Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Article in English | MEDLINE | ID: mdl-38835135

ABSTRACT

Melanoma, a cancer arising from melanocytes, requires a novel treatment strategy because of the ineffectiveness of conventional therapies in certain patients. Fustin is a flavanonol found in young fustic (Cotinus coggygria). However, little is known about its antimelanoma effects. Our study demonstrates that fustin suppresses the growth of B16 melanoma cells. Phalloidin staining of cytoskeletal actin revealed that fustin induced a conformational change in the actin structure of melanoma cells, accompanied by suppressed phosphorylation of myosin regulatory light chain 2 (MLC2), a regulator of actin structure. Furthermore, the protein kinase A (cAMP-dependent protein kinase) inhibitor H89 completely attenuated fustin-induced downregulation of phosphorylated myosin phosphatase targeting subunit 1 (MYPT1), which is involved in dephosphorylation of MLC2. In a mouse model, administration of fustin suppressed tumor growth in B16 melanoma cells without adverse effects. In conclusion, our findings suggest that fustin effectively suppresses melanoma cell growth both in vitro and in vivo.

2.
Biochem Biophys Res Commun ; 672: 154-160, 2023 09 10.
Article in English | MEDLINE | ID: mdl-37354608

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a type of cancer with a high mortality rate. Current treatments for PDACs often have side effects, and drug resistance in cancer stem cells (CSCs) would be also a problem. Cyclic guanosine monophosphate (cGMP) suppresses the mitochondrial function of PDACs and inhibits their CSC properties. Metabolic regulation plays a crucial role in the maintenance of CSC phenotype, and we hypothesized that cGMP induction suppresses cancer stem cell properties in the cancer cell through energy-related signaling pathways. We demonstrated that induction of cGMP upregulated the PPARα/PDK4 pathway and suppressed CSC properties in PDAC, and patients with pancreatic cancer with high PDK4 gene expression had a better prognosis than those with low gene expression. Therefore, these mechanisms may provide new therapeutic targets for the eradication of pancreatic CSCs.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Peroxisome Proliferator-Activated Receptors/metabolism , Pancreatic Neoplasms/pathology , Carcinoma, Pancreatic Ductal/pathology , Pancreas/metabolism , Neoplastic Stem Cells/pathology , Cell Line, Tumor , Pancreatic Neoplasms
3.
Mamm Genome ; 33(1): 55-65, 2022 03.
Article in English | MEDLINE | ID: mdl-34482425

ABSTRACT

Recombinase alleles and transgenes can be used to facilitate spatio-temporal specificity of gene disruption or transgene expression. However, the versatility of this in vivo recombination system relies on having detailed and accurate characterization of recombinase expression and activity to enable selection of the appropriate allele or transgene. The CrePortal ( http://www.informatics.jax.org/home/recombinase ) leverages the informatics infrastructure of Mouse Genome Informatics to integrate data from the scientific literature, direct data submissions from the scientific community at-large, and from major projects developing new recombinase lines and characterizing recombinase expression and specificity patterns. Searching the CrePortal by recombinase activity or specific recombinase gene driver provides users with a recombinase alleles and transgenes activity tissue summary and matrix comparison of gene expression and recombinase activity with links to generation details, a recombinase activity grid, and associated phenotype annotations. Future improvements will add cell type-based activity annotations. The CrePortal provides a comprehensive presentation of recombinase allele and transgene data to assist researchers in selection of the recombinase allele or transgene based on where and when recombination is desired.


Subject(s)
Integrases , Recombinases , Alleles , Animals , Integrases/genetics , Integrases/metabolism , Mice , Mice, Transgenic , Recombinases/genetics , Transgenes
4.
Prog Oceanogr ; 1802020 Jan.
Article in English | MEDLINE | ID: mdl-33184522

