Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
J Am Heart Assoc ; 12(4): e028442, 2023 02 21.
Article in English | MEDLINE | ID: mdl-36752267

ABSTRACT

Background Viral myocarditis is characterized by leukocyte infiltration of the heart and cardiomyocyte death. We recently identified C-C chemokine ligand (CCL) 17 as a proinflammatory effector of C-C chemokine receptor 2-positive macrophages and dendritic cells that are recruited to the heart and contribute to adverse left ventricular remodeling following myocardial infarction and pressure overload. Methods and Results Mouse encephalomyocarditis virus was used to investigate the function of CCL17 in a viral myocarditis model. Ccl17Gfp reporter and knockout mice were used to identify the cell types that express CCL17 and delineate the functional importance of CCL17 in encephalomyocarditis virus clearance and myocardial inflammation. Cardiac CCL17 was expressed in C-C chemokine receptor 2-positive macrophages and dendritic cells following encephalomyocarditis virus infection. Colony-stimulating factor 2 (granulocyte-macrophage colony-stimulating factor) signaling was identified as a key regulator of CCL17 expression. Ccl17 deletion resulted in impaired encephalomyocarditis virus clearance, increased cardiomyocyte death, and higher mortality during infection early stage, and aggravated hypertrophy and fibrotic responses in infection long-term stage. An increased abundance of regulatory T cells was detected in the myocardium of injured Ccl17-deficient mice. Depletion of regulatory T cells in Ccl17-deficient mice abrogated the detrimental role of CCL17 deletion by restoring interferon signaling. Conclusions Collectively, these findings identify CCL17 as an important mediator of the host immune response during cardiac viral infection early stage and suggest that CCL17 targeted therapies should be avoided in acute viral myocarditis.


Subject(s)
Myocarditis , Virus Diseases , Mice , Animals , Myocarditis/genetics , Myocarditis/prevention & control , T-Lymphocytes, Regulatory , Macrophages/metabolism , Mice, Knockout , Receptors, Chemokine/metabolism , Chemokine CCL17/metabolism
2.
Nat Commun ; 13(1): 6531, 2022 11 01.
Article in English | MEDLINE | ID: mdl-36319634

ABSTRACT

DNA replication forks are tightly controlled by a large protein network consisting of well-known core regulators and many accessory factors which remain functionally undefined. In this study, we report previously unknown nuclear functions of the actin-binding factor profilin-1 (PFN1) in DNA replication, which occur in a context-dependent fashion and require its binding to poly-L-proline (PLP)-containing proteins instead of actin. In unperturbed cells, PFN1 increases DNA replication initiation and accelerates fork progression by binding and stimulating the PLP-containing nucleosome remodeler SNF2H. Under replication stress, PFN1/SNF2H increases fork stalling and functionally collaborates with fork reversal enzymes to enable the over-resection of unprotected forks. In addition, PFN1 binds and functionally attenuates the PLP-containing fork protector BODL1 to increase the resection of a subset of stressed forks. Accordingly, raising nuclear PFN1 level decreases genome stability and cell survival during replication stress. Thus, PFN1 is a multi-functional regulator of DNA replication with exploitable anticancer potential.


Subject(s)
Actins , Profilins , Humans , Actins/metabolism , DNA Helicases/metabolism , DNA Replication , Genomic Instability , Profilins/metabolism , Adenosine Triphosphatases/metabolism , Chromosomal Proteins, Non-Histone/metabolism
3.
Front Cell Dev Biol ; 9: 692269, 2021.
Article in English | MEDLINE | ID: mdl-34235154

ABSTRACT

The essential actin-binding factor profilin-1 (Pfn1) is a non-classical tumor suppressor with the abilities toboth inhibit cellular proliferation and augment chemotherapy-induced apoptosis. Besides actin, Pfn1 interacts with proteins harboring the poly-L-proline (PLP) motifs. Our recent work demonstrated that both nuclear localization and PLP-binding are required for tumor growth inhibition by Pfn1, and this is at least partially due to Pfn1 association with the PLP-containing ENL protein in the Super Elongation Complex (SEC) and the transcriptional inhibition of pro-cancer genes. In this paper, by identifying a phosphorylation event of Pfn1 at Ser71 capable of inhibiting its actin-binding and nuclear export, we provide in vitro and in vivo evidence that chemotherapy-induced apoptotic sensitization by Pfn1 requires its cytoplasmic localization and actin-binding. With regard to tumor growth inhibition byPfn1, our data indicate a requirement for dynamic actin association and dissociation rendered by reversible Ser71phosphorylation and dephosphorylation. Furthermore, genetic and pharmacological experiments showed that Ser71 of Pfn1 can be phosphorylated by protein kinase A (PKA). Taken together, our data provide novel mechanistic insights into the multifaceted anticancer activities of Pfn1 and how they are spatially-defined in the cell and differentially regulated by ligand-binding.

