Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Nat Commun ; 14(1): 8095, 2023 Dec 13.
Article in English | MEDLINE | ID: mdl-38092728

ABSTRACT

Nicotinamide phosphoribosyltransferase (NAMPT) plays a major role in NAD biosynthesis in many cancers and is an attractive potential cancer target. However, factors dictating therapeutic efficacy of NAMPT inhibitors (NAMPTi) are unclear. We report that neuroendocrine phenotypes predict lung and prostate carcinoma vulnerability to NAMPTi, and that NAMPTi therapy against those cancers is enhanced by dietary modification. Neuroendocrine differentiation of tumor cells is associated with down-regulation of genes relevant to quinolinate phosphoribosyltransferase-dependent de novo NAD synthesis, promoting NAMPTi susceptibility in vitro. We also report that circulating nicotinic acid riboside (NAR), a non-canonical niacin absent in culture media, antagonizes NAMPTi efficacy as it fuels NAMPT-independent but nicotinamide riboside kinase 1-dependent NAD synthesis in tumors. In mouse transplantation models, depleting blood NAR by nutritional or genetic manipulations is synthetic lethal to tumors when combined with NAMPTi. Our findings provide a rationale for simultaneous targeting of NAR metabolism and NAMPT therapeutically in neuroendocrine carcinoma.


Subject(s)
Carcinoma, Neuroendocrine , Niacin , Male , Mice , Animals , Nicotinamide Phosphoribosyltransferase/metabolism , Niacin/pharmacology , Niacin/metabolism , NAD/metabolism , Cytokines/metabolism , Carcinoma, Neuroendocrine/drug therapy , Cell Line, Tumor
2.
J Steroid Biochem Mol Biol ; 221: 106103, 2022 07.
Article in English | MEDLINE | ID: mdl-35367370

ABSTRACT

Obesity has been known to increase the risks of breast cancer (BC) development and also to be associated with adverse clinical outcome of the patients. Abnormalities of cholesterol metabolism are not only related to obesity but also to biological or clinical behavior of BC patients. However, which metabolites or pathways of cholesterol metabolism could represent the characteristics of BC patients have remained virtually unknown. Therefore, in this study, we attempted to perform bird's eye view or comprehensive analysis of in situ or intra-tumoral cholesterol metabolic pathways using the multimodal approaches in order to elucidate the possible significance of cholesterol metabolites and its metabolic enzymes including CYP27A1, CYP7A1, and CYP46A1. GC-MS study using BC specimens was first performed in 60 BCE patients to evaluate cholesterol metabolism from cholesterol through oxysterols in both BC and normal tissues. Results of those analyses above lead to evaluating immunoreactivity and mRNA expression of CYP27A1, CYP7A1 and CYP46A1 in 213 and 153 BCE cases, respectively. Results of comprehensive GC-MS analysis did reveal that three oxysterols, 27-HC, 7α-HC and 24-HC were all related to malignant phenotypes in BC. 27-HC abundance was significantly associated with higher tumor stage (P = 0.0475) of BC patients. Luminal B type BC patients harboring high CYP27A1, the enzyme responsible for production of 27-HC were significantly associated with worse disease-free survival than those with low CYP27A1 (P = 0.0463). 7α-HC tended to be more abundant in HER2 positive and TNBC subtypes and higher levels of 7α-HC were also significantly associated with higher Ki-67 labeling index (P = 0.0022) and histological grade (P = 0.0286). CYP7A1, the enzyme involved in production of 7α-HC, was significantly more abundant in TNBC than other subtypes (vs Luminal A; P = 0.0321, vs Luminal B; P = 0.0048, vs HER2; P = 0.0103). The levels of 24-HC in BC were lower than normal breast tissues regardless of its subtypes. CYP46A1, the enzyme involved in the production of 24-HC, was detected only in 33 (15.5%) out of 213 BCE cases examined in this study. Results of our bird's eye view analysis of in situ or intra-tumoral cholesterol metabolism in BC patients did firstly reveal BC subtype dependent involvement of its different pathways. Results also indicated the therapeutic possibility of subtype dependent modification of cholesterol metabolizing pathways in BC patients.