ABSTRACT

Seven South Pacific anguillid eel species live from New Guinea to French Polynesia, but their spawning areas and life histories are mostly unknown despite previous sampling surveys. A July-October 2016 research cruise was conducted to study the spawning areas and times, and larval distributions of South Pacific anguillid eels, which included a short 155°E station-line northeast of New Guinea and five long transects (5-25°S, 160°E-140°W) crossing the South Equatorial (SEC) and other currents. This survey collected nearly 4000 anguilliform leptocephali at 179 stations using an Isaacs-Kidd Midwater Trawl accompanied by 104 CTD casts. Based on mor-phometric observations and DNA sequencing, 74 anguillid leptocephali were collected, which in the southern areas included 29 larvae of six species: Anguilla bicolor pacifica, A. marmorata, A. australis, A. reinhardtii, A. megastoma, and A. obscura (all anguillid species of the region were caught except A. dieffenbachii). Small A. australis (9.0-16.8 mm) and A. reinhardtii (12.4, 12.5 mm) leptocephali were collected south of the Solomon Islands, other A. australis (10.8-12.0 mm) larvae were caught northwest of Fiji along with an A. obscura (20.0 mm) larva, and an A. marmorata (7.8 mm) larva was collected near Samoa. Considering collection sites, larval ages from otolith analysis, and westward SEC drift, multiple spawning locations occurred from south of the Solomon Islands and the Fiji area (16-20 days old larvae) to near Samoa (19 days old larva) during June and July in areas where high-salinity Subtropical Underwater (STUW, ~150 m depth) and the warm, low-salinity surface Fresh Pool were present. Five long hydrographic sections showed the strong Fresh Pool in the west and the STUW formation area in the east.

5.
J Immunol ; 199(9): 3261-3269, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28954885

ABSTRACT

TLR signaling is critical to innate immune system regulation; however, aberrant TLR signaling is involved in several diseases, including insulin resistance, Alzheimer's disease, and tumor metastasis. Moreover, a recent study found that TLR-4 signaling pathway inhibition might be a target for the suppression of chronic inflammatory disorders. In this article, we show that the green tea polyphenol epigallocatechin-3-O-gallate (EGCG) increases the expression of Toll interacting protein, a strong inhibitor of TLR4 signaling, by suppressing the expression of E74-like ETS transcription factor 1 (Elf-1). A mechanistic study revealed that EGCG suppressed Elf-1 expression via protein phosphatase 2A/cyclic GMP (cGMP)-dependent mechanisms. We also confirmed that orally administered EGCG and a cGMP inducer upregulated Toll interacting protein expression, increased intracellular levels of cGMP in macrophages, and suppressed Elf-1 expression. These data support EGCG and a cGMP inducer as potential candidate suppressors of TLR4 signaling.


Subject(s)
Catechin/analogs & derivatives , DNA-Binding Proteins/immunology , Intracellular Signaling Peptides and Proteins/immunology , Nuclear Proteins/immunology , Second Messenger Systems/immunology , Tea/chemistry , Transcription Factors/immunology , Up-Regulation/immunology , Animals , Catechin/chemistry , Catechin/pharmacology , Cyclic GMP/genetics , Cyclic GMP/immunology , DNA-Binding Proteins/genetics , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Inbred BALB C , Nuclear Proteins/genetics , Protein Phosphatase 2/genetics , Protein Phosphatase 2/immunology , Second Messenger Systems/genetics , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Transcription Factors/genetics
6.
J Biol Chem ; 292(26): 10813-10823, 2017 06 30.
Article in English | MEDLINE | ID: mdl-28507102

