Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Biol Chem ; 299(8): 105038, 2023 08.
Article in English | MEDLINE | ID: mdl-37442231

ABSTRACT

Covalent amino acid modification significantly expands protein functional capability in regulating biological processes. Tyrosine residues can undergo phosphorylation, sulfation, adenylation, halogenation, and nitration. These posttranslational modifications (PTMs) result from the actions of specific enzymes: tyrosine kinases, tyrosyl-protein sulfotransferase(s), adenylate transferase(s), oxidoreductases, peroxidases, and metal-heme containing proteins. Whereas phosphorylation, sulfation, and adenylation modify the hydroxyl group of tyrosine, tyrosine halogenation and nitration target the adjacent carbon residues. Because aberrant tyrosine nitration has been associated with human disorders and with animal models of disease, we have created an updated and curated database of 908 human nitrated proteins. We have also analyzed this new resource to provide insight into the role of tyrosine nitration in cancer biology, an area that has not previously been considered in detail. Unexpectedly, we have found that 879 of the 1971 known sites of tyrosine nitration are also sites of phosphorylation suggesting an extensive role for nitration in cell signaling. Overall, the review offers several forward-looking opportunities for future research and new perspectives for understanding the role of tyrosine nitration in cancer biology.


Subject(s)
Neoplasms , Proteins , Tyrosine , Animals , Humans , Phosphorylation , Proteins/metabolism , Signal Transduction , Tyrosine/metabolism
2.
Cell Rep ; 30(9): 3164-3170.e3, 2020 03 03.
Article in English | MEDLINE | ID: mdl-32130915

ABSTRACT

Aberrant hyperphosphorylation of the protein phosphatase 2A catalytic subunit (PP2Ac) at Tyr307 has been associated with aggressive disease and poor clinical outcome in multiple cancers. However, the study of reversible phosphorylation at this site has relied entirely upon the use of antibodies-most prominently, the clone E155. Here, we provide evidence that the E155 and F-8 phospho-Tyr307 antibodies cannot differentiate between phosphorylated and unphosphorylated forms of PP2Ac. The form of PP2Ac bound by these antibodies in H358 cells is unphosphorylated at the C-terminal tail. Furthermore, these antibodies are sensitive to additional protein modifications that occur near Tyr307, including Thr304 phosphorylation and Leu309 methylation, when these post-translational modifications are present. Thus, studies that used these antibodies to report PP2Ac hyperphosphorylation require reinterpretation, as these antibodies cannot be reliably used as readouts for a single PP2Ac post-translational modification (PTM) change.


Subject(s)
Antibodies/metabolism , Phosphotyrosine/metabolism , Protein Phosphatase 2/metabolism , Research , Alkaline Phosphatase/metabolism , Amino Acid Sequence , Cell Line , Cell Line, Tumor , Humans , Leucine/metabolism , Methylation , Mutation/genetics , Peptides/chemistry , Peptides/metabolism , Phosphorylation/drug effects , Protein Phosphatase 2/chemistry , Protein Phosphatase 2/genetics , Protein Processing, Post-Translational , Vanadates/pharmacology
3.
J Biol Chem ; 295(3): 757-770, 2020 01 17.
Article in English | MEDLINE | ID: mdl-31822503

ABSTRACT

The tumor suppressor protein phosphatase 2A (PP2A) is a serine/threonine phosphatase whose activity is inhibited in most human cancers. One of the best-characterized PP2A substrates is MYC proto-oncogene basic helix-loop-helix transcription factor (MYC), whose overexpression is commonly associated with aggressive forms of this disease. PP2A directly dephosphorylates MYC, resulting in its degradation. To explore the therapeutic potential of direct PP2A activation in a diverse set of MYC-driven cancers, here we used biochemical assays, recombinant cell lines, gene expression analyses, and immunohistochemistry to evaluate a series of first-in-class small-molecule activators of PP2A (SMAPs) in Burkitt lymphoma, KRAS-driven non-small cell lung cancer, and triple-negative breast cancer. In all tested models of MYC-driven cancer, the SMAP treatment rapidly and persistently inhibited MYC expression through proteasome-mediated degradation, inhibition of MYC transcriptional activity, decreased cancer cell proliferation, and tumor growth inhibition. Importantly, we generated a series of cell lines expressing PP2A-dependent phosphodegron variants of MYC and demonstrated that the antitumorigenic activity of SMAPs depends on MYC degradation. Collectively, the findings presented here indicate a pharmacologically tractable approach to drive MYC degradation by using SMAPs for the management of a broad range of MYC-driven cancers.