Subject(s)
Breast Neoplasms , Oxysterols , Triple Negative Breast Neoplasms , Breast Neoplasms/metabolism , Cholesterol/metabolism , Cholesterol 24-Hydroxylase/metabolism , Cytochrome P-450 Enzyme System/metabolism , Female , Humans , Metabolic Networks and Pathways , Obesity , Oxysterols/metabolism
3.
Cancer Sci ; 113(5): 1613-1624, 2022 May.
Article in English | MEDLINE | ID: mdl-35247012

ABSTRACT

To examine effects of PP6 gene (Ppp6c) deficiency on pancreatic tumor development, we developed pancreas-specific, tamoxifen-inducible Cre-mediated KP (KRAS(G12D) plus Trp53-deficient) mice (cKP mice) and crossed them with Ppp6cflox / flox mice. cKP mice with the homozygous Ppp6c deletion developed pancreatic tumors, became emaciated and required euthanasia within 150 days of mutation induction, phenotypes that were not seen in heterozygous or wild-type (WT) mice. At 30 days, a comparative analysis of genes commonly altered in homozygous versus WT Ppp6c cKP mice revealed enhanced activation of Erk and NFκB pathways in homozygotes. By 80 days, the number and size of tumors and number of precancerous lesions had significantly increased in the pancreas of Ppp6c homozygous relative to heterozygous or WT cKP mice. Ppp6c-/- tumors were pathologically diagnosed as pancreatic ductal adenocarcinoma (PDAC) undergoing the epithelial-mesenchymal transition (EMT), and cancer cells had invaded surrounding tissues in three out of six cases. Transcriptome and metabolome analyses indicated an enhanced cancer-specific glycolytic metabolism in Ppp6c-deficient cKP mice and the increased expression of inflammatory cytokines. Individual Ppp6c-/- cKP mice showed weight loss, decreased skeletal muscle and adipose tissue, and increased circulating tumor necrosis factor (TNF)-α and IL-6 levels, suggestive of systemic inflammation. Overall, Ppp6c deficiency in the presence of K-ras mutations and Trp53 gene deficiency promoted pancreatic tumorigenesis with generalized cachexia and early death. This study provided the first evidence that Ppp6c suppresses mouse pancreatic carcinogenesis and supports the use of Ppp6c-deficient cKP mice as a model for developing treatments for cachexia associated with pancreatic cancer.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Phosphoprotein Phosphatases/metabolism , Animals , Cachexia/genetics , Carcinogenesis/genetics , Carcinoma, Pancreatic Ductal/pathology , Humans , Mice , Mutation , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Pancreatic Neoplasms
4.
Cancer Med ; 10(13): 4451-4464, 2021 07.
Article in English | MEDLINE | ID: mdl-34145991

ABSTRACT

BACKGROUND: Effective treatments for cancer harboring mutant RAS are lacking. In Drosophila, it was reported that PP6 suppresses tumorigenicity of mutant RAS. However, the information how PP6 regulates oncogenic RAS in mammals is limited. METHODS: We examined the effects of PP6 gene (Ppp6c) deficiency on tongue tumor development in K (K-rasG12D)- and KP (K-rasG12D + Trp53-deficient)-inducible mice. RESULTS: Mice of K and KP genotypes developed squamous cell carcinoma in situ in the tongue approximately 2 weeks after the induction of Ppp6c deficiency and was euthanized due to 20% loss of body weight. Transcriptome analysis revealed significantly different gene expressions between tissues of Ppp6c-deficient tongues and those of Ppp6c wild type, while Trp53 deficiency had a relatively smaller effect. We then analyzed genes commonly altered by Ppp6c deficiency, with or without Trp53 deficiency, and identified a group concentrated in KEGG database pathways defined as 'Pathways in Cancer' and 'Cytokine-cytokine receptor interaction'. We then evaluated signals downstream of oncogenic RAS and those regulated by PP6 substrates and found that in the presence of K-rasG12D, Ppp6c deletion enhanced the activation of the ERK-ELK1-FOS, AKT-4EBP1, and AKT-FOXO-CyclinD1 axes. Ppp6c deletion combined with K-rasG12D also enhanced DNA double-strand break (DSB) accumulation and activated NFκB signaling, upregulating IL-1ß, COX2, and TNF.