ABSTRACT

In 95% of patients with pancreatic ductal adenocarcinoma, recurrence is observed following chemotherapy. Findings from several studies have indicated that cancer stem cells (CSCs) are resistant to anticancer agents and may be involved in cancer recurrence and metastasis. The CD44 protein is a major CSC marker, and CD44 also plays an indispensable role in the CSC properties in several cancers, including pancreatic cancer; however, no clinical approach exists to inhibit CD44 activity. Here, we have performed knock-in/knockdown experiments, and we demonstrate that the forkhead box O3 (FOXO3)/liver kinase B1 (LKB1)/AMP-activated protein kinase/peroxisome proliferator-activated receptor-γ co-activator-1ß (PGC-1ß)/pyruvate dehydrogenase-A1 pathway is essential for CD44 expression and CSC properties. We observed that patients exhibiting high pyruvate dehydrogenase-A1 expression have a poor prognosis. Systemic PGC-1ß knock-out mice are fertile and viable and do not exhibit an overt phenotype under normal conditions. This suggests that cGMP induction and PGC-1ß inhibition represent potential strategies for treating patients with pancreatic ductal adenocarcinoma.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Carrier Proteins/metabolism , Forkhead Box Protein O3/metabolism , Neoplasm Proteins/metabolism , Pancreatic Neoplasms/metabolism , Signal Transduction , Animals , Biomarkers, Tumor/genetics , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Carrier Proteins/genetics , Cell Line, Tumor , Female , Forkhead Box Protein O3/genetics , Humans , Male , Mice, Knockout , Neoplasm Proteins/genetics , Neoplastic Stem Cells , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , RNA-Binding Proteins
8.
Proc Natl Acad Sci U S A ; 108(10): 4129-34, 2011 Mar 08.
Article in English | MEDLINE | ID: mdl-21325052

ABSTRACT

Although aerobic glycolysis (the Warburg effect) is a hallmark of cancer, key questions, including when, how, and why cancer cells become highly glycolytic, remain less clear. For a largely unknown regulatory mechanism, a rate-limiting glycolytic enzyme pyruvate kinase M2 (PKM2) isoform is exclusively expressed in embryonic, proliferating, and tumor cells, and plays an essential role in tumor metabolism and growth. Because the receptor tyrosine kinase/PI3K/AKT/mammalian target of rapamycin (RTK/PI3K/AKT/mTOR) signaling cascade is a frequently altered pathway in cancer, we explored its potential role in cancer metabolism. We identified mTOR as a central activator of the Warburg effect by inducing PKM2 and other glycolytic enzymes under normoxic conditions. PKM2 level was augmented in mouse kidney tumors due to deficiency of tuberous sclerosis complex 2 and consequent mTOR activation, and was reduced in human cancer cells by mTOR suppression. mTOR up-regulation of PKM2 expression was through hypoxia-inducible factor 1α (HIF1α)-mediated transcription activation, and c-Myc-heterogeneous nuclear ribonucleoproteins (hnRNPs)-dependent regulation of PKM2 gene splicing. Disruption of PKM2 suppressed oncogenic mTOR-mediated tumorigenesis. Unlike normal cells, mTOR hyperactive cells were more sensitive to inhibition of mTOR or glycolysis. Dual suppression of mTOR and glycolysis synergistically blunted the proliferation and tumor development of mTOR hyperactive cells. Even though aerobic glycolysis is not required for breach of senescence for immortalization and transformation, the frequently deregulated mTOR signaling during multistep oncogenic processes could contribute to the development of the Warburg effect in many cancers. Components of the mTOR/HIF1α/Myc-hnRNPs/PKM2 glycolysis signaling network could be targeted for the treatment of cancer caused by an aberrant RTK/PI3K/AKT/mTOR signaling pathway.


Subject(s)
Neoplasms/pathology , Pyruvate Kinase/metabolism , TOR Serine-Threonine Kinases/physiology , Aerobiosis , Animals , Cell Proliferation , Genes, myc , Glycolysis , Humans , Mice , Neoplasms/enzymology , Neoplasms/metabolism , TOR Serine-Threonine Kinases/metabolism , Up-Regulation
9.
iScience ; 27(1): 108730, 2024 Jan 19.
Article in English | MEDLINE | ID: mdl-38235326

ABSTRACT

Cirrhosis is becoming one of the most common diseases worldwide. Abnormal upregulation of transforming growth factor ß (TGF-ß) signaling plays a pivotal role in the excess activation of hepatic stellate cells. However, an efficient countermeasure against abnormal hepatic stellate cell activation is yet to be established because TGF-ß signaling is involved in several biological processes. Herein, we demonstrated the antifibrotic effect of miR-12135, a microRNA with unknown function upregulated by isoflavone. Comprehensive transcriptome assay demonstrated that miR-12135 suppressed Integrin Subunit Alpha 11 (ITGA11) and that ITGA11 expression is correlated with alpha smooth muscle actin expression in patients with cirrhosis. miR-12135 suppressed the expression level of ITGA11 and liver fibrosis. Importantly, ITGA11 is overexpressed in activated hepatic stellate cells, whereas ITGA11 knockout mice are viable and fertile. In conclusions, the miR-12135/ITGA11 axis can be an ideal therapeutic target to suppress fibrosis by precisely targeting abnormally upregulated TGF-ß signaling in hepatic stellate cells.

