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










Publication year range
1.
Acta Pharmacol Sin ; 45(6): 1305-1315, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38383757

ABSTRACT

Histone deacetylase inhibitors (HDACis) are important drugs for cancer therapy, but the indistinct resistant mechanisms of solid tumor therapy greatly limit their clinical application. In this study we conducted HDACi-perturbated proteomics and phosphoproteomics analyses in HDACi-sensitive and -resistant cell lines using a tandem mass tag (TMT)-based quantitative proteomic strategy. We found that the ribosome biogenesis proteins MRTO4, PES1, WDR74 and NOP16 vital to tumorigenesis might regulate the tumor sensitivity to HDACi. By integrating HDACi-perturbated protein signature with previously reported proteomics and drug sensitivity data, we predicted and validated a series of drug combination pairs potentially to enhance the sensitivity of HDACi in diverse solid tumor. Functional phosphoproteomic analysis further identified the kinase PDK1 and ROCK as potential HDACi-resistant signatures. Overall, this study reveals the potential HDACi-resistant signatures and may provide promising drug combination strategies to attenuate the resistance of solid tumor to HDACi.


Subject(s)
Drug Resistance, Neoplasm , Histone Deacetylase Inhibitors , Neoplasms , Proteomics , Humans , Histone Deacetylase Inhibitors/pharmacology , Drug Resistance, Neoplasm/drug effects , Neoplasms/drug therapy , Neoplasms/metabolism , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use
2.
Nat Commun ; 15(1): 1465, 2024 Feb 17.
Article in English | MEDLINE | ID: mdl-38368419

ABSTRACT

Protein-modifying enzymes regulate the dynamics of myriad post-translational modification (PTM) substrates. Precise characterization of enzyme-substrate associations is essential for the molecular basis of cellular function and phenotype. Methods for direct capturing global substrates of protein-modifying enzymes in living cells are with many challenges, and yet largely unexplored. Here, we report a strategy to directly capture substrates of lysine-modifying enzymes via PTM-acceptor residue crosslinking in living cells, enabling global profiling of substrates of PTM-enzymes and validation of PTM-sites in a straightforward manner. By integrating enzymatic PTM-mechanisms, and genetically encoding residue-selective photo-crosslinker into PTM-enzymes, our strategy expands the substrate profiles of both bacterial and mammalian lysine acylation enzymes, including bacterial lysine acylases PatZ, YiaC, LplA, TmcA, and YjaB, as well as mammalian acyltransferases GCN5 and Tip60, leading to discovery of distinct yet functionally important substrates and acylation sites. The concept of direct capturing substrates of PTM-enzymes via residue crosslinking may extend to the other types of amino acid residues beyond lysine, which has the potential to facilitate the investigation of diverse types of PTMs and substrate-enzyme interactive proteomics.


Subject(s)
Lysine , Proteins , Animals , Lysine/metabolism , Proteins/metabolism , Acylation , Proteomics/methods , Protein Processing, Post-Translational , Mammals/metabolism
3.
J Am Chem Soc ; 145(46): 25283-25292, 2023 11 22.
Article in English | MEDLINE | ID: mdl-37857329

ABSTRACT

DNA-encoded chemical library (DEL) has been extensively used for lead compound discovery for decades in academia and industry. Incorporating an electrophile warhead into DNA-encoded compounds recently permitted the discovery of covalent ligands that selectively react with a particular cysteine residue. However, noncysteine residues remain underexplored as modification sites of covalent DELs. Herein, we report the design and utility of tyrosine-targeting DELs of 67 million compounds. Proteome-wide reactivity analysis of tyrosine-reactive sulfonyl fluoride (SF) covalent probes suggested three enzymes (phosphoglycerate mutase 1, glutathione s-transferase 1, and dipeptidyl peptidase 3) as models of tyrosine-targetable proteins. Enrichment with SF-functionalized DELs led to the identification of a series of tyrosine-targeting covalent inhibitors of the model enzymes. In-depth mechanistic investigation revealed their novel modes of action and reactive ligand-accessible hotspots of the enzymes. Our strategy of combining activity-based proteome profiling and covalent DEL enrichment (ABPP-CoDEL), which generated selective covalent binders against a variety of target proteins, illustrates the potential use of this methodology in further covalent drug discovery.