Subject(s)
Carcinoma in Situ/genetics , Carcinoma, Squamous Cell/genetics , Gene Deletion , Genes, ras , Phosphoprotein Phosphatases/deficiency , Tongue Neoplasms/genetics , Animals , DNA Breaks, Double-Stranded , Genotype , Mice , Mutation , Phosphoprotein Phosphatases/genetics , Transcriptome , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
5.
Cancer Sci ; 112(6): 2233-2244, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33743547

ABSTRACT

According to TCGA database, mutations in PPP6C (encoding phosphatase PP6) are found in c. 10% of tumors from melanoma patients, in which they coexist with BRAF and NRAS mutations. To assess PP6 function in melanoma carcinogenesis, we generated mice in which we could specifically induce BRAF(V600E) expression and delete Ppp6c in melanocytes. In these mice, melanoma susceptibility following UVB irradiation exhibited the following pattern: Ppp6c semi-deficient (heterozygous) > Ppp6c wild-type > Ppp6c-deficient (homozygous) tumor types. Next-generation sequencing of Ppp6c heterozygous and wild-type melanoma tumors revealed that all harbored Trp53 mutations. However, Ppp6c heterozygous tumors showed a higher Signature 1 (mitotic/mitotic clock) mutation index compared with Ppp6c wild-type tumors, suggesting increased cell division. Analysis of cell lines derived from either Ppp6c heterozygous or wild-type melanoma tissues showed that both formed tumors in nude mice, but Ppp6c heterozygous tumors grew faster compared with those from the wild-type line. Ppp6c knockdown via siRNA in the Ppp6c heterozygous line promoted the accumulation of genomic damage and enhanced apoptosis relative to siRNA controls. We conclude that in the presence of BRAF(V600E) expression and UV-induced Trp53 mutation, Ppp6c haploinsufficiency promotes tumorigenesis.


Subject(s)
Carcinogenesis/genetics , Melanoma/genetics , Phosphoprotein Phosphatases/genetics , Proto-Oncogene Proteins B-raf/genetics , Ultraviolet Rays/adverse effects , Animals , Carcinogenesis/pathology , Carcinogenesis/radiation effects , Exome/genetics , Exome/radiation effects , Genotype , Haploinsufficiency , Humans , Melanocytes/metabolism , Melanocytes/pathology , Melanocytes/radiation effects , Melanoma/pathology , Mice , Mice, Nude , Mice, Transgenic , Mutation/radiation effects , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
6.
Biochem Biophys Res Commun ; 530(1): 285-291, 2020 09 10.
Article in English | MEDLINE | ID: mdl-32828300

ABSTRACT

Recent advances in cancer biology reveal the importance of metabolic changes in cancer; however, less is known about how metabolic pathways in tumors are regulated in vivo. Here, we report analysis of the lung cancer metabolism based on different surgical procedures, namely lobectomy and partial resection. In lobectomy, but not in partial resection, pulmonary arteries and veins are ligated prior to removal of tissues, rendering tissues ischemic. We show that tumors indeed undergo ischemia upon lobectomy and that the tumor metabolome differs markedly from that of tumors removed by partial resection. Comparison of the responses to ischemia in tumor and normal lung tissues revealed that lung cancer tissue exhibits greater TCA cycle and autophagic activity than do normal lung tissues in vivo in patients. Finally, we report that deleting ATG7, which encodes a protein essential for autophagy, antagonizes growth of tumors derived from lung cancer cell lines, suggesting that autophagy confers metabolic advantages to lung cancer. Our findings shed light on divergent metabolic responses to ischemia seen in tumors and normal tissues.