10.
Sci Rep ; 13(1): 2128, 2023 02 06.
Article in English | MEDLINE | ID: mdl-36746980

ABSTRACT

Lung fibrosis, including idiopathic pulmonary fibrosis, is an intractable disease accompanied by an irreversible dysfunction in the respiratory system. Its pathogenesis involves the transforming growth factorß (TGFß)-induced overproduction of the extracellular matrix from fibroblasts; however, limited countermeasures have been established. In this study, we identified osa-miR172d-5p, a plant-derived microRNA (miR), as a potent anti-fibrotic miR. In silico analysis followed by an in vitro assay based on human lung fibroblasts demonstrated that osa-miR172d-5p suppressed the gene expression of TGF-ß activated kinase 1 (MAP3K7) binding protein 1 (Tab1). It also suppressed the TGFß-induced fibrotic gene expression in human lung fibroblasts. To assess the anti-fibrotic effect of osa-miR172d-5p, we established bleomycin-induced lung fibrosis models to demonstrate that osa-miR172d-5p ameliorated lung fibrosis. Moreover, it suppressed Tab1 expression in the lung tissues of bleomycin-treated mice. In conclusion, osa-miR172d-5p could be a potent candidate for the treatment of lung fibrosis, including idiopathic pulmonary fibrosis.


Subject(s)
Idiopathic Pulmonary Fibrosis , MicroRNAs , Humans , Mice , Animals , MicroRNAs/metabolism , Lung/pathology , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/metabolism , Fibrosis , Bleomycin/toxicity , Bleomycin/metabolism , Fibroblasts/metabolism , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism
11.
Food Chem Toxicol ; 169: 113421, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36100043

ABSTRACT

Anticancer agents can cause various side effects, including tissue damages/inflammatory reactions. Drug-responsive biomarkers are essential for evaluating drug toxicity in disease processes. S100 calcium-binding proteins A8/A9 (S100A8/A9) are highly expressed in neutrophils and monocytes/macrophages accumulated at inflammatory sites and are known to be related to tissue damage/inflammation; however, their response to drug toxicity has not been reported. Herein, we investigated the effects of anticancer agents (doxorubicin, cisplatin, and docetaxel) on S100A8/A9 gene expression profiles in four representative tissues (heart, kidney, liver, and lung) in normal C57BL/6J mice. Both S100A8/A9 expression was transiently or time-dependently elevated in four tissues within 48 h after dosing of the three anticancer agents under toxicity-inducing conditions. S100A8/A9 patterns differed among agents and tissues. This result suggests that S100A8/A9 is useful for evaluating anticancer agent-induced tissue damage. Metabolomic analysis revealed that some metabolites showed temporal patterns similar to that of S100A8/A9 expression. The amounts of fumarate (doxorubicin-treated heart), tyrosine (cisplatin-treated kidney), acetylcarnosine (doxorubicin-treated liver), and 2-phosphoglycerate (docetaxel-treated lung) showed similar patterns to that of S100A8/A9 expression. Although these metabolites showed different behaviors between tissues and serum, they may be useful marker candidates for evaluating anticancer agent-induced tissue damage at an earlier stage after dosing.


