Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Mol Cell ; 53(4): 534-48, 2014 Feb 20.
Article in English | MEDLINE | ID: mdl-24486017

ABSTRACT

Mitochondrial pyruvate dehydrogenase complex (PDC) is crucial for glucose homeostasis in mammalian cells. The current understanding of PDC regulation involves inhibitory serine phosphorylation of pyruvate dehydrogenase (PDH) by PDH kinase (PDK), whereas dephosphorylation of PDH by PDH phosphatase (PDP) activates PDC. Here, we report that lysine acetylation of PDHA1 and PDP1 is common in epidermal growth factor (EGF)-stimulated cells and diverse human cancer cells. K321 acetylation inhibits PDHA1 by recruiting PDK1, and K202 acetylation inhibits PDP1 by dissociating its substrate PDHA1, both of which are important in promoting glycolysis in cancer cells and consequent tumor growth. Moreover, we identified mitochondrial ACAT1 and SIRT3 as the upstream acetyltransferase and deacetylase, respectively, of PDHA1 and PDP1, while knockdown of ACAT1 attenuates tumor growth. Furthermore, Y381 phosphorylation of PDP1 dissociates SIRT3 and recruits ACAT1 to PDC. Together, hierarchical, distinct posttranslational modifications act in concert to control molecular composition of PDC and contribute to the Warburg effect.


Subject(s)
Acetyl-CoA C-Acetyltransferase/metabolism , Pyruvate Dehydrogenase (Lipoamide)-Phosphatase/metabolism , Pyruvate Dehydrogenase (Lipoamide)/metabolism , Sirtuin 3/metabolism , Tyrosine/chemistry , Animals , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Glycolysis , Humans , Lysine/chemistry , Male , Mice , Mice, Nude , Mitochondria/metabolism , Neoplasm Transplantation , Neoplasms/metabolism , Phosphorylation
2.
Mol Cell ; 55(4): 552-65, 2014 Aug 21.
Article in English | MEDLINE | ID: mdl-25042803

ABSTRACT

Although the oxidative pentose phosphate pathway is important for tumor growth, how 6-phosphogluconate dehydrogenase (6PGD) in this pathway is upregulated in human cancers is unknown. We found that 6PGD is commonly activated in EGF-stimulated cells and human cancer cells by lysine acetylation. Acetylation at K76 and K294 of 6PGD promotes NADP(+) binding to 6PGD and formation of active 6PGD dimers, respectively. Moreover, we identified DLAT and ACAT2 as upstream acetyltransferases of K76 and K294, respectively, and HDAC4 as the deacetylase of both sites. Expressing acetyl-deficient mutants of 6PGD in cancer cells significantly attenuated cell proliferation and tumor growth. This is due in part to reduced levels of 6PGD products ribulose-5-phosphate and NADPH, which led to reduced RNA and lipid biosynthesis as well as elevated ROS. Furthermore, 6PGD activity is upregulated with increased lysine acetylation in primary leukemia cells from human patients, providing mechanistic insights into 6PGD upregulation in cancer cells.


Subject(s)
Acetyl-CoA C-Acetyltransferase/metabolism , Dihydrolipoyllysine-Residue Acetyltransferase/metabolism , Histone Deacetylases/metabolism , Leukemia/pathology , Lung Neoplasms/pathology , Lysine/metabolism , Phosphogluconate Dehydrogenase/metabolism , Acetylation , Animals , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Leukemia/metabolism , Lung Neoplasms/metabolism , Mice , NADP/metabolism , Neoplasms, Experimental , Protein Binding/physiology , Protein Multimerization
3.
Mol Cell ; 44(6): 864-77, 2011 Dec 23.
Article in English | MEDLINE | ID: mdl-22195962

ABSTRACT

Many tumor cells rely on aerobic glycolysis instead of oxidative phosphorylation for their continued proliferation and survival. Myc and HIF-1 are believed to promote such a metabolic switch by, in part, upregulating gene expression of pyruvate dehydrogenase (PDH) kinase 1 (PDHK1), which phosphorylates and inactivates mitochondrial PDH and consequently pyruvate dehydrogenase complex (PDC). Here we report that tyrosine phosphorylation enhances PDHK1 kinase activity by promoting ATP and PDC binding. Functional PDC can form in mitochondria outside of the matrix in some cancer cells and PDHK1 is commonly tyrosine phosphorylated in human cancers by diverse oncogenic tyrosine kinases localized to different mitochondrial compartments. Expression of phosphorylation-deficient, catalytic hypomorph PDHK1 mutants in cancer cells leads to decreased cell proliferation under hypoxia and increased oxidative phosphorylation with enhanced mitochondrial utilization of pyruvate and reduced tumor growth in xenograft nude mice. Together, tyrosine phosphorylation activates PDHK1 to promote the Warburg effect and tumor growth.


