Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Cancer Res ; 84(7): 1065-1083, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38383964

ABSTRACT

Triple-negative breast cancer (TNBC) chemoresistance hampers the ability to effectively treat patients. Identification of mechanisms driving chemoresistance can lead to strategies to improve treatment. Here, we revealed that protein arginine methyltransferase-1 (PRMT1) simultaneously methylates D-3-phosphoglycerate dehydrogenase (PHGDH), a critical enzyme in serine synthesis, and the glycolytic enzymes PFKFB3 and PKM2 in TNBC cells. 13C metabolic flux analyses showed that PRMT1-dependent methylation of these three enzymes diverts glucose toward intermediates in the serine-synthesizing and serine/glycine cleavage pathways, thereby accelerating the production of methyl donors in TNBC cells. Mechanistically, PRMT1-dependent methylation of PHGDH at R54 or R20 activated its enzymatic activity by stabilizing 3-phosphoglycerate binding and suppressing polyubiquitination. PRMT1-mediated PHGDH methylation drove chemoresistance independently of glutathione synthesis. Rather, activation of the serine synthesis pathway supplied α-ketoglutarate and citrate to increase palmitate levels through activation of fatty acid synthase (FASN). Increased palmitate induced protein S-palmitoylation of PHGDH and FASN to further enhance fatty acid synthesis in a PRMT1-dependent manner. Loss of PRMT1 or pharmacologic inhibition of FASN or protein S-palmitoyltransferase reversed chemoresistance in TNBC. Furthermore, IHC coupled with imaging MS in clinical TNBC specimens substantiated that PRMT1-mediated methylation of PHGDH, PFKFB3, and PKM2 correlates with chemoresistance and that metabolites required for methylation and fatty acid synthesis are enriched in TNBC. Together, these results suggest that enhanced de novo fatty acid synthesis mediated by coordinated protein arginine methylation and protein S-palmitoylation is a therapeutic target for overcoming chemoresistance in TNBC. SIGNIFICANCE: PRMT1 promotes chemoresistance in TNBC by methylating metabolic enzymes PFKFB3, PKM2, and PHGDH to augment de novo fatty acid synthesis, indicating that targeting this axis is a potential treatment strategy.


Subject(s)
Phosphoglycerate Dehydrogenase , Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Drug Resistance, Neoplasm , Serine/metabolism , Palmitates , Fatty Acids , Cell Line, Tumor , Protein-Arginine N-Methyltransferases/genetics , Repressor Proteins
2.
Mol Genet Metab Rep ; 35: 100968, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36974075

ABSTRACT

The pyruvate dehydrogenase complex serves as the main connection between cytosolic glycolysis and the tricarboxylic acid cycle within mitochondria. An infant with pyruvate dehydrogenase complex deficiency was treated with vitamin B1 supplementation and a ketogenic diet. These dietary modifications resolved the renal tubular reabsorption, central apnea, and transfusion-dependent anemia. A concurrent metabolome analysis demonstrated the resolution of the amino aciduria and an increased total amount of substrates in the tricarboxylic acid cycle, reflecting the improved mitochondrial energetics. Glutamate was first detected in the cerebrospinal fluid, accompanied by a clinical improvement, after the ketogenic ratio was increased to 3:1; thus, glutamate levels in cerebrospinal fluid may represent a biomarker for neuronal recovery. Metabolomic analyses of body fluids are useful for monitoring therapeutic effects in infants with inborn errors of carbohydrate metabolism.