Subject(s)
Antineoplastic Agents , Biomarkers, Pharmacological , Calgranulin A , Calgranulin B , Inflammation , Animals , Mice , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/toxicity , Biomarkers, Pharmacological/metabolism , Calgranulin A/genetics , Calgranulin A/metabolism , Calgranulin B/genetics , Calgranulin B/metabolism , Cisplatin/administration & dosage , Cisplatin/toxicity , Docetaxel/administration & dosage , Docetaxel/toxicity , Doxorubicin/administration & dosage , Doxorubicin/toxicity , Fumarates/analysis , Inflammation/chemically induced , Inflammation/metabolism , Mice, Inbred C57BL , Tyrosine/analysis
12.
J Nat Med ; 75(4): 1037-1042, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34100197

ABSTRACT

Animal and clinical studies have revealed that (-)-epigallocatechin-3-O-gallate (EGCG), one of the major bioactive polyphenols in green tea, showed several pharmacological effects including anti-obesity effect and anti-inflammatory effect. We previously reported that the second messenger cyclic guanosine monophosphate (cGMP) mediates its anti-inflammatory and anti-cancer properties. Here we demonstrated that glucosyl-hesperidin, enhances the cGMP-inducing effects of green tea extract in vivo. Moreover, glucosyl-hesperidin intake potentiated the green tea-elicited upregulation of the anti-inflammatory factor, toll-interacting protein.


Subject(s)
Catechin , Hesperidin , Animals , Catechin/analogs & derivatives , Catechin/pharmacology , Guanosine Monophosphate , Polyphenols/pharmacology , Tea
13.
J Neurosci ; 27(21): 5546-58, 2007 May 23.
Article in English | MEDLINE | ID: mdl-17522300

ABSTRACT

Tuberous sclerosis (TSC) is a hamartoma syndrome caused by mutations in TSC1 or TSC2 in which cerebral cortical tubers and seizures are major clinical issues. We have engineered mice in which most cortical neurons lose Tsc1 expression during embryonic development. These Tsc1 mutant mice display several neurological abnormalities beginning at postnatal day 5 with subsequent failure to thrive and median survival of 35 d. The mice also display clinical and electrographic seizures both spontaneously and with physical stimulation, and some seizures end in a fatal tonic phase. Many cortical and hippocampal neurons are enlarged and/or dysplastic in the Tsc1 mutant mice, strongly express phospho-S6, and are ectopic in multiple sites in the cortex and hippocampus. There is a striking delay in myelination in the mutant mice, which appears to be caused by an inductive neuronal defect. This new TSC brain model replicates several features of human TSC brain lesions and implicates an important function of Tsc1/Tsc2 in neuronal development.


Subject(s)
Demyelinating Diseases/pathology , Disease Models, Animal , Neurons/pathology , Seizures/pathology , Tuberous Sclerosis/pathology , Tumor Suppressor Proteins/deficiency , Animals , Demyelinating Diseases/genetics , Demyelinating Diseases/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Neurons/metabolism , Seizures/genetics , Seizures/metabolism , Survival Rate , Tuberous Sclerosis/genetics , Tuberous Sclerosis/metabolism , Tuberous Sclerosis/mortality , Tuberous Sclerosis Complex 1 Protein , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/physiology
14.
Biochem Biophys Res Commun ; 367(4): 899-905, 2008 Mar 21.
Article in English | MEDLINE | ID: mdl-18194669

ABSTRACT

PRDM14 was identified by microarray analysis and was expressed in specifically undifferentiated human ES cells. PRDM14 protein is thought to regulate gene transcription in human ES cells, as it contains a PR domain, a subtype of the SET domain which catalyzes histone methylation. To analyze the function of PRDM14, we performed knock-down and forced expression of PRDM14 in human ES cells. Knock-down of PRDM14 by siRNA induced expression of early differentiation marker genes. Forced expression of PRDM14 suppressed expression of differentiation marker genes in the embryoid body. These results suggest that PRDM14 is involved in the maintenance of the self-renewal of human ES cells by suppression of gene expression.


Subject(s)
Embryo, Mammalian/metabolism , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental/physiology , Repressor Proteins/metabolism , Transcription Factors/metabolism , Biomarkers/metabolism , Cell Differentiation/physiology , Cells, Cultured , DNA-Binding Proteins , Humans , RNA-Binding Proteins
15.
Nucleic Acids Res ; 34(17): 4878-92, 2006.
Article in English | MEDLINE | ID: mdl-16973895