Subject(s)
Protein Phosphatase 2/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Tumor Suppressor Proteins/genetics , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Proteolysis/drug effects , Proto-Oncogene Mas , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/chemistry , Small Molecule Libraries/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology
4.
Biochim Biophys Acta Mol Cell Res ; 1866(1): 51-63, 2019 01.
Article in English | MEDLINE | ID: mdl-30401535

ABSTRACT

The serine/threonine phosphatase PP2A regulates a vast portion of the phosphoproteome including pathways involved in apoptosis, proliferation and DNA damage response and PP2A inactivation is a vital step in malignant transformation. Many groups have explored the therapeutic venue of combining PP2A reactivation with kinase inhibition to counteract the very changes in tumor suppressors and oncogenes that lead to cancer development. Conversely, inhibition of PP2A to complement chemotherapy and radiation-induced cancer cell death is also an area of active investigation. Here we review the studies that utilize PP2A targeted agents as combination therapy in cancer. A potential role for PP2A in tumor immunity is also highlighted.


Subject(s)
Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , Protein Phosphatase 2/physiology , Antineoplastic Agents/therapeutic use , Apoptosis/physiology , Cell Proliferation/physiology , DNA Repair/physiology , Gene Expression Regulation, Neoplastic/genetics , Humans , Molecular Targeted Therapy/methods , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Neoplasms/physiopathology , Neoplasms/therapy , Phosphoprotein Phosphatases/metabolism , Phosphoprotein Phosphatases/physiology , Protein Subunits/physiology , Signal Transduction/physiology
5.
Mol Cancer Ther ; 17(11): 2481-2489, 2018 11.
Article in English | MEDLINE | ID: mdl-30206106

ABSTRACT

The rat is the preferred model for toxicology studies, and it offers distinctive advantages over the mouse as a preclinical research model including larger sample size collection, lower rates of drug clearance, and relative ease of surgical manipulation. An immunodeficient rat would allow for larger tumor size development, prolonged dosing and drug efficacy studies, and preliminary toxicologic testing and pharmacokinetic/pharmacodynamic studies in the same model animal. Here, we created an immunodeficient rat with a functional deletion of the Recombination Activating Gene 2 (Rag2) gene, using genetically modified spermatogonial stem cells (SSC). We targeted the Rag2 gene in rat SSCs with TALENs and transplanted these Rag2-deficient SSCs into sterile recipients. Offspring were genotyped, and a founder with a 27 bp deletion mutation was identified and bred to homozygosity to produce the Sprague-Dawley Rag2 - Rag2 tm1Hera (SDR) knockout rat. We demonstrated that SDR rat lacks mature B and T cells. Furthermore, the SDR rat model was permissive to growth of human glioblastoma cell line subcutaneously resulting in successful growth of tumors. In addition, a human KRAS-mutant non-small cell lung cancer cell line (H358), a patient-derived high-grade serous ovarian cancer cell line (OV81), and a patient-derived recurrent endometrial cancer cell line (OV185) were transplanted subcutaneously to test the ability of the SDR rat to accommodate human xenografts from multiple tissue types. All human cancer cell lines showed efficient tumor uptake and growth kinetics indicating that the SDR rat is a viable host for a range of xenograft studies. Mol Cancer Ther; 17(11); 2481-9. ©2018 AACR.


Subject(s)
DNA-Binding Proteins/deficiency , Spermatogonia/cytology , Stem Cells/metabolism , Xenograft Model Antitumor Assays , Animals , B-Lymphocytes/cytology , Base Sequence , Biomarkers/metabolism , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Gene Knockout Techniques , Genome , Humans , Male , Rats, Sprague-Dawley , Subcutaneous Tissue/pathology , T-Lymphocytes/cytology
6.
Proteomics ; 17(22)2017 Nov.
Article in English | MEDLINE | ID: mdl-28961369

ABSTRACT

Activation of protein phosphatase 2A (PP2A) is a promising anticancer therapeutic strategy, as this tumor suppressor has the ability to coordinately downregulate multiple pathways involved in the regulation of cellular growth and proliferation. In order to understand the systems-level perturbations mediated by PP2A activation, we carried out mass spectrometry-based phosphoproteomic analysis of two KRAS mutated non-small cell lung cancer (NSCLC) cell lines (A549 and H358) treated with a novel small molecule activator of PP2A (SMAP). Overall, this permitted quantification of differential signaling across over 1600 phosphoproteins and 3000 phosphosites. Kinase activity assessment and pathway enrichment implicate collective downregulation of RAS and cell cycle kinases in the case of both cell lines upon PP2A activation. However, the effects on RAS-related signaling are attenuated for A549 compared to H358, while the effects on cell cycle-related kinases are noticeably more prominent in A549. Network-based analyses and validation experiments confirm these detailed differences in signaling. These studies reveal the power of phosphoproteomics studies, coupled to computational systems biology, to elucidate global patterns of phosphatase activation and understand the variations in response to PP2A activation across genetically similar NSCLC cell lines.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Phosphoproteins/metabolism , Protein Phosphatase 2/metabolism , Proteomics/methods , Small Molecule Libraries/pharmacology , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , Mass Spectrometry , Phosphorylation , Signal Transduction
7.
J Clin Invest ; 127(6): 2081-2090, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28504649

ABSTRACT

Targeted cancer therapies, which act on specific cancer-associated molecular targets, are predominantly inhibitors of oncogenic kinases. While these drugs have achieved some clinical success, the inactivation of kinase signaling via stimulation of endogenous phosphatases has received minimal attention as an alternative targeted approach. Here, we have demonstrated that activation of the tumor suppressor protein phosphatase 2A (PP2A), a negative regulator of multiple oncogenic signaling proteins, is a promising therapeutic approach for the treatment of cancers. Our group previously developed a series of orally bioavailable small molecule activators of PP2A, termed SMAPs. We now report that SMAP treatment inhibited the growth of KRAS-mutant lung cancers in mouse xenografts and transgenic models. Mechanistically, we found that SMAPs act by binding to the PP2A Aα scaffold subunit to drive conformational changes in PP2A. These results show that PP2A can be activated in cancer cells to inhibit proliferation. Our strategy of reactivating endogenous PP2A may be applicable to the treatment of other diseases and represents an advancement toward the development of small molecule activators of tumor suppressor proteins.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Activators/pharmacology , Protein Phosphatase 2/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Drug Resistance, Neoplasm , Enzyme Activation , Enzyme Activators/chemistry , Humans , Male , Mice, Inbred BALB C , Mice, Nude , Mice, Transgenic , Protein Binding , Protein Phosphatase 2/chemistry , Signal Transduction , Tumor Burden , Xenograft Model Antitumor Assays
8.
J Clin Invest ; 122(7): 2637-51, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22653055

ABSTRACT

EGFR activation is both a key molecular driver of disease progression and the target of a broad class of molecular agents designed to treat advanced cancer. Nevertheless, resistance develops through several mechanisms, including activation of AKT signaling. Though much is known about the specific molecular lesions conferring resistance to anti-EGFR-based therapies, additional molecular characterization of the downstream mediators of EGFR signaling may lead to the development of new classes of targeted molecular therapies to treat resistant disease. We identified a transcriptional network involving the tumor suppressors Krüppel-like factor 6 (KLF6) and forkhead box O1 (FOXO1) that negatively regulates activated EGFR signaling in both cell culture and in vivo models. Furthermore, the use of the FDA-approved drug trifluoperazine hydrochloride (TFP), which has been shown to inhibit FOXO1 nuclear export, restored sensitivity to AKT-driven erlotinib resistance through modulation of the KLF6/FOXO1 signaling cascade in both cell culture and xenograft models of lung adenocarcinoma. Combined, these findings define a novel transcriptional network regulating oncogenic EGFR signaling and identify a class of FDA-approved drugs as capable of restoring chemosensitivity to anti-EGFR-based therapy for the treatment of metastatic lung adenocarcinoma.


Subject(s)
Adenocarcinoma/metabolism , ErbB Receptors/metabolism , Forkhead Transcription Factors/metabolism , Kruppel-Like Transcription Factors/metabolism , Lung Neoplasms/metabolism , Proto-Oncogene Proteins/metabolism , Active Transport, Cell Nucleus/drug effects , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Drug Resistance, Neoplasm , Drug Synergism , Enzyme Activation , ErbB Receptors/antagonists & inhibitors , Erlotinib Hydrochloride , Female , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , Humans , Kruppel-Like Factor 6 , Kruppel-Like Transcription Factors/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Mutation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt/metabolism , Quinazolines/pharmacology , Quinazolines/therapeutic use , Real-Time Polymerase Chain Reaction , Signal Transduction , Transcription, Genetic , Trifluoperazine/pharmacology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...