4.
Cell Rep ; 34(7): 108749, 2021 02 16.
Article in English | MEDLINE | ID: mdl-33596420

ABSTRACT

Aberrant expression of nuclear transporters and deregulated subcellular localization of their cargo proteins are emerging as drivers and therapeutic targets of cancer. Here, we present evidence that the nuclear exporter exportin-6 and its cargo profilin-1 constitute a functionally important and frequently deregulated axis in cancer. Exportin-6 upregulation occurs in numerous cancer types and is associated with poor patient survival. Reducing exportin-6 level in breast cancer cells triggers antitumor effects by accumulating nuclear profilin-1. Mechanistically, nuclear profilin-1 interacts with eleven-nineteen-leukemia protein (ENL) within the super elongation complex (SEC) and inhibits the ability of the SEC to drive transcription of numerous pro-cancer genes including MYC. XPO6 and MYC are positively correlated across diverse cancer types including breast cancer. Therapeutically, exportin-6 loss sensitizes breast cancer cells to the bromodomain and extra-terminal (BET) inhibitor JQ1. Thus, exportin-6 upregulation is a previously unrecognized cancer driver event by spatially inhibiting nuclear profilin-1 as a tumor suppressor.


Subject(s)
Karyopherins/metabolism , Neoplasms/metabolism , Profilins/antagonists & inhibitors , Profilins/metabolism , Animals , Cell Line, Tumor , Female , Heterografts , Humans , Karyopherins/genetics , MCF-7 Cells , Mice , Mice, Nude , Neoplasms/genetics , Profilins/genetics , Survival Analysis , Up-Regulation
5.
Cell Rep ; 31(10): 107745, 2020 06 09.
Article in English | MEDLINE | ID: mdl-32521270

ABSTRACT

Spatiotemporal protein reorganization at DNA damage sites induced by genotoxic chemotherapies is crucial for DNA damage response (DDR), which influences treatment response by directing cancer cell fate. This process is orchestrated by valosin-containing protein (VCP), an AAA+ ATPase that extracts polyubiquinated chromatin proteins and facilitates their turnover. However, because of the essential and pleiotropic effects of VCP in global proteostasis, it remains challenging practically to understand and target its DDR-specific functions. We describe a DNA-damage-induced phosphorylation event (Ser784), which selectively enhances chromatin-associated protein degradation mediated by VCP and is required for DNA repair, signaling, and cell survival. These functional effects of Ser784 phosphorylation on DDR correlate with a decrease in VCP association with chromatin, cofactors NPL4/UFD1, and polyubiquitinated substrates. Clinically, high phospho-Ser784-VCP levels are significantly associated with poor outcome among chemotherapy-treated breast cancer patients. Thus, Ser784 phosphorylation is a DDR-specific enhancer of VCP function and a potential predictive biomarker for chemotherapy treatments.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/therapy , DNA Damage/genetics , Valosin Containing Protein/metabolism , Female , Humans , Prognosis , Transfection
6.
Cancer Med ; 8(11): 5232-5241, 2019 09.
Article in English | MEDLINE | ID: mdl-31278855