ABSTRACT

We report here the isolation of 44 genes that are upregulated after serum starvation and/or contact inhibition. These genes have been termed TIGA, after Transcript Induced by Growth Arrest. We found that there are two kinds of G0 phases caused by serum starvation, namely, the shallow G0 (or G0/G1) and the deep G0 phases. The shallow G0 is induced by only a few hours of serum starvation, while deep G0 is generated after 3 days of serum starvation. We propose that mammalian cells enter deep G0 through a G0 gate, which is only opened on the third day of serum starvation. TIGA1, one of the uncharacterized TIGA genes, encodes a homolog of cyanate permease of bacteria and localizes in mitochondria. This suggests that Tiga1 is involved in the inorganic ion transport and metabolism needed to maintain the deep G0 phase. Ectopic expression of TIGA1 inhibited not only tumor cell proliferation but also anchorage-independent growth of cancer cell lines. A microsatellite marker, ENDL-1, allowed us to detect loss of heterozygosity around the TIGA1 gene region (5q21-22). Further analysis of the TIGA genes we have identified here may help us to better understand the mechanisms that regulate the G0 phase.


Subject(s)
Membrane Transport Proteins/physiology , Mitochondrial Proteins/physiology , RNA, Messenger/biosynthesis , Resting Phase, Cell Cycle/genetics , Up-Regulation , Amino Acid Sequence , Animals , Blotting, Western , Cell Line , Cell Line, Tumor , Cell Proliferation , Contact Inhibition , Culture Media, Serum-Free , Genes, Tumor Suppressor , Humans , Kinetics , Loss of Heterozygosity , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/physiology , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/genetics , Molecular Sequence Data , Neoplasms/genetics , Protein Structure, Tertiary , RNA, Long Noncoding , RNA, Messenger/metabolism , Sequence Homology, Amino Acid , Tumor Stem Cell Assay
16.
J Clin Invest ; 112(8): 1223-33, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14561707

ABSTRACT

Tuberous sclerosis (TSC) is a familial tumor syndrome due to mutations in TSC1 or TSC2, in which progression to malignancy is rare. Primary Tsc2(-/-) murine embryo fibroblast cultures display early senescence with overexpression of p21CIP1/WAF1 that is rescued by loss of TP53. Tsc2(-/-)TP53(-/-) cells, as well as tumors from Tsc2(+/-) mice, display an mTOR-activation signature with constitutive activation of S6K, which is reverted by treatment with rapamycin. Rapamycin also reverts a growth advantage of Tsc2(-/-)TP53(-/-) cells. Tsc1/Tsc2 does not bind directly to mTOR, however, nor does it directly influence mTOR kinase activity or cellular phosphatase activity. There is a marked reduction in Akt activation in Tsc2(-/-)TP53(-/-) and Tsc1(-/-) cells in response to serum and PDGF, along with a reduction in cell ruffling. PDGFRalpha and PDGFRbeta expression is markedly reduced in both the cell lines and Tsc mouse renal cystadenomas, and ectopic expression of PDGFRbeta in Tsc2-null cells restores Akt phosphorylation in response to serum, PDGF, EGF, and insulin. This activation of mTOR along with downregulation of PDGFR PI3K-Akt signaling in cells lacking Tsc1 or Tsc2 may explain why these genes are rarely involved in human cancer. This is in contrast to PTEN, which is a negative upstream regulator of this pathway.


Subject(s)
Phosphatidylinositol 3-Kinases/physiology , Protein Kinases/physiology , Protein Serine-Threonine Kinases , Proteins/physiology , Proto-Oncogene Proteins/physiology , Receptors, Platelet-Derived Growth Factor/analysis , Repressor Proteins/physiology , Signal Transduction/physiology , Animals , Cells, Cultured , Cellular Senescence , Down-Regulation , Male , Mice , Proto-Oncogene Proteins c-akt , Ribosomal Protein S6 Kinases/metabolism , TOR Serine-Threonine Kinases , Tuberous Sclerosis Complex 1 Protein , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Protein p53/analysis , Tumor Suppressor Proteins
17.
Sci Rep ; 7(1): 1917, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28507327