Subject(s)
Citric Acid Cycle , Ischemia/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/surgery , Metabolome , Animals , Autophagy , Autophagy-Related Protein 7/genetics , Autophagy-Related Protein 7/metabolism , Cell Line, Tumor , Female , Gene Deletion , Ischemia/etiology , Ischemia/genetics , Ischemia/pathology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice
7.
Molecules ; 25(16)2020 Aug 10.
Article in English | MEDLINE | ID: mdl-32785052

ABSTRACT

Inhibition of nicotinamide phosphoribosyltransferase (NAMPT) is an attractive therapeutic strategy for targeting cancer metabolism. So far, many potent NAMPT inhibitors have been developed and shown to bind to two unique tunnel-shaped cavities existing adjacent to each active site of a NAMPT homodimer. However, cytotoxicities and resistances to NAMPT inhibitors have become apparent. Therefore, there remains an urgent need to develop effective and safe NAMPT inhibitors. Thus, we designed and synthesized two close structural analogues of NAMPT inhibitors, azaindole-piperidine (3a)- and azaindole-piperazine (3b)-motif compounds, which were modified from the well-known NAMPT inhibitor FK866 (1). Notably, 3a displayed considerably stronger enzyme inhibitory activity and cellular potency than did 3b and 1. The main reason for this phenomenon was revealed to be due to apparent electronic repulsion between the replaced nitrogen atom (N1) of piperazine in 3b and the Nδ atom of His191 in NAMPT by our in silico binding mode analyses. Indeed, 3b had a lower binding affinity score than did 3a and 1, although these inhibitors took similar stable chair conformations in the tunnel region. Taken together, these observations indicate that the electrostatic enthalpy potential rather than entropy effects inside the tunnel cavity has a significant impact on the different binding affinity of 3a from that of 3b in the disparate enzymatic and cellular potencies. Thus, it is better to avoid or minimize interactions with His191 in designing further effective NAMPT inhibitors.


Subject(s)
Drug Design , Enzyme Inhibitors/chemistry , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Binding Sites , Catalytic Domain , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/metabolism , Humans , Indoles/chemistry , Kinetics , Molecular Docking Simulation , Nicotinamide Phosphoribosyltransferase/metabolism , Piperazine/chemistry , Piperidines/chemistry
8.
FEBS Lett ; 594(9): 1379-1388, 2020 05.
Article in English | MEDLINE | ID: mdl-31950503

ABSTRACT

It is of current interest to target cancer metabolism as treatment for many malignancies, including ovarian cancer (OVC), in which few druggable driver mutations have been identified. Nicotinamide phosphoribosyltransferase (NAMPT), a rate-limiting enzyme in the NAD salvage pathway, is a potential therapeutic target in OVC. However, factors that determine responsiveness to NAMPT inhibition are not fully understood. Here, we report that OVC cell lines can be divided into subgroups exhibiting NAMPT-dependent or NAMPT-independent glycolysis, and these metabolic differences correlate with vulnerability to NAMPT inhibition. Interestingly, cells showing NAMPT-dependent glycolysis were enriched in a group of cells lacking BRCA1/2 gene mutations. Our findings suggest the importance of selecting appropriate patients for NAMPT-targeting therapy in OVC.


Subject(s)
Cytokines/antagonists & inhibitors , Cytokines/metabolism , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Nicotinamide Phosphoribosyltransferase/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Acrylamides/pharmacology , Cell Line, Tumor , Female , Glycolysis/drug effects , Humans , Lactic Acid/metabolism , NAD/metabolism , Niacin/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Piperidines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology
9.
Biochem Biophys Res Commun ; 522(3): 690-696, 2020 02 12.
Article in English | MEDLINE | ID: mdl-31787238

ABSTRACT

Lung adenocarcinoma (LUAC) is a unique lung cancer subtype that is responsive to several therapeutic agents. The KRAS gene is the second most frequently mutated gene in LUAC and the majority of KRAS mutations are one of three classical activating mutations (G12, G13, and Q61). Recently, other types of "minor" KRAS mutation have been identified among LUAC patients and some may have similar transforming activities to those of the classical KRAS mutations. Here we describe minor KRAS mutations in LUAC patients, some of which (A66T, A66V, and G75E) may have tumor-forming activity in mouse embryonic fibroblasts in an allograft model. RNA-Seq analysis revealed that mouse embryonic fibroblasts overexpressing these three minor KRAS mutations have distinct expression profiles compared with overexpression of the wild type but similar expression profiles compared with overexpression of the classical KRAS mutants. Our results indicate that some of the minor KRAS mutations cause varying tumor formation activity and are important targets for developing anti-RAS agents as chemotherapeutic agents.