ABSTRACT

BACKGROUND: Malignant peripheral nerve sheath tumors (MPNSTs) are aggressive sarcomas that arise most commonly in the setting of the Neurofibromatosis Type 1 (NF1) cancer predisposition syndrome. Despite aggressive multimodality therapy, outcomes are dismal and most patients die within 5 years of diagnosis. Prior genomic studies in our laboratory identified tyrosine kinase 2 (TYK2) as a frequently mutated gene in MPNST. Herein, we explored the function of TYK2 in MPNST pathogenesis. METHODS: Immunohistochemistry was utilized to examine expression of TYK2 in MPNSTs and other sarcomas. To establish a role for TYK2 in MPNST pathogenesis, murine and human TYK2 knockdown and knockout cells were established using shRNA and CRISPR/Cas9 systems, respectively. RESULTS: We have demonstrated that TYK2 was highly expressed in the majority of human MPNSTs examined. Additionally, we demonstrated that knockdown of Tyk2/TYK2 in murine and human MPNST cells significantly increased cell death in vitro. These effects were accompanied by a decrease in the levels of activated Stats and Bcl-2 as well as an increase in the levels of Cleaved Caspase-3. In addition, Tyk2-KD cells demonstrated impaired growth in subcutaneous and metastasis models in vivo. CONCLUSION: Taken together, these data illustrate the importance of TYK2 in MPNST pathogenesis and suggest that the TYK2 pathway may be a potential therapeutic target for these deadly cancers.


Subject(s)
Biomarkers, Tumor , Nerve Sheath Neoplasms/genetics , Nerve Sheath Neoplasms/pathology , TYK2 Kinase/genetics , Adult , Aged , Animals , Cell Death/genetics , Cell Line, Tumor , Cell Proliferation , Disease Progression , Disease Susceptibility , Female , Gene Expression Regulation, Neoplastic , Gene Targeting , Humans , Immunohistochemistry , Male , Mice , Middle Aged , Neoplasm Grading , Neoplasm Staging , Nerve Sheath Neoplasms/metabolism , Signal Transduction , TYK2 Kinase/metabolism , Young Adult
7.
Biomed Pharmacother ; 96: 551-562, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29032339

ABSTRACT

Major limitations of chalcones as clinical anticancer agents are water insolubility and poor bioavailability, which may be improved by a classic phosphate prodrug strategy that targets non-specific alkaline phosphatase (ALP) for releasing the parent drug in vivo. In this study, we found that BOC26P, a phosphate prodrug of chalcone OC26, exhibits excellent water solubility and improved plasma concentration in vivo by either i.v. or p.o. compared with the parent drug. In pace with decreased inhibitory activity of BOC26P against microtubule polymerization in vitro and in cells, the antiproliferative activity of BOC26P is attenuated in A549 and HLF cells. However, the antitumor effect of BOC26P increases in an A549 xenograft model as compared to the equimolar concentration of OC26, suggesting that complex tumor microenvironment would be another important influence factor to regulate the antitumor activity of BOC26Pin vivo. In conclusion, these observations showed that the traditional phosphate prodrug strategy would be a promising and easy method to increase water solubility and anticancer activity of chalcones for the clinical developments of anticancer agents.


Subject(s)
Antineoplastic Agents/chemical synthesis , Chalcones/chemical synthesis , Phosphates/chemical synthesis , Prodrugs/chemical synthesis , A549 Cells , Animals , Antineoplastic Agents/pharmacology , Cell Survival/drug effects , Cell Survival/physiology , Chalcones/pharmacology , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Phosphates/pharmacology , Prodrugs/pharmacology , Rats , Rats, Sprague-Dawley , Solubility , Water/chemistry , Xenograft Model Antitumor Assays/methods
8.
Biochem Pharmacol ; 137: 29-50, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28476333

ABSTRACT

The proteasomal 19S regulatory particle (RP) associated deubiquitinases (DUBs) have attracted much attention owing to their potential as a therapeutic target for cancer therapy. Identification of new entities against 19S RP associated DUBs and illustration of the underlying mechanisms is crucial for discovery of novel proteasome blockers. In this study, a series of 4-arylidene curcumin analogues were identified as potent proteasome inhibitor by preferentially blocking deubiquitinase function of proteasomal 19S RP with moderate 20S CP inhibition. The most active compound 33 exhibited a major inhibitory effect on 19S RP-associated ubiquitin-specific proteases 14, along with a minor effect on ubiquitin C-terminal hydrolase 5, which resulted in dysfunction of proteasome, and subsequently accumulated ubiquitinated proteins (such as IκB) in several cancer cells. Remarkably, though both 19S RP and 20S CP inhibition induced significantly endoplasmic reticulum stress and triggered caspase-12/9 pathway activation to promote cancer cell apoptosis, the 19S RP inhibition by 33 avoided slow onset time, Bcl-2 overexpression, and PERK-phosphorylation, which contribute to the deficiencies of clinical drug Bortezomib. These systematic studies provided insights in the development of novel proteasome inhibitors for cancer treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Curcumin/analogs & derivatives , Curcumin/pharmacology , Deubiquitinating Enzymes/antagonists & inhibitors , Proteasome Endopeptidase Complex , Proteasome Inhibitors/pharmacology , A549 Cells , Animals , Antineoplastic Agents/chemistry , CHO Cells , Cell Proliferation/drug effects , Cell Proliferation/physiology , Cricetinae , Cricetulus , Deubiquitinating Enzymes/metabolism , Dose-Response Relationship, Drug , Drug Delivery Systems/methods , Humans , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/chemistry
9.
Acta Pharmacol Sin ; 38(5): 638-650, 2017 May.
Article in English | MEDLINE | ID: mdl-28239158