ABSTRACT

Recurrence following chemotherapy is observed in the majority of patients with pancreatic ductal adenocarcinoma (PDAC). Recent studies suggest that cancer stem cells (CSCs) may be involved in PDAC recurrence and metastasis. However, an efficient approach to targeting pancreatic CSCs remains to be established. Here we show that in cancer cells overexpressing the 67-kDa laminin receptor (67LR)-dependent cyclic GMP (cGMP) inducer, epigallocatechin-3-O-gallate (EGCG) and a phosphodiesterase 3 (PDE3) inhibitor in combination significantly suppressed the Forkhead box O3 and CD44 axis, which is indispensable for the CSC properties of PDAC. We confirmed that the EGCG and PDE3 inhibitor in combination strongly suppressed tumour formation and liver metastasis in vivo. We also found that a synthesized EGCG analog capable of inducing strong cGMP production drastically suppressed the CSC properties of PDAC and extended the survival period in vivo. In conclusion, the combination treatment of EGCG and a PDE3 inhibitor as a strong cGMP inducer could be a potential treatment candidate for the eradication of CSCs of PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Catechin/analogs & derivatives , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Phosphodiesterase 3 Inhibitors/pharmacology , Animals , Biomarkers, Tumor , Catechin/chemistry , Catechin/pharmacology , Cell Line, Tumor , Cyclic GMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 3/genetics , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Disease Models, Animal , Drug Synergism , Fluorescent Antibody Technique , Gene Expression , Humans , Mice , Phosphodiesterase 3 Inhibitors/chemistry , Xenograft Model Antitumor Assays
18.
DNA Res ; 13(4): 169-83, 2006 Aug 31.
Article in English | MEDLINE | ID: mdl-17082220

ABSTRACT

We have comprehensively identified the genes whose expressions are augmented in bone marrow-derived mononuclear cells (BMMC) from patients with Rheumatoid Arthritis (RA) as compared with BMMCs from Osteoarthritis (OA) patients, and named them AURA after augmented in RA. Both stepwise subtractive hybridization and microarray analyses were used to identify AURA genes, which were confirmed by northern blot analysis and/or reverse transcription polymerase chain reaction (RT-PCR). We also assessed their expression levels in individual patients by quantitative real-time RT-PCR. Of 103 AURA genes we have identified, the mRNA levels of the following 10 genes, which are somehow related to immune responses, were increased in many of the RA patients: AREG (=AURA9), FK506-binding protein 5 (FKBP5 = AURA45), C-type lectin superfamily member 9 (CLECSF9 = AURA24), tyrosylprotein sulfotransferase 1 (TPST1 = AURA52), lymphocyte G0/G1 switch gene (G0S2 = AURA8), chemokine receptor 4 (CXCR4 = AURA86), nuclear factor-kappa B (NF-kappaB = AURA25) and two genes of unknown function (FLJ11106 = AURA1, BC022398 = AURA2 and XM_058513 = AURA17). Since AREG was most significantly increased in many of the RA patients, we subjected it to further analysis and found that AREG-epidermal growth factor receptor signaling is highly activated in synovial cells isolated from RA patients, but not in OA synoviocytes. We propose that the expression profiling of these AURA genes may improve our understanding of the pathogenesis of RA.


Subject(s)
Arthritis, Rheumatoid/genetics , Bone Marrow Cells/metabolism , DNA, Complementary/isolation & purification , Gene Expression Profiling/methods , Amphiregulin , Cell Proliferation , Cells, Cultured , EGF Family of Proteins , Genes/genetics , Glycoproteins/physiology , Humans , Intercellular Signaling Peptides and Proteins/physiology , Leukocytes/metabolism , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Models, Biological , Oligonucleotide Array Sequence Analysis , Osteoarthritis/genetics , Synovial Fluid/cytology , Up-Regulation
19.
Oncogene ; 21(25): 4050-9, 2002 Jun 06.
Article in English | MEDLINE | ID: mdl-12037687