Subject(s)
Adenocarcinoma of Lung/genetics , Lung Neoplasms/genetics , Point Mutation , Proto-Oncogene Proteins p21(ras)/genetics , Adenocarcinoma of Lung/pathology , Animals , Carcinogenesis/genetics , Carcinogenesis/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Lung Neoplasms/pathology , Mice , Models, Molecular , Transcriptome
10.
Mol Cell Oncol ; 5(4): e1472054, 2018.
Article in English | MEDLINE | ID: mdl-30250920

ABSTRACT

Isoform selection of pyruvate kinase M (PKM), a glycolytic enzyme, influences fates of glucose-derived carbons in cellular metabolic networks. We recently developed novel mouse lines to study PKM isoform function and identified PKM1 as a potential target in a subset of human lung cancers. This work provides new insight into cancer metabolism.

11.
Oncotarget ; 9(64): 32278-32279, 2018 Aug 17.
Article in English | MEDLINE | ID: mdl-30190785
12.
Cancer Sci ; 109(7): 2178-2187, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29758119

ABSTRACT

Here, we address the function of protein phosphatase 6 (PP6) loss on K-ras-initiated tumorigenesis in keratinocytes. To do so, we developed tamoxifen-inducible double mutant (K-rasG12D -expressing and Ppp6c-deficient) mice in which K-rasG12D expression is driven by the cytokeratin 14 (K14) promoter. Doubly-mutant mice showed early onset tumor formation in lips, nipples, external genitalia, anus and palms, and had to be killed by 3 weeks after induction by tamoxifen, while comparably-treated K-rasG12D -expressing mice did not. H&E-staining of lip tumors before euthanasia revealed that all were papillomas, some containing focal squamous cell carcinomas. Immunohistochemical analysis of lips of doubly-mutant vs K-rasG12D mice revealed that cell proliferation and cell size increased approximately 2-fold relative to K-rasG12D -expressing mutants, and epidermal thickness of lip tissue greatly increased relative to that seen in K-rasG12D -only mice. Moreover, AKT phosphorylation increased in K-rasG12D -expressing/Ppp6c-deficient cells, as did phosphorylation of the downstream effectors 4EBP1, S6 and GSK3, suggesting that protein synthesis and survival signals are enhanced in lip tissues of doubly-mutant mice. Finally, increased numbers of K14-positive cells were present in the suprabasal layer of doubly-mutant mice, indicating abnormal keratinocyte differentiation, and γH2AX-positive cells accumulated, indicating perturbed DNA repair. Taken together, Ppp6c deficiency enhances K-rasG12D -dependent tumor promotion.


Subject(s)
Carcinogenesis/genetics , Keratinocytes/enzymology , Phosphoprotein Phosphatases/metabolism , Skin Neoplasms/enzymology , Animals , Mice , Mice, Mutant Strains , Proto-Oncogene Proteins p21(ras)/genetics , Skin Neoplasms/genetics
13.
Cancer Cell ; 33(3): 355-367.e7, 2018 03 12.
Article in English | MEDLINE | ID: mdl-29533781

ABSTRACT

Expression of PKM2, which diverts glucose-derived carbon from catabolic to biosynthetic pathways, is a hallmark of cancer. However, PKM2 function in tumorigenesis remains controversial. Here, we show that, when expressed rather than PKM2, the PKM isoform PKM1 exhibits a tumor-promoting function in KRASG12D-induced or carcinogen-initiated mouse models or in some human cancers. Analysis of Pkm mutant mouse lines expressing specific PKM isoforms established that PKM1 boosts tumor growth cell intrinsically. PKM1 activated glucose catabolism and stimulated autophagy/mitophagy, favoring malignancy. Importantly, we observed that pulmonary neuroendocrine tumors (NETs), including small-cell lung cancer (SCLC), express PKM1, and that PKM1 expression is required for SCLC cell proliferation. Our findings provide a rationale for targeting PKM1 therapeutically in certain cancer subtypes, including pulmonary NETs.


