Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Bioorg Med Chem ; 30: 115931, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33341501

ABSTRACT

The ubiquitin-proteasome system (UPS) plays an important role in maintaining protein homeostasis by degrading intracellular proteins. In the proteasome, poly-ubiquitinated proteins are deubiquitinated by three deubiquitinases (DUBs) associated with 19S regulatory particle before degradation via 20S core particle. Ubiquitin carboxyl-terminal hydrolase L5 (UCHL5) is one of three proteasome-associated DUBs that control the fate of ubiquitinated substrates implicated in cancer survival and progression. In this study, we have performed virtual screening of an FDA approved drug library with UCHL5 and discovered tiaprofenic acid (TA) as a potential binder. With molecular docking analysis and in-vitro DUB assay, we have designed, synthesized, and evaluated a series of TA derivatives for inhibition of UCHL5 activity. We demonstrate that one TA derivative, TAB2, acts as an inhibitor of UCHL5.


Subject(s)
Enzyme Inhibitors/pharmacology , Propionates/pharmacology , Ubiquitin Thiolesterase/antagonists & inhibitors , Cell Line, Tumor , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Molecular Docking Simulation , Molecular Structure , Propionates/chemical synthesis , Propionates/chemistry , Structure-Activity Relationship , Ubiquitin Thiolesterase/metabolism
2.
Eur J Pharmacol ; 889: 173609, 2020 Dec 15.
Article in English | MEDLINE | ID: mdl-33031796

ABSTRACT

Cancer cells rely heavily on molecular chaperones, such as heat shock protein 90 (HSP90), and their co-chaperones. The development of HSP90 inhibitors is an attractive therapeutic approach that has the potential to affect multiple hallmarks of cancer. Such approach is particularly needed for tumors that carry large mutational burdens, including cutaneous squamous cell carcinomas (cSCC). We previously identified sulfoxythiocarbamate S-4 as an HSP90 inhibitor. In this study, we investigated the mechanism(s) by which S-4 compromises the viability of human cSCC cells. S-4 inhibits HSP90 and causes depletion of its clients HER2, a tyrosine kinase oncoprotein, and Bcl-2, an anti-apoptotic protein. The decrease in Bcl-2 is accompanied by cytochrome c release from mitochondria into the cytoplasm, suggesting apoptosis. In the surviving cells, depletion of the HSP90 clients cyclin D and CDK4 by S-4 prevents phosphorylation of the retinoblastoma protein Rb and the release of transcription factor E2F, inhibiting G1-S cell cycle progression and cell division. These findings illustrate the comprehensive effectiveness of S-4 and encourage future development of compounds of this type for cancer prevention and treatment.


Subject(s)
Carcinoma, Squamous Cell/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , Skin Neoplasms/metabolism , Thiocarbamates/pharmacology , Animals , Carcinoma, Squamous Cell/pathology , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Isoxazoles/chemistry , Isoxazoles/pharmacology , Keratinocytes/drug effects , Keratinocytes/pathology , Mice , NIH 3T3 Cells , Resorcinols/chemistry , Resorcinols/pharmacology , Skin Neoplasms/pathology , Thiocarbamates/chemistry
3.
Chembiochem ; 21(6): 853-859, 2020 03 16.
Article in English | MEDLINE | ID: mdl-31560820

ABSTRACT

Protein S-glutathionylation is one of the important cysteine oxidation events that regulate various redox-mediated biological processes. Despite several existing methods, there are few proteomic approaches to identify and quantify specific cysteine residues susceptible to S-glutathionylation. We previously developed a clickable glutathione approach that labels intracellular glutathione with azido-Ala by using a mutant form of glutathione synthetase. In this study, we developed a quantification strategy with clickable glutathione by using isotopically labeled heavy and light derivatives of azido-Ala, which provides the relative quantification of glutathionylated peptides in mass spectrometry-based proteomic analysis. We applied isotopically labeled clickable glutathione to HL-1 cardiomyocytes, quantifying relative levels of 1398 glutathionylated peptides upon addition of hydrogen peroxide. Importantly, we highlight elevated levels of glutathionylation on sarcomere-associated muscle proteins while validating glutathionylation of two structural proteins, α-actinin and desmin. Our report provides a chemical proteomic strategy to quantify specific glutathionylated cysteines.