Subject(s)
Proteome , Tyrosine , Proteome/chemistry , Drug Discovery/methods , Small Molecule Libraries/pharmacology , Ligands , DNA
4.
Acta Pharmacol Sin ; 44(8): 1701-1711, 2023 Aug.
Article in English | MEDLINE | ID: mdl-36932232

ABSTRACT

Eriocalyxin B (EB), 17-hydroxy-jolkinolide B (HJB), parthenolide (PN), xanthatin (XT) and andrographolide (AG) are terpenoid natural products with a variety of promising antitumor activities, which commonly bear electrophilic groups (α,ß-unsaturated carbonyl groups and/or epoxides) capable of covalently modifying protein cysteine residues. However, their direct targets and underlying molecular mechanisms are still largely unclear, which limits the development of these compounds. In this study, we integrated activity-based protein profiling (ABPP) and quantitative proteomics approach to systematically characterize the covalent targets of these natural products and their involved cellular pathways. We first demonstrated the anti-proliferation activities of these five compounds in triple-negative breast cancer cell MDA-MB-231. Tandem mass tag (TMT)-based quantitative proteomics showed all five compounds commonly affected the ubiquitin mediated proteolysis pathways. ABPP platform identified the preferentially modified targets of EB and PN, two natural products with high anti-proliferation activity. Biochemical experiments showed that PN inhibited the cell proliferation through targeting ubiquitin carboxyl-terminal hydrolase 10 (USP10). Together, this study uncovered the covalently modified targets of these natural products and potential molecular mechanisms of their antitumor activities.


Subject(s)
Biological Products , Biological Products/pharmacology , Biological Products/chemistry , Proteomics , Proteins/metabolism , Ubiquitins
5.
Cell Death Differ ; 29(1): 1-13, 2022 01.
Article in English | MEDLINE | ID: mdl-34215846

ABSTRACT

SKP1-CUL1-F-box (SCF) ubiquitin ligases play fundamental roles in cellular functions. Typically, substrate phosphorylation is required for SCF recognition and subsequent degradation. However, phospho-dependent substrates remain largely unidentified. Here, using quantitative phoshoproteome approach, we performed a system-wide investigation of phospho-dependent SCF substrates. This strategy identified diverse phospho-dependent candidates. Biochemical verification revealed a mechanism by which SCFFBXO22 recognizes the motif XXPpSPXPXX as a conserved phosphodegron to target substrates for destruction. We further demonstrated BAG3, a HSP70 co-chaperone, is a bona fide substrate of SCFFBXO22. FBXO22 mediates BAG3 ubiquitination and degradation that requires ERK-dependent BAG3 phosphorylation at S377. FBXO22 depletion or expression of a stable BAG3 S377A mutant promotes tumor growth via defects in apoptosis and cell cycle progression in vitro and in vivo. In conclusion, our study identified broad phosphorylation-dependent SCF substrates and demonstrated a phosphodegron recognized by FBXO22 and a novel ERK-FBXO22-BAG3 axis involved in tumorigenesis.


Subject(s)
Carcinogenesis , F-Box Proteins , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Cell Transformation, Neoplastic , F-Box Proteins/genetics , F-Box Proteins/metabolism , Humans , Phosphorylation , Receptors, Cytoplasmic and Nuclear/metabolism , Ubiquitin/metabolism , Ubiquitination
6.
Acta Pharmacol Sin ; 41(9): 1246-1254, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32210356