3.
Metabolites ; 11(12)2021 Dec 17.
Article in English | MEDLINE | ID: mdl-34940639

ABSTRACT

Cardiac dysfunction is induced by multifactorial mechanisms in diabetes. Deranged fatty acid (FA) utilization, known as lipotoxicity, has long been postulated as one of the upstream events in the development of diabetic cardiomyopathy. CD36, a transmembrane glycoprotein, plays a major role in FA uptake in the heart. CD36 knockout (CD36KO) hearts exhibit reduced rates of FA transport with marked enhancement of glucose use. In this study, we explore whether reduced FA use by CD36 ablation suppresses the development of streptozotocin (STZ)-induced diabetic cardiomyopathy. We found that cardiac contractile dysfunction had deteriorated 16 weeks after STZ treatment in CD36KO mice. Although accelerated glucose uptake was not reduced in CD36KO-STZ hearts, the total energy supply, estimated by the pool size in the TCA cycle, was significantly reduced. The isotopomer analysis with 13C6-glucose revealed that accelerated glycolysis, estimated by enrichment of 13C2-citrate and 13C2-malate, was markedly suppressed in CD36KO-STZ hearts. Levels of ceramides, which are cardiotoxic lipids, were not elevated in CD36KO-STZ hearts compared to wild-type-STZ ones. Furthermore, increased energy demand by transverse aortic constriction resulted in synergistic exacerbation of contractile dysfunction in CD36KO-STZ mice. These findings suggest that CD36KO-STZ hearts are energetically compromised by reduced FA use and suppressed glycolysis; therefore, the limitation of FA utilization is detrimental to cardiac energetics in this model of diabetic cardiomyopathy.

5.
Redox Biol ; 41: 101926, 2021 05.
Article in English | MEDLINE | ID: mdl-33752108

ABSTRACT

Chemosensitivity to cisplatin derivatives varies among individual patients with intractable malignancies including ovarian cancer, while how to unlock the resistance remain unknown. Ovarian cancer tissues were collected the debulking surgery in discovery- (n = 135) and validation- (n = 47) cohorts, to be analyzed with high-throughput automated immunohistochemistry which identified cystathionine γ-lyase (CSE) as an independent marker distinguishing non-responders from responders to post-operative platinum-based chemotherapy. We aimed to identify CSE-derived metabolites responsible for chemoresistant mechanisms: gold-nanoparticle (AuN)-based surface-enhanced Raman spectroscopy (SERS) was used to enhance electromagnetic fields which enabled to visualize multiple sulfur-containing metabolites through detecting scattering light from Au-S vibration two-dimensionally. Clear cell carcinoma (CCC) who turned out less sensitive to cisplatin than serous adenocarcinoma was classified into two groups by the intensities of SERS intensities at 480 cm-1; patients with greater intensities displayed the shorter overall survival after the debulking surgery. The SERS signals were eliminated by topically applied monobromobimane that breaks sulfane-sulfur bonds of polysulfides to result in formation of sulfodibimane which was detected at 580 cm-1, manifesting the presence of polysulfides in cancer tissues. CCC-derived cancer cell lines in culture were resistant against cisplatin, but treatment with ambroxol, an expectorant degrading polysulfides, renders the cells CDDP-susceptible. Co-administration of ambroxol with cisplatin significantly suppressed growth of cancer xenografts in nude mice. Furthermore, polysulfides, but neither glutathione nor hypotaurine, attenuated cisplatin-induced disturbance of DNA supercoiling. Polysulfide detection by on-tissue SERS thus enables to predict prognosis of cisplatin-based chemotherapy. The current findings suggest polysulfide degradation as a stratagem unlocking cisplatin chemoresistance.


Subject(s)
Antineoplastic Agents , Ovarian Neoplasms , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cisplatin , Drug Resistance, Neoplasm , Female , Humans , Mice , Mice, Nude , Ovarian Neoplasms/drug therapy , Spectrum Analysis, Raman , Sulfides
6.
Cell Rep ; 34(1): 108579, 2021 01 05.
Article in English | MEDLINE | ID: mdl-33406421

ABSTRACT

O-linked N-acetylglucosamine (O-GlcNAc) transferase (OGT) is a unique enzyme introducing O-GlcNAc moiety on target proteins, and it critically regulates various cellular processes in diverse cell types. However, its roles in hematopoietic stem and progenitor cells (HSPCs) remain elusive. Here, using Ogt conditional knockout mice, we show that OGT is essential for HSPCs. Ogt is highly expressed in HSPCs, and its disruption induces rapid loss of HSPCs with increased reactive oxygen species and apoptosis. In particular, Ogt-deficient hematopoietic stem cells (HSCs) lose quiescence, cannot be maintained in vivo, and become vulnerable to regenerative and competitive stress. Interestingly, Ogt-deficient HSCs accumulate defective mitochondria due to impaired mitophagy with decreased key mitophagy regulator, Pink1, through dysregulation of H3K4me3. Furthermore, overexpression of PINK1 restores mitophagy and the number of Ogt-deficient HSCs. Collectively, our results reveal that OGT critically regulates maintenance and stress response of HSCs by ensuring mitochondrial quality through PINK1-dependent mitophagy.