Subject(s)
Alanine/chemistry , Azides/chemistry , Glutathione/chemistry , Protein S/analysis , Click Chemistry , Cysteine/chemistry , Cysteine/metabolism , Isotope Labeling , Protein S/metabolism
4.
J Proteome Res ; 18(4): 1806-1818, 2019 04 05.
Article in English | MEDLINE | ID: mdl-30831029

ABSTRACT

Reactive oxygen species (ROS) are important signaling molecules, but their overproduction is associated with many cardiovascular diseases, including cardiomyopathy. ROS induce various oxidative modifications, among which glutathionylation is one of the significant protein oxidations that occur under oxidative stress. Despite previous efforts, direct and site-specific identification of glutathionylated proteins in cardiomyocytes has been limited. In this report, we used a clickable glutathione approach in a HL-1 mouse cardiomyocyte cell line under exposure to hydrogen peroxide, finding 1763 glutathionylated peptides with specific Cys modification sites, which include many muscle-specific proteins. Bioinformatic and cluster analyses found 125 glutathionylated proteins, whose mutations or dysfunctions are associated with cardiomyopathy, many of which include sarcomeric structural and contractile proteins, chaperone, and other signaling or regulatory proteins. We further provide functional implication of glutathionylation for several identified proteins, including CSRP3/MLP and complex I, II, and III, by analyzing glutathionylated sites in their structures. Our report establishes a chemoselective method for direct identification of glutathionylated proteins and provides potential target proteins whose glutathionylation may contribute to muscle diseases.


Subject(s)
Glutathione , Myocytes, Cardiac/metabolism , Proteins , Proteome , Animals , Cell Line , Glutathione/chemistry , Glutathione/metabolism , Mice , Protein Processing, Post-Translational , Proteins/analysis , Proteins/chemistry , Proteins/metabolism , Proteome/analysis , Proteome/chemistry , Proteome/metabolism , Proteomics , Reactive Oxygen Species/metabolism
5.
Nat Commun ; 9(1): 4341, 2018 10 18.
Article in English | MEDLINE | ID: mdl-30337525

ABSTRACT

Reactive oxygen species (ROS) contribute to the etiology of multiple muscle-related diseases. There is emerging evidence that cellular stress can lead to destabilization of sarcomeres, the contractile unit of muscle. However, it is incompletely understood how cellular stress induces structural destabilization of sarcomeres. Here we report that glutathionylation of SMYD2 contributes to a loss of myofibril integrity and degradation of sarcomeric proteins mediated by MMP-2 and calpain 1. We used a clickable glutathione approach in a cardiomyocyte cell line and found selective glutathionylation of SMYD2 at Cys13. Biochemical analysis demonstrated that SMYD2 upon oxidation or glutathionylation at Cys13 loses its interaction with Hsp90 and N2A, a domain of titin. Upon dissociation from SMYD2, N2A or titin is degraded by activated MMP-2, suggesting a protective role of SMYD2 in sarcomere stability. Taken together, our results support that SMYD2 glutathionylation is a novel molecular mechanism by which ROS contribute to sarcomere destabilization.


Subject(s)
Glutathione/metabolism , Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/metabolism , Proteolysis , Sarcomeres/metabolism , Amino Acid Sequence , Animals , Cell Line , Cell Survival , Cysteine/metabolism , HSP90 Heat-Shock Proteins/metabolism , Myofibrils/metabolism , Oxidation-Reduction , Rats , Reactive Oxygen Species/metabolism
6.
J Biol Chem ; 291(53): 27343-27353, 2016 12 30.
Article in English | MEDLINE | ID: mdl-27856637