Subject(s)
Mitochondria/enzymology , Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , Tyrosine/metabolism , Animals , Female , Mice , Mice, Nude , Mitochondria/metabolism , Neoplasm Transplantation , Neoplasms/pathology , Phosphorylation , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Transplantation, Heterologous
4.
J Biol Chem ; 289(38): 26533-26541, 2014 Sep 19.
Article in English | MEDLINE | ID: mdl-25104357

ABSTRACT

The mitochondrial pyruvate dehydrogenase complex (PDC) plays a crucial role in regulation of glucose homoeostasis in mammalian cells. PDC flux depends on catalytic activity of the most important enzyme component pyruvate dehydrogenase (PDH). PDH kinase inactivates PDC by phosphorylating PDH at specific serine residues, including Ser-293, whereas dephosphorylation of PDH by PDH phosphatase restores PDC activity. The current understanding suggests that Ser-293 phosphorylation of PDH impedes active site accessibility to its substrate pyruvate. Here, we report that phosphorylation of a tyrosine residue Tyr-301 also inhibits PDH α 1 (PDHA1) by blocking pyruvate binding through a novel mechanism in addition to Ser-293 phosphorylation. In addition, we found that multiple oncogenic tyrosine kinases directly phosphorylate PDHA1 at Tyr-301, and Tyr-301 phosphorylation of PDHA1 is common in EGF-stimulated cells as well as diverse human cancer cells and primary leukemia cells from human patients. Moreover, expression of a phosphorylation-deficient PDHA1 Y301F mutant in cancer cells resulted in increased oxidative phosphorylation, decreased cell proliferation under hypoxia, and reduced tumor growth in mice. Together, our findings suggest that phosphorylation at distinct serine and tyrosine residues inhibits PDHA1 through distinct mechanisms to impact active site accessibility, which act in concert to regulate PDC activity and promote the Warburg effect.


Subject(s)
Protein Processing, Post-Translational , Pyruvate Dehydrogenase (Lipoamide)/metabolism , 3T3 Cells , Amino Acid Substitution , Animals , Carbohydrate Metabolism , Catalytic Domain , Cell Hypoxia , Cell Line, Tumor , Cell Proliferation , Epidermal Growth Factor/physiology , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Oxidative Phosphorylation , Phosphorylation , Protein Binding , Pyruvate Dehydrogenase (Lipoamide)/chemistry , Pyruvate Dehydrogenase (Lipoamide)/genetics , Pyruvic Acid/chemistry , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Tumor Burden , Tyrosine/metabolism
5.
J Biol Chem ; 289(31): 21413-22, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24962578

ABSTRACT

Many cancer cells rely more on aerobic glycolysis (the Warburg effect) than mitochondrial oxidative phosphorylation and catabolize glucose at a high rate. Such a metabolic switch is suggested to be due in part to functional attenuation of mitochondria in cancer cells. However, how oncogenic signals attenuate mitochondrial function and promote the switch to glycolysis remains unclear. We previously reported that tyrosine phosphorylation activates and inhibits mitochondrial pyruvate dehydrogenase kinase (PDK) and phosphatase (PDP), respectively, leading to enhanced inhibitory serine phosphorylation of pyruvate dehydrogenase (PDH) and consequently inhibition of pyruvate dehydrogenase complex (PDC) in cancer cells. In particular, Tyr-381 phosphorylation of PDP1 dissociates deacetylase SIRT3 and recruits acetyltransferase ACAT1 to PDC, resulting in increased inhibitory lysine acetylation of PDHA1 and PDP1. Here we report that phosphorylation at another tyrosine residue, Tyr-94, inhibits PDP1 by reducing the binding ability of PDP1 to lipoic acid, which is covalently attached to the L2 domain of dihydrolipoyl acetyltransferase (E2) to recruit PDP1 to PDC. We found that multiple oncogenic tyrosine kinases directly phosphorylated PDP1 at Tyr-94, and Tyr-94 phosphorylation of PDP1 was common in diverse human cancer cells and primary leukemia cells from patients. Moreover, expression of a phosphorylation-deficient PDP1 Y94F mutant in cancer cells resulted in increased oxidative phosphorylation, decreased cell proliferation under hypoxia, and reduced tumor growth in mice. Together, our findings suggest that phosphorylation at different tyrosine residues inhibits PDP1 through independent mechanisms, which act in concert to regulate PDC activity and promote the Warburg effect.