ABSTRACT

Tuberous sclerosis complex (TSC) is an autosomal dominant tumor predisposition syndrome characterized by benign proliferations (hamartomas). In the brain, individuals with TSC develop autism, mental retardation and seizures associated with focal cortical dysplasias, subependymal nodules, and subependymal giant cell astrocytomas (SEGAs). We hypothesize that dysregulated astrocyte function due to mutations in the tumor suppressor genes, TSC1 and TSC2, may contribute to the pathogenesis of these brain abnormalities. In this report, we demonstrate that mice heterozygous for a targeted defect in either the Tsc1 or Tsc2 genes(Tsc1+/- and Tsc2+/- mice) exhibit a 1.5-fold increase in the number of astrocytes in vivo. Whereas increased astrocyte numbers in vivo were suggestive of a proliferative advantage, Tsc2+/- primary astrocyte cultures did not show a cell-autonomous growth advantage, anchorage-independent growth, increased saturation density, or increased fluid-phase endocytosis compared to wild type astrocytes. Tsc2 null mouse embryonic fibroblasts (MEFs) however, did exhibit increased saturation density compared to Tsc2 wild type controls. In both Tsc2+/- astrocytes and Tsc2 null mouse embryonic fibroblasts, p27-Kip1 expression was decreased compared to wild type cells, and was reversed by tuberin re-expression in Tsc2-/- MEFs. In contrast, no change in endocytosis was observed upon tuberin re-expression in Tsc2-/- MEFs. Collectively, these results suggest Tsc heterozygosity may provide a non-cell-autonomous growth advantage for astrocytes that may involve p27-Kip1 expression.


Subject(s)
Astrocytes/pathology , Cell Cycle Proteins/metabolism , Proteins/genetics , Repressor Proteins/genetics , Tuberous Sclerosis/genetics , Tumor Suppressor Proteins/metabolism , Animals , Blotting, Western , Cell Division/physiology , Cyclin-Dependent Kinase Inhibitor p27 , Endocytosis , Fibroblasts/metabolism , Genes, Tumor Suppressor , Heterozygote , Loss of Heterozygosity , Mice , Mice, Knockout , Proteins/metabolism , Repressor Proteins/metabolism , Tuberous Sclerosis/metabolism , Tuberous Sclerosis/pathology , Tuberous Sclerosis Complex 1 Protein , Tuberous Sclerosis Complex 2 Protein
20.
Hum Gene Ther ; 16(12): 1367-76, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16390268

ABSTRACT

Tuberous sclerosis (TSC) is an autosomal dominant genetic disorder characterized by abnormalities in cellular migration, proliferation, and differentiation in many tissues. Benign hamartomas develop in multiple organs, believed to be caused by somatic mutation in addition to germ line mutation to cause loss of both alleles of either the TSC1 or TSC2 tumor suppressor gene, with resultant dysregulated growth due to loss of hamartin or tuberin function, respectively. This study focuses on detecting spontaneous lesions in a knockout mouse model of TSC2 by magnetic resonance imaging (MRI) and exploring the efficiency of introducing gene products into lesions, using transduced endothelial cells as gene vehicles. MRI was shown to be effective in detecting spontaneous lesions in multiple tissues as a means of assessing the prevalence of tumors. Tsc(2+/) heterozygous mice were screened at 12-24 months of age. MRI detected 100% of the renal lesions (cystadenomas, renal cell carcinomas) and 75% of the hepatic lesions (hemangiosarcomas), later identified by histology. Cell-mediated gene delivery was evaluated by immunohistochemical analysis of renal, hepatic, and lung lesions after intravenous delivery of MS1 mouse endothelial cells, transduced to express an enhanced form of green fluorescent protein (EGFP). Preliminary immunohistochemical analysis, using a polyclonal antibody to EGFP and a horseradish peroxidase-diaminobenzidine detection system, revealed these cells throughout liver, kidney, and lung sections from injected animals, organs that are frequently affected in TSC2 patients, as well as within the lesions themselves.


Subject(s)
Gene Transfer Techniques , Genetic Therapy/methods , Tuberous Sclerosis/pathology , Tuberous Sclerosis/therapy , Tumor Suppressor Proteins/genetics , Animals , Brain/pathology , Cell Line , Disease Models, Animal , Endothelial Cells , Genes, Tumor Suppressor , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Kidney Neoplasms/pathology , Lung/pathology , Magnetic Resonance Imaging , Mice , Mice, Knockout , Transduction, Genetic , Transgenes , Tuberous Sclerosis/genetics , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL