitin E3 ligase Skp2 and is critical to its biological functions. Targeting of Skp1 by a small compound 6-O-angeloylplenolin (6-OAP) results in dissociation and degradation of Skp2 and mitotic arrest of lung cancer cells. Here, by using a proteome microarray containing 16,368 proteins and a biotinylated 6-OAP, we identified 99 proteins that could bind 6-OAP, with Skp1 and STAT3 sitting at the central position of the 6-OAP interactome. 6-OAP formed hydrogen bonds with Ser611/Ser613/Arg609 at the SH2 domain of STAT3 and inhibited the constitutive and interleukin-6-induced phosphorylated STAT3 (pSTAT3), leading to inhibitory effects on lung cancer cells and suppression of Skp2 transcription. STAT3 was overexpressed in tumor samples compared to counterpart normal lung tissues and was inversely associated with prognosis of the patients. 6-OAP inhibited tumor growth in SCID mice intravenously injected with lung cancer cells, and downregulated both STAT3 and Skp2 in tumor samples. Given that 6-OAP is a Skp1 inhibitor, our data suggest that this compound may target Skp1 and STAT3 to suppress Skp2, augmenting its anti-lung cancer activity.


Subject(s)
Antineoplastic Agents/pharmacology , Oncogene Proteins/metabolism , Protein Kinase Inhibitors/pharmacology , Proteomics , S-Phase Kinase-Associated Proteins/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Gene Expression Regulation , Humans , Lactones/chemistry , Lactones/pharmacology , Mice , Models, Molecular , Molecular Conformation , Oncogene Proteins/antagonists & inhibitors , Oncogene Proteins/chemistry , Protein Binding , Protein Kinase Inhibitors/chemistry , Proteomics/methods , S-Phase Kinase-Associated Proteins/chemistry , S-Phase Kinase-Associated Proteins/genetics , S-Phase Kinase-Associated Proteins/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/chemistry , Sesquiterpenes/chemistry , Sesquiterpenes/pharmacology , Structure-Activity Relationship , Transcription, Genetic , Xenograft Model Antitumor Assays
20.
Acta Pharmacol Sin ; 37(11): 1516-1524, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27374490

ABSTRACT

AIM: Retinoic acid receptor-related orphan nuclear receptors (RORs) are orphan nuclear receptors that show constitutive activity in the absence of ligands. Among 3 subtypes of RORs, RORc is a promising therapeutic target for the treatment of Th17-mediated autoimmune diseases. Here, we report novel RORc inverse agonists discovered through structure-based drug design. METHODS: Based on the structure of compound 8, a previously described agonist of RORa, a series of 4-(4-(benzyloxy)phenyl)-3,4-dihydropyrimidin-2(1H)-one derivatives were designed and synthesized. The interaction between the compounds and RORc was detected at molecular level using AlphaScreen assay. The compounds were further examined in 293T cells transfected with RORc and luciferase reporter gene. Thermal stability shift assay was used to evaluate the effects of the compounds on protein stability. RESULTS: A total of 27 derivatives were designed and synthesized. Among them, the compound 22b was identified as the most potent RORc inverse agonist. Its IC50 values were 2.39 µmol/L in AlphaScreen assay, and 0.82 µmol/L in inhibition of the cell-based luciferase reporter activity. Furthermore, the compound 22b displayed a 120-fold selectivity for RORc over other nuclear receptors. Moreover, a molecular docking study showed that the structure-activity relationship was consistent with the binding mode of compound 22b in RORc. CONCLUSION: 4-(4-(Benzyloxy)phenyl)-3,4-dihydropyrimidin-2(1H)-one derivatives are promising candidates for the treatment of Th17-mediated autoimmune diseases, such as rheumatoid arthritis, psoriasis, and multiple sclerosis.


Subject(s)
Benzene Derivatives/chemistry , Nuclear Receptor Subfamily 1, Group F, Member 3/agonists , Pyrimidinones/chemistry , Autoimmune Diseases/drug therapy , Autoimmune Diseases/immunology , Benzene Derivatives/chemical synthesis , Benzene Derivatives/pharmacology , Drug Inverse Agonism , Genes, Reporter , HEK293 Cells , Humans , Luciferases, Renilla/genetics , Molecular Docking Simulation , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Pyrimidinones/chemical synthesis , Pyrimidinones/pharmacology , Structure-Activity Relationship , Th17 Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...