Subject(s)
Cell Division , Neoplasms/pathology , Pyruvate Dehydrogenase (Lipoamide)-Phosphatase/antagonists & inhibitors , Tyrosine/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cell Line, Tumor , DNA Primers , Humans , Lactic Acid/metabolism , Molecular Sequence Data , Neoplasms/enzymology , Oxygen Consumption , Phosphorylation , Pyruvate Dehydrogenase (Lipoamide)-Phosphatase/chemistry , Pyruvate Dehydrogenase (Lipoamide)-Phosphatase/genetics , Receptor, Fibroblast Growth Factor, Type 1/physiology , Sequence Homology, Amino Acid
6.
Cancer Cell ; 12(3): 201-14, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17785202

ABSTRACT

To better understand the signaling properties of oncogenic FGFR3, we performed phospho-proteomics studies to identify potential downstream signaling effectors that are tyrosine phosphorylated in hematopoietic cells expressing constitutively activated leukemogenic FGFR3 mutants. We found that FGFR3 directly tyrosine phosphorylates the serine/threonine kinase p90RSK2 at Y529, which consequently regulates RSK2 activation by facilitating inactive ERK binding to RSK2 that is required for ERK-dependent phosphorylation and activation of RSK2. Moreover, inhibition of RSK2 by siRNA or a specific RSK inhibitor fmk effectively induced apoptosis in FGFR3-expressing human t(4;14)-positive myeloma cells. Our findings suggest that FGFR3 mediates hematopoietic transformation by activating RSK2 in a two-step fashion, promoting both the ERK-RSK2 interaction and subsequent phosphorylation of RSK2 by ERK.


Subject(s)
Cell Transformation, Neoplastic/metabolism , MAP Kinase Signaling System , Multiple Myeloma/enzymology , Receptor, Fibroblast Growth Factor, Type 3/physiology , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Apoptosis , Binding Sites , Cell Line, Tumor , Cell Proliferation , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Mitogen-Activated Protein Kinase Kinases/metabolism , Models, Biological , Multiple Myeloma/metabolism , Phosphorylation , RNA Interference , Ribosomal Protein S6 Kinases, 90-kDa/antagonists & inhibitors , Tyrosine/metabolism
7.
J Biol Chem ; 288(45): 32528-32538, 2013 Nov 08.
Article in English | MEDLINE | ID: mdl-24085294

ABSTRACT

Metastasis is the leading cause of death in patients with breast, lung, and head and neck cancers. However, the molecular mechanisms underlying metastases in these cancers remain unclear. We found that the p90 ribosomal S6 kinase 2 (RSK2)-cAMP response element-binding protein (CREB) pathway is commonly activated in diverse metastatic human cancer cells, leading to up-regulation of a CREB transcription target Fascin-1. We also observed that the protein expression patterns of RSK2 and Fascin-1 correlate in primary human tumor tissue samples from head and neck squamous cell carcinoma patients. Moreover, knockdown of RSK2 disrupts filopodia formation and bundling in highly invasive cancer cells, leading to attenuated cancer cell invasion in vitro and tumor metastasis in vivo, whereas expression of Fascin-1 significantly rescues these phenotypes. Furthermore, targeting RSK2 with the small molecule RSK inhibitor FMK-MEA effectively attenuated the invasive and metastatic potential of cancer cells in vitro and in vivo, respectively. Taken together, our findings for the first time link RSK2-CREB signaling to filopodia formation and bundling through the up-regulation of Fascin-1, providing a proinvasive and prometastatic advantage to human cancers. Therefore, protein effectors of the RSK2-CREB-Fascin-1 pathway represent promising biomarkers and therapeutic targets in the clinical prognosis and treatment of metastatic human cancers.


Subject(s)
Biomarkers, Tumor/metabolism , CREB-Binding Protein/metabolism , Carrier Proteins/biosynthesis , Gene Expression Regulation, Neoplastic , Microfilament Proteins/biosynthesis , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Signal Transduction , Animals , Biomarkers, Tumor/genetics , CREB-Binding Protein/genetics , Carrier Proteins/genetics , Cell Line, Tumor , Gene Knockdown Techniques , Humans , Mice , Mice, Nude , Microfilament Proteins/genetics , Neoplasm Metastasis , Neoplasm Proteins/genetics , Neoplasms/genetics , Neoplasms/pathology , Pseudopodia/genetics , Pseudopodia/metabolism , Pseudopodia/pathology , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Up-Regulation/genetics
8.
Cancer Cell ; 10(1): 65-75, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16843266

ABSTRACT

Tyrosine kinases are aberrantly activated in numerous malignancies, including acute myeloid leukemia (AML). To identify tyrosine kinases activated in AML, we developed a screening strategy that rapidly identifies tyrosine-phosphorylated proteins using mass spectrometry. This allowed the identification of an activating mutation (A572V) in the JAK3 pseudokinase domain in the acute megakaryoblastic leukemia (AMKL) cell line CMK. Subsequent analysis identified two additional JAK3 alleles, V722I and P132T, in AMKL patients. JAK3(A572V), JAK3(V722I), and JAK3(P132T) each transform Ba/F3 cells to factor-independent growth, and JAK3(A572V) confers features of megakaryoblastic leukemia in a murine model. These findings illustrate the biological importance of gain-of-function JAK3 mutations in leukemogenesis and demonstrate the utility of proteomic approaches to identifying clinically relevant mutations.