Subject(s)
Hematopoietic Stem Cells/metabolism , Histones/metabolism , Mitochondria/metabolism , Mitophagy , N-Acetylglucosaminyltransferases/metabolism , Protein Kinases/metabolism , Acetylglucosamine/metabolism , Animals , Apoptosis , Cell Cycle , Cell Line , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , N-Acetylglucosaminyltransferases/genetics , Reactive Oxygen Species/metabolism , Stress, Physiological
7.
Sci Rep ; 10(1): 20809, 2020 11 30.
Article in English | MEDLINE | ID: mdl-33257783

ABSTRACT

Diabetes is an independent risk factor for the development of heart failure. Increased fatty acid (FA) uptake and deranged utilization leads to reduced cardiac efficiency and accumulation of cardiotoxic lipids, which is suggested to facilitate diabetic cardiomyopathy. We studied whether reduced FA uptake in the heart is protective against streptozotocin (STZ)-induced diabetic cardiomyopathy by using mice doubly deficient in fatty acid binding protein 4 (FABP4) and FABP5 (DKO mice). Cardiac contractile dysfunction was aggravated 8 weeks after STZ treatment in DKO mice. Although compensatory glucose uptake was not reduced in DKO-STZ hearts, total energy supply, estimated by the pool size in the TCA cycle, was significantly reduced. Tracer analysis with 13C6-glucose revealed that accelerated glycolysis in DKO hearts was strongly suppressed by STZ treatment. Levels of ceramides, cardiotoxic lipids, were similarly elevated by STZ treatment. These findings suggest that a reduction in total energy supply by reduced FA uptake and suppressed glycolysis could account for exacerbated contractile dysfunction in DKO-STZ hearts. Thus, enhanced FA uptake in diabetic hearts seems to be a compensatory response to reduced energy supply from glucose, and therefore, limited FA use could be detrimental to cardiac contractile dysfunction due to energy insufficiency.


Subject(s)
Diabetic Cardiomyopathies/metabolism , Diabetic Cardiomyopathies/physiopathology , Fatty Acids/metabolism , Acetylation , Animals , Ceramides/metabolism , Citric Acid Cycle , Energy Metabolism , Female , Glucose/metabolism , Glycolysis , Ketone Bodies/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Myocardial Contraction , Streptozocin , Ventricular Dysfunction, Left
8.
Commun Biol ; 3(1): 450, 2020 08 17.
Article in English | MEDLINE | ID: mdl-32807853

ABSTRACT

Under hypoxic conditions, nitroimidazoles can replace oxygen as electron acceptors, thereby enhancing the effects of radiation on malignant cells. These compounds also accumulate in hypoxic cells, where they can act as cytotoxins or imaging agents. However, whether these effects apply to cancer stem cells has not been sufficiently explored. Here we show that the 2-nitroimidazole doranidazole potentiates radiation-induced DNA damage in hypoxic glioma stem cells (GSCs) and confers a significant survival benefit in mice harboring GSC-derived tumors in radiotherapy settings. Furthermore, doranidazole and misonidazole, but not metronidazole, manifested radiation-independent cytotoxicity for hypoxic GSCs that was mediated by ferroptosis induced partially through blockade of mitochondrial complexes I and II and resultant metabolic alterations in oxidative stress responses. Doranidazole also limited the growth of GSC-derived subcutaneous tumors and that of tumors in orthotopic brain slices. Our results thus reveal the theranostic potential of 2-nitroimidazoles as ferroptosis inducers that enable targeting GSCs in their hypoxic niche.