Subject(s)
Carrier Proteins/genetics , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/genetics , Membrane Proteins/genetics , Thyroid Hormones/genetics , Animals , Carcinogenesis/genetics , Carrier Proteins/metabolism , Cell Cycle/genetics , Cell Line, Tumor , Membrane Proteins/metabolism , Mice, Knockout , Protein Isoforms/genetics , Thyroid Hormone-Binding Proteins
14.
Oncol Lett ; 14(6): 6863-6868, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29344123

ABSTRACT

Lung cancer is the most common cause of cancer mortality, however, efficient methods to culture, expand and transform lung epithelial (LE) cells have not been established. In the present study, an efficient ex vivo method was applied to recapitulate lung carcinogenesis using mouse LE cells. A Matrigel-assisted three-dimensional culture was used to isolate and selectively expand LE cells from mouse lungs. Purified LE cells were passaged and expanded for at least 2 to 3 months while maintaining epidermal growth factor-dependence. LE cells were also easily transformed by genetic manipulations using retroviral vectors. A SV40 large-T antigen, suppressing p53 and pRB, plus an activated oncogene, such as KrasG12V or EGFRex19del, were required to transform LE cells. Transformed cells formed tumors resembling non-small cell lung cancer (NSCLC) in allograft models and exhibited strong oncogene addiction. This experimental system provided a unique model system to study lung tumorigenesis and develop novel therapeutics against NSCLC.

15.
Mech Dev ; 139: 1-9, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26868000

ABSTRACT

Ppp6c, which encodes the catalytic subunit of phosphoprotein phosphatase 6 (PP6), is conserved among eukaryotes from yeast to humans. In mammalian cells, PP6 targets IκBε for degradation, activates DNA-dependent protein kinase to trigger DNA repair, and is reportedly required for normal mitosis. Recently, Ppp6c mutations were identified as candidate drivers of melanoma and skin cancer. Nonetheless, little is known about the physiological role of Ppp6c. To investigate this function in vivo, we established mice lacking the Ppp6c phosphatase domain by crossing heterozygous mutants. No viable homozygous pups were born, indicative of a lethal mutation. Ppp6c homozygous mutant embryos were identified among blastocysts, which exhibited a normal appearance, but embryos degenerated by E7.5 and showed clear developmental defects at E8.5, suggesting that mutant embryos die after implantation. Accordingly, homozygous blastocysts showed significant growth failure of the inner cell mass (ICM) in in vitro blastocyst culture, and primary Ppp6c exon4-deficient MEFs showed greatly reduced proliferation. These results establish for the first time that the Ppp6c phosphatase domain is indispensable for mouse embryogenesis after implantation.


Subject(s)
Phosphoprotein Phosphatases/physiology , Animals , Blastocyst/cytology , Blastocyst/enzymology , Cell Proliferation , Cells, Cultured , Embryo Culture Techniques , Embryo Implantation , Embryonic Development , Exons , Female , Genes, Lethal , Male , Mice, Inbred C57BL , Mice, Transgenic , Sequence Deletion
16.
Cancer Lett ; 365(2): 223-8, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26054846