ABSTRACT

Mitophagy is a degradative pathway that mediates the degradation of the entire mitochondria, and defects in this process are implicated in many diseases including cancer. In mammals, mitophagy is mediated by BNIP3L (also known as NIX) that is a dual regulator of mitochondrial turnover and programmed cell death pathways. Acute myeloid leukemia (AML) cells with deficiency of BNIP3L are more sensitive to mitochondria-targeting drugs. But small molecular inhibitors for BNIP3L are currently not available. Some immunomodulatory drugs (IMiDs) have been proved by FDA for hematologic malignancies, however, the underlining molecular mechanisms are still elusive, which hindered the applications of BNIP3L inhibition for AML treatment. In this study we carried out MS-based quantitative proteomics analysis to identify the potential neosubstrates of a novel thalidomide derivative CC-885 in A549 cells. In total, we quantified 5029 proteins with 36 downregulated in CRBN+/+ cell after CC-885 administration. Bioinformatic analysis showed that macromitophagy pathway was enriched in the negative pathway after CC-885 treatment. We further found that CC-885 caused both dose- and time-dependent degradation of BNIP3L in CRBN+/+, but not CRBN-/- cell. Thus, our data uncover a novel role of CC-885 in the regulation of mitophagy by targeting BNIP3L for CRL4CRBN E3 ligase-dependent ubiquitination and degradation, suggesting that CC-885 could be used as a selective BNIP3L degradator for the further investigation. Furthermore, we demonstrated that CC-885 could enhance AML cell sensitivity to the mitochondria-targeting drug rotenone, suggesting that combining CC-885 and mitochondria-targeting drugs may be a therapeutic strategy for AML patients.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Membrane Proteins/metabolism , Mitophagy/drug effects , Phenylurea Compounds/pharmacology , Proto-Oncogene Proteins/metabolism , Thalidomide/analogs & derivatives , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Cell Line, Tumor , Drug Synergism , HEK293 Cells , Humans , Proteasome Endopeptidase Complex/metabolism , Proteolysis/drug effects , Rotenone/pharmacology , Thalidomide/pharmacology , Ubiquitination/drug effects
7.
J Proteomics ; 210: 103545, 2020 01 06.
Article in English | MEDLINE | ID: mdl-31626998

ABSTRACT

Targeting specific ubiquitin E3 ligase for degradation of disease-driven protein has recently been an important concept for cancer therapy, as exemplified by the case of thalidomide for the treatment of multiple myeloma. E7070, an aryl sulfonamide drug, exhibited anticancer activity by targeting the E3 ligase receptor DCAF15, with RBM39 as the only known substrate. Exploration of additional substrates of E7070 would facilitate elucidation of its mechanism of actions. To this end, we used a strategy combing pSILAC method with two complementary digestion approaches (LysC-Trypsin and LysN-LysArgiNase) to accurately monitor the protein turnover and increase the depth of proteome profiling. Systematically, we showed that E7070 treatment changed turnover rates of 868 proteins (1.5 fold change and p-value <.05). Several proteins displayed accelerated turnover indicating they were potential new substrates of E7070, among which, pre-mRNA splicing factor 39 (PRPF39) had been reported to be overexpressed in certain cancers. We further demonstrated that PRPF39 was ubiquitinated and degraded by E7070 in a DCAF15-dependent manner, and represented a new bona fide substrate of E7070. The degradation of PRPF39 might also be contributed to the anticancer activity of E7070. SIGNIFICANCE: Identification of degraded substrates is difficult because protein abundance is a comprehensive result regulated by protein production and degradation at the same time. Pulsed SILAC (pSILAC), a method to measure protein turnover, would provide higher sensitivity than total protein quantification. In addition, some peptide sequences are not amenable to MS analysis after LysC-Trypsin digestion. LysN-LysargiNase, as a mirror protease combination of LysC-Trypsin, can be complementary for peptide identification with LysC-Trypsin. By combining pSILAC with two complementary digestion approaches (LysC-Trypsin and LysN-LysArgiNase), we systematically investigated E7070-dependent protein degradation. As a result, we found several potential degradation substrates of E7070 including PRPF39. Further, by exploiting a series of biological assays, we demonstrated that E7070 can lead to the ubiquitination and proteasomal degradation of PRPF39 by promoting the recruitment of PRPF39 to the CUL4-DCAF15 E3 ubiquitin ligase.


Subject(s)
Colonic Neoplasms/pathology , DNA Repair Enzymes/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , Proteolysis , Proteomics/methods , RNA Splicing Factors/metabolism , Sulfonamides/pharmacology , Ubiquitin-Protein Ligases/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Humans , Mass Spectrometry/methods , Ubiquitin/chemistry , Ubiquitin/metabolism
8.
Zootaxa ; 3985(4): 583-90, 2015 Jul 14.
Article in English | MEDLINE | ID: mdl-26250166

ABSTRACT

In this paper, a new species Sovia lii spec. nov. is described from Qinling-Daba Mountains in S. Gansu province and S. Shaanxi province, W. China. Variability of wing pattern, differences with its similar congeners and some biological information of this new taxon are introduced. External variability of Sovia lucasii, which is discovered from Shaanxi for the first time and is sympatric with the new species there, is illustrated and discussed. A brief analysis of the distributional pattern of the genus Sovia is provided. A key to the genus is given.


Subject(s)
Lepidoptera/classification , Animal Distribution , Animal Structures/anatomy & histology , Animal Structures/growth & development , Animals , Body Size , China , Ecosystem , Female , Lepidoptera/anatomy & histology , Lepidoptera/growth & development , Male , Organ Size
9.
Tumour Biol ; 33(4): 1085-93, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22351562

ABSTRACT

Although it has been known that basic fibroblast growth factor (bFGF) is involved in tumor progression, few studies addressed the role of bFGF in hematopoietic system malignancies including chronic myeloid leukemia (CML). An elevated level of bFGF was recently found in CML patients, and bFGF was considered to play an important role in stimulating the growth of leukemia cells. Suppression of the mitogenic activity of bFGF may contribute to CML therapy. We have previously obtained a novel bFGF-binding peptide (named P7) with strong inhibitory activity against bFGF-induced cell proliferation. In this study, we investigated the effects of P7 on the proliferation of K562 cells derived from CML. The results demonstrated that P7 inhibited bFGF-stimulated proliferation, arrested the cell cycle at the G0/G1 phase, repressed the activation of MAP kinase, reversed the effects of bFGF on cell membrane ultrastructure, and caused significant changes in the expression of proteins related to proliferation. Our results suggested that the bFGF-binding peptide may have a potential antitumor effect on CML from the point of view of targeting bFGF.


Subject(s)
Cell Proliferation/drug effects , Fibroblast Growth Factor 2/pharmacology , Peptide Fragments/pharmacology , Ribonucleotide Reductases/pharmacology , Adaptor Proteins, Signal Transducing/metabolism , Blotting, Western , Cell Cycle/drug effects , Cell Membrane/drug effects , Dose-Response Relationship, Drug , Drug Antagonism , Electrophoresis, Gel, Two-Dimensional , Enzyme Activation/drug effects , Humans , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mass Spectrometry , Microscopy, Atomic Force , Mitogen-Activated Protein Kinases/metabolism , Proteome/metabolism , Proteomics/methods , RNA-Binding Proteins/metabolism , Up-Regulation/drug effects
10.
Cancer Sci ; 101(5): 1212-8, 2010 May.
Article in English | MEDLINE | ID: mdl-20331620

ABSTRACT

Colon cancer is a leading cause of morbidity and mortality in Western countries. Basic fibroblast growth factor (bFGF) was up-regulated in patients with colon cancer and was considered as a potential therapeutic target. In this study, we first demonstrated that a novel bFGF-binding peptide (named P7) inhibited proliferation of several colon cancer cell lines including HT-29, LoVo, and Caco2 cells stimulated by bFGF. Further investigations with HT-29 cells indicated that P7 arrested the cell cycle at the G0/G1 phase of bFGF-stimulated cells, reduced the levels of phospho-Erk1/Erk2 induced by bFGF, and caused significant changes in the expression of proteins related to proliferation, cell cycle, and cancer. Our results suggested that the bFGF-binding peptide has a potential antitumor effect on colon cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Carrier Proteins/pharmacology , Colonic Neoplasms/drug therapy , Fibroblast Growth Factor 2/antagonists & inhibitors , Cell Cycle/drug effects , Colonic Neoplasms/pathology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Fibroblast Growth Factor 2/metabolism , HT29 Cells , Humans , Proteomics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
SELECTION OF CITATIONS
SEARCH DETAIL
...