ABSTRACT

We previously identified AG-690/11026014 (6014) as a novel poly(ADP-ribose) polymerase-1 (PARP-1) inhibitor that effectively prevented angiotensin II (Ang II)-induced cardiomyocyte hypertrophy. In the present study, we reported a new synthesis route for 6014, and investigated its protective effects on Ang II-induced cardiac remodeling and cardiac dysfunction and the underlying mechanisms in mice. We designed a new synthesis route to obtain a sufficient quantity of 6014 for this in vivo study. C57BL/6J mice were infused with Ang II and treated with 6014 (10, 30, 90 mg·kg-1·d-1, ig) for 4 weeks. Then two-dimensional echocardiography was performed to assess the cardiac function and structure. Histological changes of the hearts were examined with HE staining and Masson's trichrome staining. The protein expression was evaluated by Western blot, immunohistochemistry and immunofluorescence assays. The activities of sirtuin-1 (SIRT-1) and the content of NAD+ were detected with the corresponding test kits. Treatment with 6014 dose-dependently improved cardiac function, including LVEF, CO and SV and reversed the changes of cardiac structure in Ang II-infused mice: it significantly ameliorated Ang II-induced cardiac hypertrophy evidenced by attenuating the enlargement of cardiomyocytes, decreased HW/BW and LVW/BW, and decreased expression of hypertrophic markers ANF, BNP and ß-MHC; it also prevented Ang II-induced cardiac fibrosis, as implied by the decrease in excess accumulation of extracellular matrix (ECM) components collagen I, collagen III and FN. Further studies revealed that treatment with 6014 did not affect the expression levels of PARP-1, but dose-dependently inhibited the activity of PARP-1 and subsequently restored the activity of SIRT-1 in heart tissues due to the decreased consumption of NAD+ and attenuated Poly-ADP-ribosylation (PARylation) of SIRT-1. In conclusion, the novel PARP-1 inhibitor 6014 effectively protects mice against AngII-induced cardiac remodeling and improves cardiac function. Thus, 6014 might be a potential therapeutic agent for heart diseases..


Subject(s)
Cardiomegaly/therapy , Cardiotonic Agents/therapeutic use , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Thioglycolates/therapeutic use , Ventricular Remodeling/drug effects , Xanthines/therapeutic use , Angiotensin II/pharmacology , Animals , Cardiomegaly/chemically induced , Cardiotonic Agents/chemical synthesis , Fibrosis/chemically induced , Fibrosis/therapy , Male , Mice, Inbred C57BL , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Sirtuin 1/metabolism , Thioglycolates/chemical synthesis , Xanthines/chemical synthesis
10.
Eur J Pharmacol ; 796: 139-148, 2017 Feb 05.
Article in English | MEDLINE | ID: mdl-28024945