Subject(s)
Brain Neoplasms/pathology , Ferroptosis , Glioma/pathology , Mitochondria/pathology , Neoplastic Stem Cells/pathology , Nitroimidazoles/pharmacology , Stress, Physiological , Animals , Brain/pathology , Brain Neoplasms/metabolism , Cell Hypoxia/drug effects , Cell Proliferation/drug effects , Female , Ferroptosis/drug effects , Glioma/metabolism , Imidazoles/pharmacology , Metabolome , Mice, Inbred C57BL , Mitochondria/drug effects , Neoplastic Stem Cells/drug effects , Radiation-Sensitizing Agents/pharmacology , Stress, Physiological/drug effects
9.
Cancer Res ; 80(3): 471-483, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31767627

ABSTRACT

Recent studies have shown that stem cell memory T (TSCM) cell-like properties are important for successful adoptive immunotherapy by the chimeric antigen receptor-engineered-T (CAR-T) cells. We previously reported that both human and murine-activated T cells are converted into stem cell memory-like T (iTSCM) cells by coculture with stromal OP9 cells expressing the NOTCH ligand. However, the mechanism of NOTCH-mediated iTSCM reprogramming remains to be elucidated. Here, we report that the NOTCH/OP9 system efficiently converted conventional human CAR-T cells into TSCM-like CAR-T, "CAR-iTSCM" cells, and that mitochondrial metabolic reprogramming played a key role in this conversion. NOTCH signaling promoted mitochondrial biogenesis and fatty acid synthesis during iTSCM formation, which are essential for the properties of iTSCM cells. Forkhead box M1 (FOXM1) was identified as a downstream target of NOTCH, which was responsible for these metabolic changes and the subsequent iTSCM differentiation. Like NOTCH-induced CAR-iTSCM cells, FOXM1-induced CAR-iTSCM cells possessed superior antitumor potential compared with conventional CAR-T cells. We propose that NOTCH- or FOXM1-driven CAR-iTSCM formation is an effective strategy for improving cancer immunotherapy. SIGNIFICANCE: Manipulation of signaling and metabolic pathways important for directing production of stem cell memory-like T cells may enable development of improved CAR-T cells.


Subject(s)
Forkhead Box Protein M1/metabolism , Immunologic Memory/immunology , Leukemia/immunology , Organelle Biogenesis , Receptors, Chimeric Antigen/immunology , Receptors, Notch/metabolism , T-Lymphocytes/immunology , Animals , Cell Differentiation , Coculture Techniques , Humans , Immunotherapy, Adoptive , Leukemia/metabolism , Leukemia/pathology , Lymphocyte Activation , Mice , Mice, Inbred NOD , Mice, SCID , Signal Transduction , Stem Cells/immunology , Stromal Cells/immunology , Stromal Cells/metabolism , Stromal Cells/pathology
10.
JCI Insight ; 4(22)2019 11 14.
Article in English | MEDLINE | ID: mdl-31723053

ABSTRACT

Although oxidative stress plays central roles in postischemic renal injury, region-specific alterations in energy and redox metabolism caused by short-duration ischemia remain unknown. Imaging mass spectrometry enabled us to reveal spatial heterogeneity of energy and redox metabolites in the postischemic murine kidney. After 10-minute ischemia and 24-hour reperfusion (10mIR), in the cortex and outer stripes of the outer medulla, ATP substantially decreased, but not in the inner stripes of the outer medulla and inner medulla. 10mIR caused renal injury with elevation of fractional excretion of sodium, although histological damage by oxidative stress was limited. Ischemia-induced NADH elevation in the cortex indicated prolonged production of reactive oxygen species by xanthine oxidase (XOD). However, consumption of reduced glutathione after reperfusion suggested the amelioration of oxidative stress. An XOD inhibitor, febuxostat, which blocks the degradation pathway of adenine nucleotides, promoted ATP recovery and exerted renoprotective effects in the postischemic kidney. Because effects of febuxostat were canceled by silencing of the hypoxanthine phosphoribosyl transferase 1 gene in cultured tubular cells, mechanisms for the renoprotective effects appear to involve the purine salvage pathway, which uses hypoxanthine to resynthesize adenine nucleotides, including ATP. These findings suggest a novel therapeutic approach for acute ischemia/reperfusion renal injury with febuxostat through salvaging high-energy adenine nucleotides.


Subject(s)
Acute Kidney Injury , Adenine Nucleotides , Enzyme Inhibitors/pharmacology , Reperfusion Injury , Xanthine Oxidase/antagonists & inhibitors , Acute Kidney Injury/metabolism , Acute Kidney Injury/physiopathology , Adenine Nucleotides/analysis , Adenine Nucleotides/metabolism , Animals , Febuxostat/pharmacology , Kidney/chemistry , Kidney/drug effects , Kidney/pathology , Male , Mice , Mice, Inbred C57BL , Oxidative Stress/drug effects , Reactive Oxygen Species/analysis , Reactive Oxygen Species/metabolism , Reperfusion Injury/metabolism , Reperfusion Injury/physiopathology
11.
Sci Rep ; 8(1): 12035, 2018 08 13.
Article in English | MEDLINE | ID: mdl-30104639

ABSTRACT

The energy metabolism of the failing heart is characterized by reduced fatty acid (FA) oxidation and an increase in glucose utilization. However, little is known about how energy metabolism-function relationship is relevant to pathophysiology of heart failure. Recent study showed that the genetic deletion of CD36 (CD36KO), which causes reduction in FA use with an increased reliance on glucose, accelerates the progression from compensated hypertrophy to heart failure. Here, we show the mechanisms by which CD36 deletion accelerates heart failure in response to pressure overload. CD36KO mice exhibited contractile dysfunction and death from heart failure with enhanced cardiac hypertrophy and interstitial fibrosis when they were subjected to transverse aortic constriction (TAC). The pool size in the TCA cycle and levels of high-energy phosphate were significantly reduced in CD36KO-TAC hearts despite an increase in glycolytic flux. De novo synthesis of non-essential amino acids was facilitated in CD36KO-TAC hearts, which could cause a further decline of the pool size. The ingestion of a diet enriched in medium-chain FA improved cardiac dysfunction in CD36KO-TAC hearts. These findings suggest that myocardial FA uptake through CD36 is indispensable for sufficient ATP production and for preventing an increased glycolytic flux-mediated structural remodeling during pressure overload-induced hypertrophy.


Subject(s)
CD36 Antigens/metabolism , Cardiomegaly/physiopathology , Energy Metabolism/physiology , Fatty Acids/metabolism , Heart Failure/physiopathology , Myocardium/metabolism , Amino Acids/biosynthesis , Animals , CD36 Antigens/genetics , Cardiomegaly/genetics , Citric Acid Cycle/physiology , Fibrosis/pathology , Heart/physiology , Heart Failure/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout
12.
Mol Cell Biol ; 37(16)2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28606929

ABSTRACT

Metformin is one of the most widely used therapeutics for type 2 diabetes mellitus and also has anticancer and antiaging properties. However, it is known to induce metformin-associated lactic acidosis (MALA), a severe medical condition with poor prognosis, especially in individuals with renal dysfunction. Inhibition of prolyl hydroxylase (PHD) is known to activate the transcription factor hypoxia-inducible factor (HIF) that increases lactate efflux as a result of enhanced glycolysis, but it also enhances gluconeogenesis from lactate in the liver that contributes to reducing circulating lactate levels. Here, we investigated the outcome of pharmaceutical inhibition of PHD in mice with MALA induced through the administration of metformin per os and an intraperitoneal injection of lactic acid. We found that the PHD inhibitors significantly increased the expression levels of genes involved in gluconeogenesis in the liver and the kidney and significantly improved the survival of mice with MALA. Furthermore, the PHD inhibitor also improved the rate of survival of MALA induced in mice with chronic kidney disease (CKD). Thus, PHD represents a new therapeutic target for MALA, which is a critical complication of metformin therapy.


Subject(s)
Acidosis, Lactic/chemically induced , Acidosis, Lactic/enzymology , Metformin/adverse effects , Oxygen/metabolism , Procollagen-Proline Dioxygenase/metabolism , Acidosis, Lactic/pathology , Adenine , Animals , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Gluconeogenesis/drug effects , Kidney/drug effects , Kidney/metabolism , Liver/drug effects , Liver/metabolism , Male , Mice, Inbred C57BL , Procollagen-Proline Dioxygenase/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Messenger/metabolism , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/pathology , Survival Analysis , Up-Regulation/genetics
13.
Proc Natl Acad Sci U S A ; 112(37): 11642-7, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26324945