ABSTRACT

We previously reported that deficiency in the gene encoding the catalytic subunit of protein phosphatase 6 (Ppp6c) predisposes mouse skin tissue to papilloma formation initiated by DMBA. Here, we demonstrate that Ppp6c loss acts as a tumor promoter in UVB-induced squamous cell carcinogenesis. Following UVB irradiation, mice with Ppp6c-deficient keratinocytes showed a higher incidence of skin squamous cell carcinoma than did control mice. Time course experiments showed that following UVB irradiation, Ppp6c-deficient keratinocytes upregulated expression of p53, PUMA, BAX, and cleaved caspase-3 proteins. UVB-induced tumors in Ppp6c-deficient keratinocytes exhibited a high frequency of both p53- and γH2AX-positive cells, suggestive of DNA damage. Epidemiological and molecular data strongly suggest that UVB from sunlight induces p53 gene mutations in keratinocytes and is the primary causative agent of human skin cancers. Our analysis suggests that PP6 deficiency underlies molecular events that drive outgrowth of initiated keratinocytes harboring UVB-induced mutated p53. Understanding PP6 function in preventing UV-induced tumorigenesis could suggest strategies to prevent and treat this condition.


Subject(s)
Carcinogenesis/radiation effects , Carcinoma, Squamous Cell/genetics , Keratinocytes/metabolism , Phosphoprotein Phosphatases/genetics , Ultraviolet Rays/adverse effects , Animals , Apoptosis/radiation effects , Apoptosis Regulatory Proteins/biosynthesis , Carcinogenesis/genetics , Caspase 3/metabolism , Cell Proliferation , DNA Damage/genetics , Histones/biosynthesis , Mice , Mice, Knockout , Skin/pathology , Skin/radiation effects , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Proteins/biosynthesis , bcl-2-Associated X Protein/biosynthesis
17.
Oncol Rep ; 28(3): 931-6, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22711061

ABSTRACT

Mitogen-activated protein kinase phosphatase 5 (MKP-5)/DUSP10 acts as a phosphatase of stress-activated kinases (JNK and p38), but its activity towards ERK has not been demonstrated. In the present study we observed that MKP-5 interacts with ERK, retains it in the cytoplasm, suppresses its activation and downregulates ERK-dependent transcription. These data suggested a novel MKP-5 function as a scaffold protein for the ERK pathway. We analyzed MKP-5 gene expression in several tumors, and found that it is frequently upregulated in colorectal but not in lung and breast cancer, suggesting its association with the malignant phenotype of colon cancer.


Subject(s)
Carcinoma/enzymology , Colonic Neoplasms/enzymology , Dual-Specificity Phosphatases/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Mitogen-Activated Protein Kinase Phosphatases/metabolism , Cell Line, Tumor , Dual-Specificity Phosphatases/genetics , Genes, Reporter , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Luciferases/biosynthesis , Luciferases/genetics , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase Phosphatases/genetics , Phosphorylation , Protein Processing, Post-Translational , Response Elements , Transcription, Genetic , Up-Regulation , p38 Mitogen-Activated Protein Kinases/metabolism
18.
Mol Cell Biochem ; 352(1-2): 155-62, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21360282

ABSTRACT

The dual-specificity phosphatase (DUSP) 13 gene encodes two atypical DUSPs, DUSP13B/TMDP and DUSP13A/MDSP using alternative exons. DUSP13B protein is most highly expressed in testis, particularly in spermatocytes and round spermatids of the seminiferous tubules, while that of DUSP13A is restricted to skeletal muscle. Here, we show that DUSP13B inactivated MAPK activation in the order of selectivity, JNK = p38>ERK in cells, while DUSP13A did not show MAPK phosphatase activity. Reporter gene analysis showed that DUSP13B had significant inhibitory effect on AP-1-dependent gene expression, but DUSP13A did not. To our knowledge, DUSP13B is the first identified testis-specific phosphatase that inhibits stress-activated MAPKs. These data suggest an important role for DUSP13B in protection from external stress during spermatogenesis.