ABSTRACT

Previously, we showed that levels of sphingosine-1 phosphate receptor 3 (S1PR3) are increased in a panel of cultured human lung adenocarcinoma cell lines, and that S1PR3-mediated signaling pathways regulate proliferation, soft agar growth, and invasion of human lung adenocarcinoma cells in vitro In the present study, we examine S1PR3 levels in human lung adenocarcinoma specimens. cDNA array and tumor microarray analysis shows that mRNA and protein levels of S1PR3 are significantly increased in human lung adenocarcinomas when compared with normal lung epithelial cells. Promoter analysis shows 16 candidate SMAD3 binding sites in the promoter region of S1PR3. ChIP indicates that TGF-ß treatment stimulates the binding of SMAD3 to the promoter region of S1PR3. Luciferase reporter assay demonstrates that SMAD3 transactivates S1PR3 promoter. TGF-ß stimulation or ectopic expression of TGF-ß up-regulates S1PR3 levels in vitro and ex vivo Pharmacologic inhibition of TGF-ß receptor or SMAD3 abrogates the TGF-ß-stimulated S1PR3 up-regulation. Moreover, S1PR3 knockdown dramatically inhibits tumor growth and lung metastasis, whereas ectopic expression of S1PR3 promotes the growth of human lung adenocarcinoma cells in animals. Pharmacological inhibition of S1PR3 profoundly inhibits the growth of lung carcinoma in mice. Our studies suggest that levels of S1PR3 are up-regulated in human lung adenocarcinomas, at least in part due to the TGF-ß/SMAD3 signaling axis. Furthermore, S1PR3 activity promotes the progression of human lung adenocarcinomas. Therefore, S1PR3 may represent a novel therapeutic target for the treatment of deadly lung adenocarcinomas.


Subject(s)
Adenocarcinoma/secondary , Lung Neoplasms/pathology , Receptors, Lysosphingolipid/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Animals , Apoptosis , Blotting, Western , Cell Proliferation , Cells, Cultured , Female , Humans , Lung/metabolism , Lung/pathology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Lysosphingolipid/genetics , Reverse Transcriptase Polymerase Chain Reaction , Smad3 Protein/genetics , Sphingosine-1-Phosphate Receptors , Transforming Growth Factor beta/genetics , Xenograft Model Antitumor Assays
7.
J Biol Chem ; 291(31): 16001-10, 2016 07 29.
Article in English | MEDLINE | ID: mdl-27246851

ABSTRACT

Excessive adipocyte lipolysis generates lipid mediators and triggers inflammation in adipose tissue. However, the specific roles of lipolysis-generated mediators in adipose inflammation remain to be elucidated. In the present study, cultured 3T3-L1 adipocytes were treated with isoproterenol to activate lipolysis and the fatty acyl lipidome of released lipids was determined by using LC-MS/MS. We observed that ß-adrenergic activation elevated levels of approximately fifty lipid species, including metabolites of cyclooxygenases, lipoxygenases, epoxygenases, and other sources. Moreover, we found that ß-adrenergic activation induced cyclooxygenase 2 (COX-2), not COX-1, expression in a manner that depended on activation of hormone-sensitive lipase (HSL) in cultured adipocytes and in the epididymal white adipose tissue (EWAT) of C57BL/6 mice. We found that lipolysis activates the JNK/NFκB signaling pathway and inhibition of the JNK/NFκB axis abrogated the lipolysis-stimulated COX-2 expression. In addition, pharmacological inhibition of COX-2 activity diminished levels of COX-2 metabolites during lipolytic activation. Inhibition of COX-2 abrogated the induction of CCL2/MCP-1 expression by ß-adrenergic activation and prevented recruitment of macrophage/monocyte to adipose tissue. Collectively, our data indicate that excessive adipocyte lipolysis activates the JNK/NFκB pathway leading to the up-regulation of COX-2 expression and recruitment of inflammatory macrophages.


Subject(s)
Adipocytes/enzymology , Cyclooxygenase 2/biosynthesis , Eicosanoids/biosynthesis , Lipolysis , Panniculitis/enzymology , Signal Transduction , 3T3-L1 Cells , Adipocytes/pathology , Animals , Chemokine CCL2/metabolism , Inflammation/enzymology , Inflammation/pathology , MAP Kinase Kinase 4/metabolism , Macrophages/metabolism , Macrophages/pathology , Mice , NF-kappa B/metabolism , Panniculitis/pathology , Sterol Esterase/metabolism
8.
Bioorg Med Chem Lett ; 26(5): 1452-6, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26832785