ABSTRACT

Loss of prolyl hydroxylase 2 (PHD2) activates the hypoxia-inducible factor-dependent hypoxic response, including anaerobic glycolysis, which causes large amounts of lactate to be released from cells into the circulation. We found that Phd2-null mouse embryonic fibroblasts (MEFs) produced more lactate than wild-type MEFs, as expected, whereas systemic inactivation of PHD2 in mice did not cause hyperlacticacidemia. This unexpected observation led us to hypothesize that the hypoxic response activated in the liver enhances the Cori cycle, a lactate-glucose carbon recycling system between muscle and liver, and thereby decreases circulating lactate. Consistent with this hypothesis, blood lactate levels measured after a treadmill or lactate tolerance test were significantly lower in Phd2-liver-specific knockout (Phd2-LKO) mice than in control mice. An in vivo (13)C-labeled lactate incorporation assay revealed that the livers of Phd2-LKO mice produce significantly more glucose derived from (13)C-labeled lactate than control mice, suggesting that blockade of PHD2 in the liver ameliorates lactic acidosis by activating gluconeogenesis from lactate. Phd2-LKO mice were resistant to lactic acidosis induced by injection of a lethal dose of lactate, displaying a significant elongation of survival. Moreover, oral administration of a PHD inhibitor improved survival in an endotoxin shock mice model. These data suggest that PHD2 is a potentially novel drug target for the treatment of lactic acidosis, which is a serious and often fatal complication observed in some critically ill patients.


Subject(s)
Acidosis, Lactic/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Liver/metabolism , Oxygen/metabolism , Animals , Blood Gas Analysis , Blood Glucose/metabolism , Genotype , Hepatocytes/metabolism , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lactates/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Physical Conditioning, Animal , Sepsis/metabolism
14.
J Biol Chem ; 289(42): 28956-70, 2014 Oct 17.
Article in English | MEDLINE | ID: mdl-25190814

ABSTRACT

Histone H3K4 methylation has been linked to transcriptional activation. KDM5A (also known as RBP2 or JARID1A), a member of the KDM5 protein family, is an H3K4 demethylase, previously implicated in the regulation of transcription and differentiation. Here, we show that KDM5A is physically and functionally associated with two histone deacetylase complexes. Immunoaffinity purification of KDM5A confirmed a previously described association with the SIN3B-containing histone deacetylase complex and revealed an association with the nucleosome remodeling and deacetylase (NuRD) complex. Sucrose density gradient and sequential immunoprecipitation analyses further confirmed the stable association of KDM5A with these two histone deacetylase complexes. KDM5A depletion led to changes in the expression of hundreds of genes, two-thirds of which were also controlled by CHD4, the NuRD catalytic subunit. Gene ontology analysis confirmed that the genes commonly regulated by both KDM5A and CHD4 were categorized as developmentally regulated genes. ChIP analyses suggested that CHD4 modulates H3K4 methylation levels at the promoter and coding regions of target genes. We further demonstrated that the Caenorhabditis elegans homologues of KDM5 and CHD4 function in the same pathway during vulva development. These results suggest that KDM5A and the NuRD complex cooperatively function to control developmentally regulated genes.


Subject(s)
Gene Expression Regulation , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Retinoblastoma-Binding Protein 2/metabolism , Animals , Autoantigens/metabolism , Caenorhabditis elegans/metabolism , Cell Line, Tumor , Chromatin/metabolism , Gene Expression Profiling , HeLa Cells , Histones/metabolism , Humans , MCF-7 Cells , Methylation , Nucleosomes/metabolism , Protein Binding , RNA, Small Interfering/metabolism , Repressor Proteins/metabolism , Transcription, Genetic
15.
Mol Cancer Res ; 11(9): 973-85, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23741060

ABSTRACT

UNLABELLED: Activation of aerobic glycolysis in cancer cells is well known as the Warburg effect, although its relation to cell- cycle progression remains unknown. In this study, human colon cancer cells were labeled with a cell-cycle phase-dependent fluorescent marker Fucci to distinguish cells in G1-phase and those in S + G2/M phases. Fucci-labeled cells served as splenic xenograft transplants in super-immunodeficient NOG mice and exhibited multiple metastases in the livers, frozen sections of which were analyzed by semiquantitative microscopic imaging mass spectrometry. Results showed that cells in G1-phase exhibited higher concentrations of ATP, NADH, and UDP-N-acetylglucosamine than those in S and G2-M phases, suggesting accelerated glycolysis in G1-phase cells in vivo. Quantitative determination of metabolites in cells synchronized in S, G2-M, and G1 phases suggested that efflux of lactate was elevated significantly in G1-phase. By contrast, ATP production in G2-M was highly dependent on mitochondrial respiration, whereas cells in S-phase mostly exhibited an intermediary energy metabolism between G1 and G2-M phases. Isogenic cells carrying a p53-null mutation appeared more active in glycolysis throughout the cell cycle than wild-type cells. Thus, as the cell cycle progressed from G2-M to G1 phases, the dependency of energy production on glycolysis was increased while the mitochondrial energy production was reciprocally decreased. IMPLICATIONS: These results shed light on distinct features of the phase-specific phenotypes of metabolic systems in cancer cells.