ABSTRACT

(E)-1-(3'-fluoro-[1,1'-biphenyl-3-yl)-3-(3-hydroxy-4-methoxyphenyl)prop-2-en-1-one) (MC37), a novel mono-carbonyl curcumin analog, was previously synthesized in our laboratory as a nuclear factor kappa B (NF-κB) inhibitor with excellent cytotoxicity against several cancer cell lines. In this study, our further investigations showed that the potent growth inhibitory activity of MC37 in human colorectal cancer cells was associated with the arrest of cell cycle progression and the induction of apoptosis. As a multi-targeted agent, MC37 inhibited the intracellular microtubule assembly, altered the expression of cyclin-dependent kinase 1 (CDK1), and ultimately induced G2/M cell cycle arrest. Moreover, MC37 collapsed the mitochondrial membrane potential (MMP), increased the Bax/Bcl-2 ratio, activated the caspase-9/3 cascade, and finally led to cancer cells apoptosis, suggesting that the mitochondrial-mediated apoptotic pathway was involved in MC37-induced apoptosis. In conclusion, these observations demonstrated that mono-carbonyl curcumin analogs would serve as multi-targeted lead for promising anti-colorectal cancer agent development.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Biphenyl Compounds/pharmacology , Colorectal Neoplasms/pathology , Curcumin/pharmacology , G2 Phase Cell Cycle Checkpoints/drug effects , M Phase Cell Cycle Checkpoints/drug effects , Mitochondria/drug effects , Propane/pharmacology , Caspase 3/metabolism , Caspase 9/metabolism , Cell Line, Tumor , Enzyme Activation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Membrane Potential, Mitochondrial/drug effects , Microtubules/drug effects , Microtubules/pathology , Mitochondria/metabolism , Mitochondria/pathology , Proto-Oncogene Proteins c-bcl-2/metabolism
11.
ACS Chem Biol ; 11(2): 425-34, 2016 Feb 19.
Article in English | MEDLINE | ID: mdl-26653078

ABSTRACT

A mitochondria-targeted approach was developed to increase the cellular bioactivities of thioredoxin reductase (TrxR) inhibitors. By being conjugated with a triphenylphosphine (TPP) motif to a previously found TrxR inhibitor 2a, the resulted compound TPP2a can target subcellular mitochondria and efficiently inhibit cellular TrxR, leading to remarkably increased cellular ROS level and mitochondrial apoptosis of HeLa cancer cells. The cellular bioactivities of TPP2a, including its cytotoxicity against a panel of cancer cell lines, dramatically elevated compared with its parental compound 2a. The selectively and covalently interaction of TPP2a with subcellular mitochondrial TrxR was validated by fluorescent microscopy. Moreover, a nonspecific signal quenching coupled strategy was proposed based on the environmentally sensitive fluorescence of TPP2a, which makes it possible to label TrxR by removing the nonspecific backgrounds caused by TPP2a under complex biosettings such as cellular lysates and living cells, implicating a potential of TPP2a for TrxR-specific labeling.


Subject(s)
Enzyme Inhibitors/pharmacology , Mitochondria/drug effects , Organophosphorus Compounds/pharmacology , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Enzyme Inhibitors/chemistry , HeLa Cells , Humans , Mitochondria/metabolism , Models, Molecular , Organophosphorus Compounds/chemistry , Reactive Oxygen Species/metabolism , Thioredoxin-Disulfide Reductase/metabolism
12.
Chem Commun (Camb) ; 51(69): 13400-3, 2015 Sep 07.
Article in English | MEDLINE | ID: mdl-26214302

ABSTRACT

A highly specific fluorescent probe (OC9) was discovered exhibiting tubulin-specific affinity fluorescence, which allowed selective labeling of cellular tubulin in microtubules. Moreover, distinct tubulin dynamics in various cellular bio-settings such as drug resistant or epithelial-mesenchymal transition (EMT) cancer cells were directly observed for the first time via OC9 staining.


Subject(s)
Aniline Compounds/chemistry , Chalcones/chemistry , Fluorescent Dyes/chemistry , Tubulin/chemistry , Cell Line, Tumor , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition , Humans , MCF-7 Cells , Microscopy, Fluorescence , Microtubules/chemistry , Microtubules/metabolism , Tubulin/metabolism
13.
Bioorg Med Chem Lett ; 25(6): 1240-3, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25682561

ABSTRACT

Fifteen taxanes (1-15) including a new taxane glucoside, 7ß,9α,10ß-triacetoxy-13α-hydroxy-5α-O-(ß-d-glucopyranosyl)taxa-4(20),11-diene (1), were isolated from the barks of Taxus wallichiana var. mairei. Compounds 1-15 representing three sub-types of 6/8/6-taxane were evaluated in vitro for anti-proliferative activity against a panel of parental and drug-resistant cancer cells. Potent compounds were found while several exhibited selective cytotoxicity. Especially, 3, 8, and 10 showed selective inhibition to breast carcinoma cell line MCF-7, while 13 selectively inhibited taxol resistant human ovarian carcinoma cell line A2780/TAX (IC50=0.19µM), being more potent than the clinical drugs taxol (IC50=4.4µM) and docetaxol (IC50=0.42µM), and less cytotoxic to mouse embryonic fibroblast cell line NIH-3T3, a cell line close to normal cell line. The possible P-glycoprotein evasion mechanism of 13 against A2780/TAX and the preliminary structure-activity relationships (SARs) of this group of compounds were also discussed.