ABSTRACT

Small molecules that block the altered metabolism in cancer or increase the production of reactive oxygen species (ROS) are emerging as potential anti-cancer agents. Considering that various carbohydrates can be used for cellular energetics or protein N-glycosylation of which interruption can lead to cellular stress, we have synthesized and evaluated a library of N-aryl glycosides for induction of ROS and cytotoxicity in H1299 cancer cell line. Two N-aryl glycosides (K8 and H8) were identified that induce about 2-fold induction of ROS and cytotoxicity in H1299 cells. We further showed that the acetylated form of K8 (K8A) activates AMPK, and stabilizes p53 in HEK293 cells, and induce a higher cytotoxicity than 2-deoxy-d-glucose in H1299 cell line.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Glycosides/pharmacology , Reactive Oxygen Species/metabolism , Stress, Physiological/drug effects , Antineoplastic Agents/chemical synthesis , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Glycosides/chemical synthesis , Glycosides/chemistry , HEK293 Cells , Humans , Molecular Structure , Structure-Activity Relationship
9.
J Biol Chem ; 289(46): 32178-32185, 2014 Nov 14.
Article in English | MEDLINE | ID: mdl-25253697

ABSTRACT

Adipocyte lipolysis can increase the production of inflammatory cytokines such as interleukin-6 (IL-6) that promote insulin resistance. However, the mechanisms that link lipolysis with inflammation remain elusive. Acute activation of ß3-adrenergic receptors (ADRB3) triggers lipolysis and up-regulates production of IL-6 in adipocytes, and both of these effects are blocked by pharmacological inhibition of hormone-sensitive lipase. We report that stimulation of ADRB3 induces expression of sphingosine kinase 1 (SphK1) and increases sphingosine 1-phosphate production in adipocytes in a manner that also depends on hormone-sensitive lipase activity. Mechanistically, we found that adipose lipolysis-induced SphK1 up-regulation is mediated by the c-Jun N-terminal kinase (JNK)/activating protein-1 signaling pathway. Inhibition of SphK1 by sphingosine kinase inhibitor 2 diminished the ADRB3-induced IL-6 production both in vitro and in vivo. Induction of IL-6 by ADRB3 activation was suppressed by siRNA knockdown of Sphk1 in cultured adipocytes and was severely attenuated in Sphk1 null mice. Conversely, ectopic expression of SphK1 increased IL-6 expression in adipocytes. Collectively, these data demonstrate that SphK1 is a critical mediator in lipolysis-triggered inflammation in adipocytes.


Subject(s)
Adipocytes/cytology , Inflammation/metabolism , Interleukin-6/metabolism , Lipolysis , Phosphotransferases (Alcohol Group Acceptor)/metabolism , 3T3-L1 Cells , Adipocytes/metabolism , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , MAP Kinase Kinase 4/metabolism , Male , Mice , Mice, Inbred C57BL , RNA, Small Interfering/metabolism , Signal Transduction , Sphingolipids/chemistry , Tandem Mass Spectrometry
10.
J Am Chem Soc ; 136(33): 11566-9, 2014 Aug 20.
Article in English | MEDLINE | ID: mdl-25079194

ABSTRACT

Glutathionylation involves reversible protein cysteine modification that regulates the function of numerous proteins in response to redox stimuli, thereby altering cellular processes. Herein we developed a selective and versatile approach to identifying glutathionylation by using a mutant of glutathione synthetase (GS). GS wild-type catalyzes coupling of γGlu-Cys to Gly to form glutathione. We generated a GS mutant that catalyzes azido-Ala in place of Gly with high catalytic efficiency and selectivity. Transfection of this GS mutant (F152A/S151G) and incubation of azido-Ala in cells efficiently afford the azide-containing glutathione derivative, γGlu-Cys-azido-Ala. Upon H2O2 treatment, clickable glutathione allowed for selective and sensitive detection of glutathionylated proteins by Western blotting or fluorescence after click reaction with biotin-alkyne or rhodamine-alkyne. This approach affords the efficient metabolic tagging of intracellular glutathione with small clickable functionality, providing a versatile handle for characterizing glutathionylation.


Subject(s)
Click Chemistry , Glutathione Synthase/metabolism , Glutathione/biosynthesis , Biocatalysis , Glutathione/chemistry , Glutathione/metabolism , Glutathione Synthase/genetics , HEK293 Cells , Humans , Hydrogen Peroxide/chemistry , Hydrogen Peroxide/metabolism , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...