Subject(s)
Leukemia, Experimental/genetics , Leukemia, Megakaryoblastic, Acute/genetics , Protein-Tyrosine Kinases/genetics , Alleles , Animals , Apoptosis/drug effects , Benzamides , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Imatinib Mesylate , Janus Kinase 2 , Janus Kinase 3 , K562 Cells , Leukemia, Experimental/metabolism , Leukemia, Experimental/pathology , Leukemia, Megakaryoblastic, Acute/metabolism , Leukemia, Megakaryoblastic, Acute/pathology , Mice , Mice, Inbred C57BL , Models, Molecular , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Phosphorylation/drug effects , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Structure, Tertiary/genetics , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Pyrimidines/pharmacology , RNA, Small Interfering/genetics , TYK2 Kinase
9.
Blood ; 112(5): 1981-92, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18559972

ABSTRACT

Oncogenic tyrosine kinases, such as BCR-ABL, TEL-ABL, TEL-PDGFbetaR, and FLT3-ITD, play a major role in the development of hematopoietic malignancy. They activate many of the same signal transduction pathways. To identify the critical target genes required for transformation in hematopoietic cells, we used a comparative gene expression strategy in which selective small molecules were applied to 32Dcl3 cells that had been transformed to factor-independent growth by these respective oncogenic alleles. We identified inhibitor of DNA binding 1 (Id1), a gene involved in development, cell cycle, and tumorigenesis, as a common target of these oncogenic kinases. These findings were prospectively confirmed in cell lines and primary bone marrow cells engineered to express the respective tyrosine kinase alleles and were also confirmed in vivo in murine models of disease. Moreover, human AML cell lines Molm-14 and K562, which express the FLT3-ITD and BCR-ABL tyrosine kinases, respectively, showed high levels of Id1 expression. Antisense and siRNA based knockdown of Id1-inhibited growth of these cells associated with increased p27(Kip1) expression and increased sensitivity to Trail-induced apoptosis. These findings indicate that Id1 is an important target of constitutively activated tyrosine kinases and may be a therapeutic target for leukemias associated with oncogenic tyrosine kinases.


Subject(s)
Inhibitor of Differentiation Protein 1/metabolism , Leukemia/metabolism , Protein-Tyrosine Kinases/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis/genetics , Apoptosis/physiology , Benzamides , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Gene Expression/drug effects , HL-60 Cells , Humans , Imatinib Mesylate , Inhibitor of Differentiation Protein 1/antagonists & inhibitors , Inhibitor of Differentiation Protein 1/genetics , K562 Cells , Leukemia/drug therapy , Leukemia/etiology , Leukemia/genetics , Leukemia, Experimental/drug therapy , Leukemia, Experimental/etiology , Leukemia, Experimental/genetics , Leukemia, Experimental/metabolism , Mice , Oncogenes , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics , Pyrimidines/pharmacology , Quinazolines/pharmacology , RNA, Small Interfering/genetics , Signal Transduction
10.
Clin Cancer Res ; 15(9): 3023-8, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19366827

ABSTRACT

PURPOSE: Activating mutations within the tyrosine kinase domain of epidermal growth factor receptor (EGFR) are found in approximately 10% to 20% of non-small-cell lung cancer (NSCLC) patients and are associated with response to EGFR inhibitors. The most common NSCLC-associated EGFR mutations are deletions in exon 19 and L858R mutation in exon 21, together accounting for 90% of EGFR mutations. To develop a simple, sensitive, and reliable clinical assay for the identification of EGFR mutations in NSCLC patients, we generated mutation-specific rabbit monoclonal antibodies against each of these two most common EGFR mutations and aimed to evaluate the detection of EGFR mutations in NSCLC patients by immunohistochemistry. EXPERIMENTAL DESIGN: We tested mutation-specific antibodies by Western blot, immunofluorescence, and immunohistochemistry. In addition, we stained 40 EGFR genotyped NSCLC tumor samples by immunohistochemistry with these antibodies. Finally, with a panel of four antibodies, we screened a large set of NSCLC patient samples with unknown genotype and confirmed the immunohistochemistry results by DNA sequencing. RESULTS: These two antibodies specifically detect the corresponding mutant form of EGFR by Western blotting, immunofluorescence, and immunohistochemistry. Screening a panel of 340 paraffin-embedded NSCLC tumor samples with these antibodies showed that the sensitivity of the immunohistochemistry assay is 92%, with a specificity of 99% as compared with direct and mass spectrometry-based DNA sequencing. CONCLUSIONS: This simple assay for detection of EGFR mutations in diagnostic human tissues provides a rapid, sensitive, specific, and cost-effective method to identify lung cancer patients responsive to EGFR-based therapies.