Subject(s)
Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Energy Metabolism , G1 Phase , Glycolysis , Adenosine Triphosphate/metabolism , Animals , Cell Line , G2 Phase , HCT116 Cells , Heterografts , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/secondary , Mice , Mitochondria/metabolism , Neoplasm Transplantation , Oxidative Phosphorylation , S Phase
16.
J Cell Sci ; 123(Pt 7): 1124-30, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20332121

ABSTRACT

PALB2 physically and functionally connects the proteins encoded by the BRCA1 and BRCA2 breast and ovarian cancer genes into a DNA-damage-response network. However, it remains unclear how these proteins associate with chromatin that contains damaged DNA. We show here that PALB2 binds directly to a conserved chromodomain protein, MRG15, which is a component of histone acetyltransferase-deacetylase complexes. This interaction was identified by analysis of purified MRG15- and PALB2-containing protein complexes. Furthermore, MRG15 interacts with the entire BRCA complex, which contains BRCA1, PALB2, BRCA2 and RAD51. Interestingly, MRG15-deficient cells, similarly to cells deficient in PALB2 or BRCA2, showed reduced efficiency for homology-directed DNA repair and hypersensitivity to DNA interstrand crosslinking agents. Additionally, knockdown of MRG15 diminished the recruitment of PALB2, BRCA2 and RAD51 to sites of DNA damage and reduced chromatin loading of PALB2 and BRCA2. These results suggest that MRG15 mediates DNA-damage-response functions of the BRCA complex in chromatin.


Subject(s)
Breast Neoplasms/genetics , DNA Repair , DNA , Epithelial Cells/metabolism , Nuclear Proteins/metabolism , Ovarian Neoplasms/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Chromatin/metabolism , Chromosome Breakage , Comet Assay , Cross-Linking Reagents/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/pathology , Fanconi Anemia Complementation Group N Protein , Female , Genes, BRCA1 , Genetic Predisposition to Disease , HeLa Cells , Humans , Mitomycin/pharmacology , Nuclear Proteins/genetics , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Protein Binding , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics
17.
Genes Cells ; 12(6): 811-26, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17573780

ABSTRACT

MRG15 is a conserved chromodomain protein that associates with histone deacetylases (HDACs) and Tip60-containing histone acetyltransferase (HAT) complexes. Here we further characterize MRG15-containing complexes and show a functional link between MRG15 and histone H3K4 demethylase activity in mammalian cells. MRG15 was predominantly localized to discrete nuclear subdomains enriched for Ser(2)-phosphorylated RNA polymerase II, suggesting it is involved specifically with active transcription. Protein analysis of the MRG15-containing complexes led to the identification of RBP2, a JmjC domain-containing protein. Remarkably, over-expression of RBP2 greatly reduced the H3K4 methylation in culture human cells in vivo, and recombinant RBP2 efficiently removed H3K4 methylation of histone tails in vitro. Knockdown of RBP2 resulted in increased H3K4 methylation levels within transcribed regions of active genes. Our findings demonstrate that RBP2 associated with MRG15 complex to maintain reduced H3K4 methylation at transcribed regions, which may ensure the transcriptional elongation state.


Subject(s)
Down-Regulation , Histone-Lysine N-Methyltransferase/physiology , Histones/chemistry , Lysine/chemistry , Retinol-Binding Proteins/physiology , Transcription Factors/physiology , Cell Line , Gene Expression Regulation, Fungal , Histone-Lysine N-Methyltransferase/chemistry , Humans , Methylation , Models, Biological , Models, Genetic , Phosphorylation , Retinol-Binding Proteins/chemistry , Retinol-Binding Proteins, Cellular , Saccharomyces cerevisiae/metabolism , Serine/chemistry , Transcription Factors/chemistry , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...