Subject(s)
Dual-Specificity Phosphatases/physiology , Gene Expression Regulation/physiology , Mitogen-Activated Protein Kinases/metabolism , Transcription Factor AP-1/physiology , Animals , Cell Line , Enzyme Activation , Exons , Humans , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology
19.
Am J Physiol Lung Cell Mol Physiol ; 300(5): L740-52, 2011 May.
Article in English | MEDLINE | ID: mdl-21239537

ABSTRACT

Hypoxia-inducible factor-1α (HIF-1α), a transcription factor that functions as a master regulator of oxygen homeostasis, has been implicated in fibrinogenesis. Here, we explore the role of HIF-1α in transforming growth factor-ß (TGF-ß) signaling by examining the effects of TGF-ß(1) on the expression of plasminogen activator inhibitor-1 (PAI-1). Immunohistochemistry of lung tissue from a mouse bleomycin (BLM)-induced pulmonary fibrosis model revealed that expression of HIF-1α and PAI-1 was predominantly induced in alveolar macrophages. Real-time RT-PCR and ELISA analysis showed that PAI-1 mRNA and activated PAI-1 protein level were strongly induced 7 days after BLM instillation. Stimulation of cultured mouse alveolar macrophages (MH-S cells) with TGF-ß(1) induced PAI-1 production, which was associated with HIF-1α protein accumulation. This accumulation of HIF-1α protein was inhibited by SB431542 (type I TGF-ß receptor/ALK receptor inhibitor) but not by PD98059 (MEK1 inhibitor) and SB203580 (p38 MAP kinase inhibitor). Expression of prolyl-hydroxylase domain (PHD)-2, which is essential for HIF-1α degradation, was inhibited by TGF-ß(1), and this decrease was abolished by SB431542. TGF-ß(1) induction of PAI-1 mRNA and its protein expression were significantly attenuated by HIF-1α silencing. Transcriptome analysis by cDNA microarray of MH-S cells after HIF-1α silencing uncovered several pro-fibrotic genes whose regulation by TGF-ß(1) required HIF-1α, including platelet-derived growth factor-A. Taken together, these findings expand our concept of the role of HIF-1α in pulmonary fibrosis in mediating the effects of TGF-ß(1) on the expression of the pro-fibrotic genes in activated alveolar macrophages.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Macrophages, Alveolar/metabolism , Plasminogen Activator Inhibitor 1/biosynthesis , Transforming Growth Factor beta/pharmacology , Animals , Benzamides/pharmacology , Bleomycin , Cell Hypoxia/physiology , Dioxoles/pharmacology , Macrophages, Alveolar/drug effects , Male , Mice , Mice, Inbred C57BL , Pulmonary Fibrosis/chemically induced , Transforming Growth Factor beta/biosynthesis
20.
J Neurooncol ; 100(1): 43-9, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20217459

ABSTRACT

Cell division cycle 25 (CDC25) phosphatases are cell-cycle regulatory proteins which are overexpressed in a significant number of human cancers. This study evaluated the role of CDC25 phosphatases in human glioma proliferation. Upregulation of CDC25A was observed in human glioma specimens and human glioma cell lines. Comparison of expression levels of CDC25A and CDC25B messenger ribonucleic acid (RNA) to Ki-67 labeling index in glioma tissues found that Ki-67 labeling index was significantly correlated with the expression of CDC25A, but not with that of CDC25B. Depletion of CDC25A by small interfering RNA and inhibition of CDC25 suppressed cell proliferation and induced apoptosis in glioma cell lines, indicating that CDC25A is a potential target for the development of new therapy for glioma.


Subject(s)
Brain Neoplasms/metabolism , Gene Expression/physiology , Glioma/metabolism , Ki-67 Antigen/metabolism , RNA, Messenger/metabolism , Statistics as Topic , cdc25 Phosphatases/genetics , Adult , Aged , Aged, 80 and over , Benzoquinones/pharmacology , Brain Neoplasms/genetics , Caspase 3/metabolism , Caspase 7/metabolism , Cell Death/genetics , Cell Line, Tumor , Cell Proliferation , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Ethylamines/pharmacology , Female , Glioma/genetics , Humans , Ki-67 Antigen/genetics , Male , Middle Aged , Nitro Compounds/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Thiazoles/pharmacology , Time Factors , Transfection/methods , Up-Regulation/genetics , Young Adult , cdc25 Phosphatases/antagonists & inhibitors , cdc25 Phosphatases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...