Subject(s)
Taxoids/chemistry , Taxus/chemistry , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Humans , MCF-7 Cells , Mice , Molecular Conformation , NIH 3T3 Cells , Paclitaxel/pharmacology , Plant Bark/chemistry , Plant Bark/metabolism , Structure-Activity Relationship , Taxoids/isolation & purification , Taxoids/pharmacology , Taxus/metabolism
14.
J Med Chem ; 57(15): 6364-82, 2014 Aug 14.
Article in English | MEDLINE | ID: mdl-25061803

ABSTRACT

A series of new ortho-aryl chalcones have been designed and synthesized. Many of these compounds were found to exhibit significant antiproliferation activity toward a panel of cancer cell lines. Selected compounds show potent cytotoxicity against several drug resistant cell lines including paclitaxel (Taxol) resistant human ovarian carcinoma cells, vincristine resistant human ileocecum carcinoma cells, and doxorubicin resistant human breast carcinoma cells. Further investigation revealed that active analogues could inhibit the microtubule polymerization by binding to colchicine site and thus induce multipolar mitosis, G2/M phase arrest, and apoptosis of cancer cells. Furthermore, affinity-based fluorescence enhancement was observed during the binding of active compounds with tubulin, which greatly facilitated the determination of tubulin binding site of the compounds. Finally, selected compound 26 was found to exhibit obvious in vivo antitumor activity in A549 tumor xenografts model. Our systematic studies implied a new scaffold targeting tubulin and mitosis for novel antitumor drug discovery.


Subject(s)
Antineoplastic Agents/chemistry , Chalcones/chemistry , Mitosis/drug effects , Tubulin Modulators/chemistry , Aniline Compounds , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Binding Sites , Chalcones/chemical synthesis , Chalcones/pharmacology , Colchicine/metabolism , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Female , Fluorescence , G2 Phase Cell Cycle Checkpoints , Heterografts , Humans , Mice, Nude , Microtubules/drug effects , Microtubules/ultrastructure , Neoplasm Transplantation , Polymerization , Stereoisomerism , Structure-Activity Relationship , Tubulin/metabolism , Tubulin Modulators/chemical synthesis , Tubulin Modulators/pharmacology
15.
Chem Commun (Camb) ; 50(53): 6987-90, 2014 Jul 07.
Article in English | MEDLINE | ID: mdl-24844612

ABSTRACT

A simple and novel furanyl acryl conjugated coumarin fluorophore was designed as an effective thioredoxin reductase (TrxR)-responding fluorescent probe by an activity-guided approach. Basically, the α,ß-unsaturated ketone moiety in the probe structure could quench the fluorescence of the coumarin, but upon the covalent modification of TrxR, a significant fluorescence could be generated, which has been confirmed to be obviously selective over Trx, GSH, Cys and DTT.


Subject(s)
Coumarins/chemistry , Coumarins/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Fluorescent Dyes/chemistry , Thioredoxins/antagonists & inhibitors , Cell Line, Tumor/enzymology , Humans , Iodoacetamide/chemistry , Ketones/chemistry , Kinetics
16.
Eur J Med Chem ; 55: 346-57, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22889562

ABSTRACT

A series of new 4-arylidene curcumin analogues (4-arylidene-1,7-bisarylhepta-1,6-diene-3,5-diones) were synthesized and found to be potent antiproliferative agents against a panel of cancer cell lines at submicromolar to low micromolar concentrations by SRB assay. Their inhibitory abilities against NF-κB was evaluated by High Content Analysis (HCA) based immunofluorescence assay; and the Akt signalling inhibition was determined by fluorescence polarization assay and western blot respectively. The Structure-Activity Relationship was discussed. Our results revealed that 4-arylidene curcumin analogues may work in a multi-targets manner in cancer cell.


Subject(s)
Curcumin/chemical synthesis , Curcumin/pharmacology , NF-kappa B/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Chemistry Techniques, Synthetic , Curcumin/chemistry , Curcumin/metabolism , Dose-Response Relationship, Drug , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Molecular Docking Simulation , Phosphorylation/drug effects , Protein Conformation , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Transport , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...