Subject(s)
Antibodies, Monoclonal , Carcinoma, Non-Small-Cell Lung/genetics , ErbB Receptors/genetics , Lung Neoplasms/genetics , Mutation/immunology , Animals , Biological Assay , Blotting, Western , Carcinoma, Non-Small-Cell Lung/secondary , DNA Mutational Analysis , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Flow Cytometry , Humans , Immunoenzyme Techniques , Immunoglobulin G/immunology , Lung Neoplasms/pathology , Mice , Mice, Nude , Rabbits , Sensitivity and Specificity , Sequence Deletion , Transplantation, Heterologous , Tumor Cells, Cultured
11.
Proteomics ; 9(16): 3979-88, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19658100

ABSTRACT

The ZNF198-fibroblast growth factor receptor-1 (FGFR1) fusion kinase is a constitutively activated tyrosine kinase associated with a specific atypical myeloproliferative disease. The chimeric protein localizes to the cytoplasm, unlike the wild type FGFR1 receptor kinase, and presumably inappropriately phosphorylates specific targets as part of the oncogenic signaling cascade. Other than known targets of the FGFR1 kinase itself, few specific targets of ZNF198-FGFR1 have been identified. Using a genetically engineered HEK 293 cell system, we have identified proteins that are specifically phosphorylated in the presence of the fusion kinase using anti-phosphotyrosine immunoprecipitation and MS. Compared with 293 cells expressing exongenous wild type FGFR1, ZNF198-FGFR1 is associated with phosphorylation of several proteins including SSBP2, ABL, FLJ14235, CALM and TRIM4 proteins. The specificity of the phosphorylation events in the SSBP2 and ABL proteins, which have previously been implicated in leukemogenesis, was further confirmed independently using immunoprecipitation with protein-specific antibodies and Western blotting. The MS analysis also identified the phosphorylation events in the ZNF198 moiety in the chimeric protein that might be related to its function. These studies identify the intersection of several different leukemia-related pathways in the development of this myeloproliferative disorder and provide new insights into the substrates of FGFR1 under defined conditions.


Subject(s)
DNA-Binding Proteins/metabolism , Mass Spectrometry/methods , Myeloproliferative Disorders/enzymology , Myeloproliferative Disorders/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Transcription Factors/metabolism , Blotting, Western , Cell Line , DNA-Binding Proteins/genetics , Humans , Immunoprecipitation , Monomeric Clathrin Assembly Proteins/metabolism , Myeloproliferative Disorders/genetics , Phosphorylation , Receptor, Fibroblast Growth Factor, Type 1/genetics , Transcription Factors/genetics
12.
Leuk Res ; 30(9): 1097-104, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16464493

ABSTRACT

STAT5 is constitutively phosphorylated in leukemic cells in approximately 70% of acute myeloid leukemia (AML) patients. To identify kinase candidates potentially responsible for STAT5 phosphorylation, we used liquid chromatography-tandem mass spectrometry (LC-MS/MS) mass spectrometry to detect phosphoproteins in AML cell lines. We established TEL-ARG and BCR-ABL fusion proteins as the mechanism underlying STAT5 phosphorylation in HT-93 and KBM-3 cells, respectively. In addition, we identified a JAK2 pseudokinase domain mutation in HEL cells and using siRNA downregulation, established JAK2 as the kinase responsible for phosphorylating STAT5. This study illustrates the benefit of LC-MS/MS mass spectrometry and siRNA for the identification of novel targets and mutations.


Subject(s)
Carcinogens , Leukemia, Myeloid, Acute/genetics , Neoplasm Proteins/genetics , Carcinogens/metabolism , Cell Line, Tumor , Chromatography, Liquid , Humans , Leukemia, Myeloid, Acute/metabolism , Mass Spectrometry , Mutation , Neoplasm Proteins/metabolism , Phosphorylation , Protein Processing, Post-Translational/genetics , Proteomics
13.
Nat Cell Biol ; 17(11): 1484-96, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26479318

ABSTRACT

The oxidative pentose phosphate pathway (PPP) contributes to tumour growth, but the precise contribution of 6-phosphogluconate dehydrogenase (6PGD), the third enzyme in this pathway, to tumorigenesis remains unclear. We found that suppression of 6PGD decreased lipogenesis and RNA biosynthesis and elevated ROS levels in cancer cells, attenuating cell proliferation and tumour growth. 6PGD-mediated production of ribulose-5-phosphate (Ru-5-P) inhibits AMPK activation by disrupting the active LKB1 complex, thereby activating acetyl-CoA carboxylase 1 and lipogenesis. Ru-5-P and NADPH are thought to be precursors in RNA biosynthesis and lipogenesis, respectively; thus, our findings provide an additional link between the oxidative PPP and lipogenesis through Ru-5-P-dependent inhibition of LKB1-AMPK signalling. Moreover, we identified and developed 6PGD inhibitors, physcion and its derivative S3, that effectively inhibited 6PGD, cancer cell proliferation and tumour growth in nude mice xenografts without obvious toxicity, suggesting that 6PGD could be an anticancer target.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Lipogenesis , Neoplasms/metabolism , Pentose Phosphate Pathway , Phosphogluconate Dehydrogenase/metabolism , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinase Kinases , Humans , Neoplasms/pathology , Oxidative Stress , Ribulosephosphates/metabolism , Signal Transduction
14.
Mol Cell Biol ; 33(13): 2574-85, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23608533

ABSTRACT

How invasive and metastatic tumor cells evade anoikis induction remains unclear. We found that knockdown of RSK2 sensitizes diverse cancer cells to anoikis induction, which is mediated through phosphorylation targets including apoptosis signal-regulating kinase 1 (ASK1) and cyclic AMP (cAMP) response element-binding protein (CREB). We provide evidence to show that RSK2 inhibits ASK1 by phosphorylating S83, T1109, and T1326 through a novel mechanism in which phospho-T1109/T1326 inhibits ATP binding to ASK1, while phospho-S83 attenuates ASK1 substrate MKK6 binding. Moreover, the RSK2→CREB signaling pathway provides antianoikis protection by regulating gene expression of protein effectors that are involved in cell death regulation, including the antiapoptotic factor protein tyrosine kinase 6 (PTK6) and the proapoptotic factor inhibitor-of-growth protein 3 (ING3). PTK6 overexpression or ING3 knockdown in addition to ASK1 knockdown further rescued the increased sensitivity to anoikis induction in RSK2 knockdown cells. These data together suggest that RSK2 functions as a signal integrator to provide antianoikis protection to cancer cells in both transcription-independent and -dependent manners, in part by signaling through ASK1 and CREB, and contributes to cancer cell invasion and tumor metastasis.


Subject(s)
Anoikis/physiology , MAP Kinase Kinase Kinase 5/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Adenosine Triphosphate/metabolism , Cell Line, Tumor/pathology , Cyclic AMP Response Element-Binding Protein/metabolism , Gene Expression Regulation , Genes, Tumor Suppressor , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , MAP Kinase Kinase 6/metabolism , MAP Kinase Kinase Kinase 5/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Phosphorylation , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Signal Transduction/genetics , Transcription, Genetic , Tumor Suppressor Proteins
15.
Nat Commun ; 4: 1790, 2013.
Article in English | MEDLINE | ID: mdl-23653202

ABSTRACT

How oncogenic signalling coordinates glycolysis and anabolic biosynthesis in cancer cells remains unclear. We recently reported that the glycolytic enzyme phosphoglycerate mutase 1 (PGAM1) regulates anabolic biosynthesis by controlling intracellular levels of its substrate 3-phosphoglycerate and product 2-phosphoglycerate. Here we report a novel mechanism in which Y26 phosphorylation enhances PGAM1 activation through release of inhibitory E19 that blocks the active site, stabilising cofactor 2,3-bisphosphoglycerate binding and H11 phosphorylation. We also report the crystal structure of H11-phosphorylated PGAM1 and find that phospho-H11 activates PGAM1 at least in part by promoting substrate 3-phosphoglycerate binding. Moreover, Y26 phosphorylation of PGAM1 is common in human cancer cells and contributes to regulation of 3-phosphoglycerate and 2-phosphoglycerate levels, promoting cancer cell proliferation and tumour growth. As PGAM1 is a negative transcriptional target of TP53, and is therefore commonly upregulated in human cancers, these findings suggest that Y26 phosphorylation represents an additional acute mechanism underlying phosphoglycerate mutase 1 upregulation.


Subject(s)
Neoplasms/enzymology , Neoplasms/metabolism , Phosphoglycerate Mutase/chemistry , Phosphoglycerate Mutase/metabolism , Phosphotyrosine/metabolism , 2,3-Diphosphoglycerate/metabolism , Amino Acid Sequence , Animals , Cell Line, Tumor , Cell Proliferation , Enzyme Stability , Glyceric Acids/metabolism , Glycolysis , Histidine/metabolism , Humans , Mice , Models, Molecular , Molecular Sequence Data , Neoplasms/pathology , Phosphorylation
16.
Zhonghua Xue Ye Xue Za Zhi ; 33(3): 163-8, 2012 Mar.
Article in Zh | MEDLINE | ID: mdl-22781597

ABSTRACT

OBJECTIVE: To screen phosphopeptide specific for acute leukemia. METHODS: Mononuclear cells from bone marrow were collected from 16 newly diagnosed acute lymphoblastic leukemia (ALL) and 20 acute myeloid leukemia (AML) patients. Peptides were extracted and purified, analyzed by immunoprecipitation and liquid chromatography coupled with tandem mass spectrometry (LC-MS). RESULTS: (1) Non-receptor tyrosine kinase family members Fyn, Yes, Src widely expressed in acute leukemia; (2) Some phosphopeptides, including non-receptor tyrosine kinase family members Abl/iso1 and Abl, non-receptor Ser/Thr protein kinase family members Bcr, JNK2, JNK2 iso2, Adaptor/scaffold members Cas-L, Cbl, CrkL CENTD1 (Centaurin delta1) ZO2, transcriptor GFR-1 and phosphatase SHIP-2 were detected in Ph positive ALL, but not in other kinds of ALL. (3) Hck, Lyn and Fgr selectively expressed in AML (except AML-M(3)). CONCLUSION: Some phosphopeptides were specific for ALL and AML, and may be useful for diagnosis and therapy of acute leukemia.


Subject(s)
Leukemia, Myeloid, Acute/metabolism , Neoplasm Proteins/analysis , Phosphopeptides/analysis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Chromatography, High Pressure Liquid , Humans , Immunoprecipitation , Leukemia, Myeloid, Acute/genetics , Phosphorylation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proteomics , Tandem Mass Spectrometry
17.
Clin Cancer Res ; 18(16): 4449-57, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22661537

ABSTRACT

PURPOSE: To deepen our understanding of mutant ROS1 expression, localization, and frequency in non-small cell lung cancer (NSCLC), we developed a highly specific and sensitive immunohistochemistry (IHC)-based assay that is useful for the detection of wild-type and mutant ROS1. EXPERIMENTAL DESIGN: We analyzed 556 tumors with the ROS1 D4D6 rabbit monoclonal antibody IHC assay to assess ROS1 expression levels and localization. A subset of tumors was analyzed by FISH to determine the percentage of these tumors harboring ROS1 translocations. Using specific and sensitive IHC assays, we analyzed the expression of anaplastic lymphoma kinase (ALK), EGFR L858R, and EGFR E746-A750del mutations in a subset of lung tumors, including those expressing ROS1. RESULTS: In our NSCLC cohort of Chinese patients, we identified 9 (1.6%) tumors expressing ROS1 and 22 (4.0%) tumors expressing ALK. FISH identified tumors with ALK or ROS1 rearrangements, and IHC alone was capable of detecting all cases with ALK and ROS1 rearrangements. ROS1 fusion partners were determined by reverse transcriptase PCR identifying CD74-ROS1, SLC34A2-ROS1, and FIG-ROS1 fusions. Some of the ALK and ROS1 rearranged tumors may also harbor coexisting EGFR mutations. CONCLUSIONS: NSCLC tumors with ROS1 rearrangements are uncommon in the Chinese population and represent a distinct entity of carcinomas. The ROS1 IHC assay described here is a valuable tool for identifying patients expressing mutant ROS1 and could be routinely applied in clinical practice to detect lung cancers that may be responsive to targeted therapies.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Carrier Proteins/metabolism , Lung Neoplasms/metabolism , Membrane Proteins/metabolism , Oncogene Proteins, Fusion/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Adaptor Proteins, Signal Transducing , Anaplastic Lymphoma Kinase , Animals , Base Sequence , Carcinoma, Non-Small-Cell Lung/genetics , Carrier Proteins/genetics , Cell Line , Cell Proliferation , Gene Expression , Genes, erbB-1 , Genotype , Golgi Matrix Proteins , Humans , Lung Neoplasms/genetics , Membrane Proteins/genetics , Membrane Transport Proteins , Mice , Mutation , Oncogene Proteins, Fusion/genetics , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Transplantation, Heterologous
18.
Cancer Res ; 72(13): 3312-23, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22570254

ABSTRACT

Ovarian cancer is the leading cause of death from gynecologic cancer. Improvement in the clinical outcome of patients is likely to be achieved by the identification of molecular events that underlie the oncogenesis of ovarian cancer. Here we show that the anaplastic lymphoma kinase (ALK) is aberrantly activated in ovarian cancer. Using an unbiased and global phosphoproteomic approach, we profiled 69 Chinese primary ovarian tumor tissues and found ALK to be aberrantly expressed and phosphorylated in 4 tumors. Genetic characterization of these ALK-positive tumors indicated that full-length ALK expression in two serous carcinoma patients is consistent with ALK gene copy number gain, whereas a stromal sarcoma patient carries a novel transmembrane ALK fusion gene: FN1-ALK. Biochemical and functional analysis showed that both full-length ALK and FN1-ALK are oncogenic, and tumors expressing ALK or FN1-ALK are sensitive to ALK kinase inhibitors. Furthermore, immunohistochemical analysis of ovarian tumor tissue microarray detected aberrant ALK expression in 2% to 4% serous carcinoma patients. Our findings provide new insights into the pathogenesis of ovarian cancer and identify ALK as a potential therapeutic target in a subset of serous ovarian carcinoma and stromal sarcoma patients.


Subject(s)
Ovarian Neoplasms/drug therapy , Receptor Protein-Tyrosine Kinases/metabolism , Adolescent , Adult , Aged , Amino Acid Sequence , Anaplastic Lymphoma Kinase , Base Sequence , Blotting, Western , Cell Line, Tumor , Chromatography, Liquid , DNA Primers , Female , Humans , Middle Aged , Molecular Sequence Data , Phosphorylation , Receptor Protein-Tyrosine Kinases/drug effects , Receptor Protein-Tyrosine Kinases/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tandem Mass Spectrometry , Tissue Array Analysis
19.
Cancer Cell ; 22(5): 585-600, 2012 Nov 13.
Article in English | MEDLINE | ID: mdl-23153533

ABSTRACT

It is unclear how cancer cells coordinate glycolysis and biosynthesis to support rapidly growing tumors. We found that the glycolytic enzyme phosphoglycerate mutase 1 (PGAM1), commonly upregulated in human cancers due to loss of TP53, contributes to biosynthesis regulation in part by controlling intracellular levels of its substrate, 3-phosphoglycerate (3-PG), and product, 2-phosphoglycerate (2-PG). 3-PG binds to and inhibits 6-phosphogluconate dehydrogenase in the oxidative pentose phosphate pathway (PPP), while 2-PG activates 3-phosphoglycerate dehydrogenase to provide feedback control of 3-PG levels. Inhibition of PGAM1 by shRNA or a small molecule inhibitor PGMI-004A results in increased 3-PG and decreased 2-PG levels in cancer cells, leading to significantly decreased glycolysis, PPP flux and biosynthesis, as well as attenuated cell proliferation and tumor growth.


Subject(s)
Glycolysis/physiology , Neoplasms/enzymology , Phosphoglycerate Mutase/physiology , Animals , Binding, Competitive , Cell Line, Tumor , Cell Proliferation , Enzyme Activation , Gene Knockdown Techniques , Gluconates/metabolism , Glucosephosphate Dehydrogenase/metabolism , Glyceric Acids/metabolism , Glycolysis/genetics , Humans , Mice , Mice, Nude , Models, Molecular , Neoplasms/pathology , Phosphoglycerate Mutase/antagonists & inhibitors , Phosphoglycerate Mutase/genetics , Phosphoglycerate Mutase/metabolism
20.
Cell Oncol (Dordr) ; 34(2): 141-53, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21394647

ABSTRACT

BACKGROUND: Mantle cell lymphoma (MCL) is currently an incurable entity, and new therapeutic approaches are needed. We have applied a high-throughput phospho-proteomic technique to MCL cell lines to identify activated pathways and we have then validated our data in both cell lines and tumor tissues. METHODS: PhosphoScan analysis was performed on MCL cell lines. Results were validated by flow cytometry and western blotting. Functional validation was performed by blocking the most active pathway in MCL cell lines. RESULTS: PhosphoScan identified more than 300 tyrosine-phosporylated proteins, among which many protein kinases. The most abundant peptides belonged to proteins connected with B-cell receptor (BCR) signaling. Active BCR signaling was demonstrated by flow cytometry in MCL cells and by western blotting in MCL tumor tissues. Blocking BCR signaling by Syk inhibitor piceatannol induced dose/time-dependent apoptosis in MCL cell lines, as well as several modifications in the phosphorylation status of BCR pathway members and a collapse of cyclin D1 protein levels. CONCLUSION: Our data support a pro-survival role of BCR signaling in MCL and suggest that this pathway might be a candidate for therapy. Our findings also suggest that Syk activation patterns might be different in MCL compared to other lymphoma subtypes.


Subject(s)
Lymphoma, Mantle-Cell/metabolism , Phosphoproteins/metabolism , Proteomics/methods , Receptors, Antigen, B-Cell/metabolism , Blotting, Western , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Enzyme Activation/drug effects , Flow Cytometry , Fluorescent Antibody Technique , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Isoenzymes/metabolism , Lymphoma, Mantle-Cell/genetics , Lymphoma, Mantle-Cell/pathology , Models, Biological , Phosphoproteins/genetics , Phosphorylation , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Antigen, B-Cell/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/physiology , Stilbenes/pharmacology , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL