Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 102
Filter
1.
RNA ; 26(11): 1637-1653, 2020 11.
Article in English | MEDLINE | ID: mdl-32747606

ABSTRACT

Programmed cell death 4 (PDCD4) protein is a tumor suppressor that inhibits translation through the mTOR-dependent initiation factor EIF4A, but its functional role and mRNA targets in neurons remain largely unknown. Our work identified that PDCD4 is highly expressed in axons and dendrites of CNS and PNS neurons. Using loss- and gain-of-function experiments in cortical and dorsal root ganglia primary neurons, we demonstrated the capacity of PDCD4 to negatively control axonal growth. To explore PDCD4 transcriptome and translatome targets, we used Ribo-seq and uncovered a list of potential targets with known functions as axon/neurite outgrowth regulators. In addition, we observed that PDCD4 can be locally synthesized in adult axons in vivo, and its levels decrease at the site of peripheral nerve injury and before nerve regeneration. Overall, our findings demonstrate that PDCD4 can act as a new regulator of axonal growth via the selective control of translation, providing a target mechanism for axon regeneration and neuronal plasticity processes in neurons.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Axons/metabolism , Dendrites/metabolism , Peripheral Nerve Injuries/metabolism , RNA-Binding Proteins/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Cells, Cultured , Gain of Function Mutation , Gene Expression Profiling , Gene Regulatory Networks , Loss of Function Mutation , Male , Mice , PC12 Cells , Primary Cell Culture , Protein Biosynthesis , RNA-Binding Proteins/genetics , Rats , Up-Regulation
2.
J Natl Cancer Inst Monogr ; 2017(52)2017 11 01.
Article in English | MEDLINE | ID: mdl-29140488

ABSTRACT

Since 2007, the US National Cancer Institute (NCI) Office of Cancer Complementary and Alternative Medicine (OCCAM), together with the Cancer Institute of the China Academy of Chinese Medical Sciences (CICACMS), institutes at China Academy of Sciences and Chinese Academy of Medical Sciences, have engaged in collaborations on Chinese medicine (CM) and cancer research. Through these collaborations, CM drugs and compounds have been studied at NCI labs. This paper summarizes the discoveries and progress on these research projects, exploring the aspects of cancer prevention, botanical drug mechanisms of action and component analysis/quality control (QC), and anticancer activity screening. These and other related projects have been presented in various jointly convened workshops and have provided the backdrop for establishing a new organization, the International Consortium for CM and Cancer, to promote international collaborations in this field.


Subject(s)
Medicine, Chinese Traditional , Neoplasms/therapy , China , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Humans , Medicine, Chinese Traditional/methods , National Cancer Institute (U.S.) , Neoplasms/diagnosis , Neoplasms/prevention & control , Research , United States
3.
Cancer Prev Res (Phila) ; 9(7): 607-15, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27138790

ABSTRACT

There is a strong belief that garlic has medicinal properties and may even reduce the risk of developing certain cancers including those of the gastrointestinal tract. The chemopreventive effects of garlic may be attributed to the anti-inflammatory properties of the sulfur-containing constituents of garlic, which includes diallyl disulfide (DADS). Here, we demonstrate that DADS prevented colorectal tumorigenesis in a mouse model of colitis-induced colorectal cancer. Supplementation with 85 ppm of DADS (60 mg daily human equivalent dose) in the diet of FVB/N mice treated with chemical carcinogen azoxymethane (AOM) and colonic irritant dextran sodium sulfate (DSS) resulted in the reduction in tumor incidence, tumor number, and tumor burden by 21.54%, 47.3%, and 66.4%, respectively. Further analysis revealed that mice fed the DADS-supplemented diet resolved the initial DSS-induced inflammation faster than those on the control diet, preventing prolonged inflammation and cellular transformation. Subsequent mechanistic studies in vitro suggest that DADS chemopreventive effects are mediated through NF-κB signaling. When SW480 colorectal cancer cells were treated with DADS, NF-κB nuclear localization and activity were diminished. Interestingly, NF-κB suppression was found to be dependent on DADS inhibition of GSK-3ß, a positive regulator of NF-κB. Inhibition of GSK-3ß and loss of nuclear NF-κB activity were also observed in vivo in AOM/DSS-treated mice fed a diet supplemented with 85 ppm DADS. Our results indicate that DADS can prevent tumorigenesis by suppressing inflammation, a process largely involving GSK-3ß inhibition and consequential reduction in NF-κB nuclear localization. Cancer Prev Res; 9(7); 607-15. ©2016 AACR.


Subject(s)
Allyl Compounds/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Carcinogenesis/drug effects , Colorectal Neoplasms/pathology , Disulfides/pharmacology , Glycogen Synthase Kinase 3 beta/metabolism , Plant Extracts/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Colitis/chemically induced , Colitis/complications , Colorectal Neoplasms/etiology , Garlic/chemistry , Glycogen Synthase Kinase 3 beta/drug effects , Humans , Inflammation/chemically induced , Inflammation/pathology , Mice , NF-kappa B/drug effects , NF-kappa B/metabolism
4.
PLoS One ; 11(3): e0151643, 2016.
Article in English | MEDLINE | ID: mdl-26982744

ABSTRACT

The translation inhibitor and tumor suppressor Pdcd4 was reported to be lost in various tumors and put forward as prognostic marker in tumorigenesis. Decreased Pdcd4 protein stability due to PI3K-mTOR-p70S6K1 dependent phosphorylation of Pdcd4 followed by ß-TrCP1-mediated ubiquitination, and proteasomal destruction of the protein was characterized as a major mechanism contributing to the loss of Pdcd4 expression in tumors. In an attempt to identify stabilizers of Pdcd4, we used a luciferase-based high-throughput compatible cellular assay to monitor phosphorylation-dependent proteasomal degradation of Pdcd4 in response to mitogen stimulation. Following a screen of approximately 2000 compounds, we identified 1,2-bis(4-chlorophenyl)disulfide as a novel Pdcd4 stabilizer. To determine an initial structure-activity relationship, we used 3 additional compounds, synthesized according to previous reports, and 2 commercially available compounds for further testing, in which either the linker between the aryls was modified (compounds 2-4) or the chlorine residues were replaced by groups with different electronic properties (compounds 5 and 6). We observed that those compounds with alterations in the sulfide linker completely lost the Pdcd4 stabilizing potential. In contrast, modifications in the chlorine residues showed only minor effects on the Pdcd4 stabilizing activity. A reporter with a mutated phospho-degron verified the specificity of the compounds for stabilizing the Pdcd4 reporter. Interestingly, the active diaryl disulfides inhibited proliferation and viability at concentrations where they stabilized Pdcd4, suggesting that Pdcd4 stabilization might contribute to the anti-proliferative properties. Finally, computational modelling indicated that the flexibility of the disulfide linker might be necessary to exert the biological functions of the compounds, as the inactive compound appeared to be energetically more restricted.


Subject(s)
Apoptosis Regulatory Proteins/drug effects , RNA-Binding Proteins/drug effects , Sulfides/pharmacology , Tumor Suppressor Proteins/drug effects , Apoptosis Regulatory Proteins/metabolism , Cell Cycle , Cell Proliferation , HEK293 Cells , Humans , RNA-Binding Proteins/metabolism , Structure-Activity Relationship , Sulfides/chemistry , Tumor Suppressor Proteins/metabolism
5.
Mol Cell Biochem ; 406(1-2): 63-73, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25912550

ABSTRACT

Cryptotanshinone (CPT) is a natural compound extracted from herbal medicine that has been previously shown to possess antitumor properties in various types of human cancer cells. In the present study, we examined the potential role of CPT in the treatment of colorectal cancer. Using SW480, HCT116, and LOVO colorectal cancer cell lines, the effects of CPT on cell viability, apoptosis, and tumorigenicity were evaluated. The results showed that CPT significantly inhibited the growth and viability of SW480, HCT116, and LOVO cell lines by inducing apoptosis and prevented anchorage dependent growth on agar. In addition, CPT inhibited the activation of Signal transducer and activator of transcription 3 (Stat3) pathways in colorectal cancer cells. Stat3 is a transcription factor that mediates the expression of various genes associated with many cellular processes, such as inflammation and cell growth, and has been shown to promote several cancer types, including colorectal cancer. These findings indicate that CPT may be a potential candidate for the treatment and prevention of colorectal cancer in part by inhibiting the activation of Stat3.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Cell Proliferation/drug effects , Colorectal Neoplasms/drug therapy , Phenanthrenes/pharmacology , STAT3 Transcription Factor/metabolism , Apoptosis , Cell Survival/drug effects , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Drug Screening Assays, Antitumor , HCT116 Cells , Humans , Inhibitor of Apoptosis Proteins/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , Survivin
6.
Mol Carcinog ; 54(9): 669-78, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25845339

ABSTRACT

An important characteristic of cancer is that the disease can overcome the surveillance of the immune system. A possible explanation for this resistance arises from the ability of tumor cells to block the tumoricidal activity of host immune cells such as natural killer (NK) cells by inducing the localized accumulation of regulatory T (Treg) cells. Evidence exists that components in commonly consumed foods including vitamins A, D, and E, water-soluble constituents of mushrooms, polyphenolics in fruits and vegetables, and n-3 fatty acids in fish oil can modulate NK cell activities, Treg cell properties, and the interactions between those two cell types. Thus, it is extremely important for cancer prevention to understand the involvement of dietary components with the early stage dynamics of interactions among these immune cells. This review addresses the potential significance of diet in supporting the function of NK cells, Treg cells, and the balance between those two cell types, which ultimately results in decreased cancer risk.


Subject(s)
Diet , Killer Cells, Natural/immunology , Neoplasms/immunology , Neoplasms/prevention & control , T-Lymphocytes, Regulatory/immunology , Animals , Cytokines/immunology , Fatty Acids, Omega-3/immunology , Humans , Neoplasms/diet therapy , Polyphenols/immunology , Vitamin A/immunology , Vitamin D/immunology
7.
Mol Nutr Food Res ; 59(4): 795-806, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25641932

ABSTRACT

SCOPE: Aim of the study was to identify and monitor metabolite markers of dry bean consumption in parallel human and mouse studies that each had shown chemopreventive effects of dry bean consumption on colorectal neoplasia risk. METHODS AND RESULTS: Using LC/mass spectroscopy ± ESI and GC/mass spectroscopy, serum metabolites of dry beans were measured in 46 men before and after a 4-week dry bean enriched diet (250 g/day) and 12 mice that received a standardized diet containing either 0 or 10% navy bean ethanol extract for 6 weeks; we also investigated fecal metabolites in the mice. The serum metabolites identified in these controlled feeding studies were then investigated in 212 polyp-free participants from the Polyp Prevention Trial who self-reported either increased (≥+31 g/day from baseline), high dry bean intake of ≥42 g/day in year 3 or low, unchanged dry bean consumption of <8 g/day; serum was analyzed from baseline and year 3. Serum pipecolic acid and S-methyl cysteine were elevated after dry bean consumption in human and mouse studies and reflected dry bean consumption in the Polyp Prevention Trial. CONCLUSION: Serum levels of pipecolic acid and S-methyl cysteine are useful biomarkers of dry bean consumption.


Subject(s)
Biomarkers/blood , Cysteine/analogs & derivatives , Diet , Fabaceae , Pipecolic Acids/blood , Adult , Aged , Animals , Anticarcinogenic Agents/administration & dosage , Chromatography, Liquid , Colorectal Neoplasms/prevention & control , Cross-Over Studies , Cysteine/blood , Feces/chemistry , Gastrointestinal Microbiome , Humans , Intestinal Mucosa/metabolism , Intestines/microbiology , Male , Mass Spectrometry , Metabolomics , Mice , Mice, Inbred Strains , Middle Aged , Plant Extracts/administration & dosage
8.
Biochim Biophys Acta ; 1853(1): 222-32, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25447673

ABSTRACT

Valosin-containing protein (VCP or p97), a member of the AAA family (ATPases associated with diverse cellular activities), plays a key role in many important cellular activities. A genetic deficiency of VCP can cause inclusion body myopathy associated with Paget's disease of bone and frontotemporal dementia (IBMPFD). Previous studies showed that the VCP N domain is essential for the regulation of nuclear entry of VCP. Here we report that IBMPFD mutations, which are mainly located in the N domain, suppress the nuclear entry of VCP. Moreover, the peptide sequence G780AGPSQ in the C-terminal region regulates the retention of VCP in the nucleus. A mutant lacking this sequence can increase the nuclear distribution of IBMPFD VCP, suggesting that this sequence is a potential molecular target for correcting the deficient nucleocytoplasmic shuttling of IBMPFD VCP proteins.


Subject(s)
Adenosine Triphosphatases/metabolism , Cell Cycle Proteins/metabolism , Cell Nucleus/metabolism , Active Transport, Cell Nucleus , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/physiology , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Frontotemporal Dementia/genetics , HEK293 Cells , Humans , Muscular Dystrophies, Limb-Girdle/genetics , Myositis, Inclusion Body/genetics , Osteitis Deformans/genetics , Protein Structure, Tertiary , Valosin Containing Protein
9.
Mol Carcinog ; 54(10): 1096-109, 2015 Oct.
Article in English | MEDLINE | ID: mdl-24838344

ABSTRACT

Colorectal cancer, a leading cause of cancer death, has been linked to inflammation and obesity. Berberine, an isoquinoline alkaloid, possesses anti-inflammatory, anti-diabetes and anti-tumor properties. In the azoxymethane initiated and dextran sulfate sodium (AOM/DSS) promoted colorectal carcinogenesis mouse model, berberine treated mice showed a 60% reduction in tumor number (P = 0.009), a 48% reduction in tumors <2 mm, (P = 0.05); 94% reduction in tumors 2-4 mm, (P = 0.001), and 100% reduction in tumors >4 mm (P = 0.02) compared to vehicle treated mice. Berberine also decreased AOM/DSS induced Ki-67 and COX-2 expression. In vitro analysis showed that in addition to its anti-proliferation activity, berberine also induced apoptosis in colorectal cancer cell lines. Berberine activated AMP-activated protein kinase (AMPK), a major regulator of metabolic pathways, and inhibited mammalian target of rapamycin (mTOR), a downstream target of AMPK. Furthermore, 4E-binding protein-1 and p70 ribosomal S6 kinases, downstream targets of mTOR, were down regulated by berberine treatment. Berberine did not affect Liver kinase B1 (LKB1) activity or the mitogen-activated protein kinase pathway. Berberine inhibited Nuclear Factor kappa-B (NF-κB) activity, reduced the expression of cyclin D1 and survivin, induced phosphorylation of p53 and increased caspase-3 cleavage in vitro. Berberine inhibition of mTOR activity and p53 phosphorylation was found to be AMPK dependent, while inhibition NF-κB was AMPK independent. In vivo, berberine also activated AMPK, inhibited mTOR and p65 phosphorylation and activated caspase-3 cleavage. Our data suggests that berberine suppresses colon epithelial proliferation and tumorigenesis via AMPK dependent inhibition of mTOR activity and AMPK independent inhibition of NF-κB.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Berberine/pharmacology , Carcinogenesis/drug effects , Colon/drug effects , Colorectal Neoplasms/drug therapy , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Azoxymethane/pharmacology , Carcinogenesis/metabolism , Carcinogenesis/pathology , Caspase 3/metabolism , Cell Line, Tumor , Colon/metabolism , Colon/pathology , Colorectal Neoplasms/metabolism , Cyclin D1/metabolism , Cyclooxygenase 2/metabolism , Female , HCT116 Cells , Humans , Mice , NF-kappa B/metabolism , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/metabolism , eIF-2 Kinase/metabolism
10.
Cancer Cell Int ; 14: 75, 2014.
Article in English | MEDLINE | ID: mdl-25324692

ABSTRACT

BACKGROUND: The small molecule NSC676914A was previously identified as an NF-κB inhibitor in TPA-stimulated HEK293 cells (Mol Can Ther 8:571-581, 2009). We hypothesized that this effect would also be seen in ovarian cancer cells, and serve as its mechanism of cytotoxicity. OVCAR3 and HEK293 cell lines stably containing a NF-κB luciferase reporter gene were generated. METHODS: Levels of NF-κB activity were assessed by luciferase reporter assays, after stimulation with LPA, LPS, TPA, and TNFα, in the presence or absence of a known NF-κB inhibitor or NSC676914A, and cytotoxicity was measured. RESULTS: NSC676914A was toxic to both OVCAR3 and HEK293 cells. We also investigated the cytotoxicity of NSC676914A on a panel of lymphoma cell lines with well characterized mutations previously shown to determine sensitivity or resistance to NF-κB inhibition. The compound did not show predicted patterns of effects on NF-κB activity in either lymphoma, ovarian or HEK293 cell lines. In HEK293 cells, the small molecule inhibited NF-κB when cells were stimulated, while in OVCAR3 cells it only partially inhibited NF-κB. Interestingly, we observed rescue of cell death with ROS inhibition. CONCLUSIONS: The current study suggests that the effect of NSC676914A on NF-κB depends on cell type and the manner in which the pathway is stimulated. Furthermore, as it is similarly toxic to lymphoma, OVCAR3 and HEK293 cells, NSC676914A shows promising NF-κB-independent anti-cancer activity in ovarian tumor cells.

11.
Carcinogenesis ; 35(12): 2778-86, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25280562

ABSTRACT

Sporadic and non-hereditary mutations account for the majority of colorectal cancers (CRC). After the loss of adenomatous polyposis coli (APC) function and activation of the ß-catenin/LEF signaling pathway, activating mutations in Kras are major drivers of sporadic CRC. Preventing the outgrowth of cells that develop sporadic mutations will decrease CRC. Resveratrol, a naturally occurring polyphenolic compound has anti-inflammatory, anti-oxidant and anti-cancer activities. We used a genetically engineered mouse model for sporadic CRC where the APC locus is knocked out and Kras is activated specifically in the distal colon to determine the effects of resveratrol on preventing and treating CRC. Feeding mice a diet supplemented with 150 or 300 ppm resveratrol (105 and 210mg daily human equivalent dose, respectively) before tumors were visible by colonoscopy resulted in a 60% inhibition of tumor production. In the 40% of mice that did develop tumors Kras expression was lost in the tumors. In a therapeutic assay where tumors were allowed to develop prior to treatment, feeding tumor bearing mice with resveratrol resulted in a complete remission in 33% of the mice and a 97% decrease in tumor size in the remaining mice. Analysis of miRNA expression in non-tumoral and tumoral colonic tissue of resveratrol treated mice showed an increased expression of miR-96, a miRNA previously shown to regulate Kras translation. These data indicate that resveratrol can prevent the formation and growth of colorectal tumors by downregulating Kras expression.


Subject(s)
Adenomatous Polyposis Coli Protein/physiology , Anticarcinogenic Agents/therapeutic use , Cell Transformation, Neoplastic/drug effects , Colorectal Neoplasms/prevention & control , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Stilbenes/therapeutic use , Animals , Blotting, Western , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Colorectal Neoplasms/etiology , Colorectal Neoplasms/pathology , Disease Models, Animal , Female , Humans , Immunoenzyme Techniques , Male , Mice , Mice, Knockout , Mutation/genetics , Proto-Oncogene Proteins p21(ras)/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Resveratrol , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
12.
Mar Drugs ; 12(8): 4593-601, 2014 Aug 21.
Article in English | MEDLINE | ID: mdl-25196934

ABSTRACT

A cell-based high-throughput screen that assessed the cellular stability of a tumor suppressor protein PDCD4 (Programmed cell death 4) was used to identify a new guanidine-containing marine alkaloid mirabilin K (3), as well as the known compounds mirabilin G (1) and netamine M (2). The structures of these tricyclic guanidine alkaloids were established from extensive spectroscopic analyses. Compounds 1 and 2 inhibited cellular degradation of PDCD4 with EC50 values of 1.8 µg/mL and 2.8 µg/mL, respectively. Mirabilin G (1) and netamine M (2) are the first marine natural products reported to stabilize PDCD4 under tumor promoting conditions.


Subject(s)
Alkaloids/chemistry , Alkaloids/pharmacology , Apoptosis Regulatory Proteins/metabolism , Guanidine/chemistry , Guanidine/pharmacology , Porifera/chemistry , RNA-Binding Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Animals , HEK293 Cells , Humans , Oleanolic Acid/analogs & derivatives , Oleanolic Acid/chemistry , Saponins/chemistry
14.
Biochem Pharmacol ; 88(3): 313-21, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24513322

ABSTRACT

Deregulation of the phosphatidylinositol 3-kinase (PI3K)-Akt-mammalian target of rapamycin (mTOR)-70kDa ribosomal protein S6 kinase 1 (p70(S6K)) pathway is commonly observed in many tumors. This pathway controls proliferation, survival, and translation, and its overactivation is associated with poor prognosis for tumor-associated survival. Current efforts focus on the development of novel inhibitors of this pathway. In a cell-based high-throughput screening assay of 15,272 pure natural compounds, we identified pomiferin triacetate as a potent stabilizer of the tumor suppressor programmed cell death 4 (Pdcd4). Mechanistically, pomiferin triacetate appeared as a general inhibitor of the PI3K-Akt-mTOR-p70(S6K) cascade. Interference with this pathway occurred downstream of Akt but upstream of p70(S6K). Specifically, mTOR kinase emerged as the molecular target of pomiferin triacetate, with similar activities against mTOR complexes 1 and 2. In an in vitro mTOR kinase assay pomiferin triacetate dose-dependently inhibited mTOR with an IC50 of 6.2 µM. Molecular docking studies supported the interaction of the inhibitor with the catalytic site of mTOR. Importantly, pomiferin triacetate appeared to be highly selective for mTOR compared to a panel of 17 lipid and 50 protein kinases tested. As a consequence of the mTOR inhibition, pomiferin triacetate efficiently attenuated translation. In summary, pomiferin triacetate emerged as a novel and highly specific mTOR inhibitor with strong translation inhibitory effects. Thus, it might be an interesting lead structure for the development of mTOR- and translation-targeted anti-tumor therapies.


Subject(s)
Isoflavones/pharmacology , Protein Biosynthesis/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , Apoptosis Regulatory Proteins/metabolism , HEK293 Cells , Humans , MCF-7 Cells , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Molecular Docking Simulation , Multiprotein Complexes/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA-Binding Proteins/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , TOR Serine-Threonine Kinases/metabolism
15.
Carcinogenesis ; 35(5): 1177-84, 2014 May.
Article in English | MEDLINE | ID: mdl-24503444

ABSTRACT

Sulfiredoxin (Srx), the exclusive enzyme that reduces the hyperoxidized inactive form of peroxiredoxins (Prxs), has been found highly expressed in several types of human skin cancer. To determine whether Srx contributed to skin tumorigenesis in vivo, Srx null mice were generated on an FVB background. Mouse skin tumorigenesis was induced by a 7,12-dimethylbenz[α]anthracene/12-O-tetradecanoylphorbol-13-acetate (DMBA/TPA) protocol. We found that the number, volume and size of papillomas in Srx(-/-) mice were significantly fewer compared with either wild-type (Wt) or heterozygous (Het) siblings. Histopathological analysis revealed more apoptotic cells in tumors from Srx(-/-) mice. Mechanistic studies in cell culture revealed that Srx was stimulated by TPA in a redox-independent manner. This effect was mediated transcriptionally through the activation of mitogen-activated protein kinase and Jun-N-terminal kinase. We also demonstrated that Srx was capable of reducing hyperoxidized Prxs to facilitate cell survival under oxidative stress conditions. These findings suggested that loss of Srx protected mice, at least partially, from DMBA/TPA-induced skin tumorigenesis. Therefore, Srx has an oncogenic role in skin tumorigenesis and targeting Srx may provide novel strategies for skin cancer prevention or treatment.


Subject(s)
Cell Transformation, Neoplastic/genetics , Oxidoreductases Acting on Sulfur Group Donors/genetics , Skin Neoplasms/genetics , Skin/metabolism , Skin/pathology , 9,10-Dimethyl-1,2-benzanthracene/adverse effects , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Line , Cell Proliferation , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Gene Expression Regulation/drug effects , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Mice, Knockout , Mitogen-Activated Protein Kinases/metabolism , Oxidation-Reduction , Oxidoreductases Acting on Sulfur Group Donors/metabolism , Skin/drug effects , Skin Neoplasms/chemically induced , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Tetradecanoylphorbol Acetate/adverse effects , Transcriptional Activation/drug effects
16.
Carcinogenesis ; 35(7): 1469-80, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24413684

ABSTRACT

PDCD4 is a tumor suppressor induced by apoptotic stimuli that regulates both translation and transcription. Previously, we showed that overexpression of PDCD4 leads to decreased anchorage-independent growth in glioblastoma (GBM)-derived cell lines and decreased tumor growth in a GBM xenograft model. In inflammatory cells, PDCD4 stimulates tumor necrosis factor-induced activation of the transcription factor NF-κB, an oncogenic driver in many cancer sites. However, the effect of PDCD4 on NF-κB transcriptional activity in most cancers including GBM is still unknown. We studied the effect of PDCD4 on NF-κB-dependent transcriptional activity in GBM by stably overexpressing PDCD4 in U251 and LN229 cells. Stable PDCD4 expression inhibits NF-κB transcriptional activation measured by a luciferase reporter. The molecular mechanism by which PDCD4 inhibits NF-κB transcriptional activation does not involve inhibited expression of NF-κB p65 or p50 proteins. PDCD4 does not inhibit pathways upstream of NF-κB including the activation of IKKα and IKKß kinases or degradation of IκBα, events needed for nuclear transport of p65 and p50. PDCD4 overexpression does inhibit localization of p65 but not p50 in the nucleus. PDCD4 protein interacts preferentially with p65 protein as shown by co-immunoprecipitation and confocal imaging. PDCD4 overexpression inhibits the mRNA expression of two NF-κB target genes in a p65-dependent manner. These results suggest that PDCD4 can significantly inhibit NF-κB activity in GBM cells by a mechanism that involves direct or indirect protein-protein interaction independent of the expected mRNA-selective translational inhibition. These findings offer novel opportunities for NF-κB-targeted interventions to prevent or treat cancer.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , NF-kappa B p50 Subunit/metabolism , RNA-Binding Proteins/genetics , Transcription Factor RelA/metabolism , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Blotting, Western , Cell Movement , Cell Proliferation , Chromatin Immunoprecipitation , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Immunoprecipitation , NF-kappa B p50 Subunit/genetics , RNA, Messenger/genetics , RNA-Binding Proteins/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factor RelA/genetics , Transcription, Genetic , Tumor Cells, Cultured
17.
Cancer Res ; 73(17): 5473-84, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23824743

ABSTRACT

Analysis of the Polyp Prevention Trial showed an association between an isorhamnetin-rich diet and a reduced risk of advanced adenoma recurrence; however, the mechanism behind the chemoprotective effects of isorhamnetin remains unclear. Here, we show that isorhamnetin prevents colorectal tumorigenesis of FVB/N mice treated with the chemical carcinogen azoxymethane and subsequently exposed to colonic irritant dextran sodium sulfate (DSS). Dietary isorhamnetin decreased mortality, tumor number, and tumor burden by 62%, 35%, and 59%, respectively. MRI, histopathology, and immunohistochemical analysis revealed that dietary isorhamnetin resolved the DSS-induced inflammatory response faster than the control diet. Isorhamnetin inhibited AOM/DSS-induced oncogenic c-Src activation and ß-catenin nuclear translocation, while promoting the expression of C-terminal Src kinase (CSK), a negative regulator of Src family of tyrosine kinases. Similarly, in HT-29 colon cancer cells, isorhamnetin inhibited oncogenic Src activity and ß-catenin nuclear translocation by inducing expression of csk, as verified by RNA interference knockdown of csk. Our observations suggest the chemoprotective effects of isorhamnetin in colon cancer are linked to its anti-inflammatory activities and its inhibition of oncogenic Src activity and consequential loss of nuclear ß-catenin, activities that are dependent on CSK expression.


Subject(s)
Colorectal Neoplasms/prevention & control , Phytotherapy , Plant Extracts/pharmacology , Quercetin/analogs & derivatives , beta Catenin/metabolism , src-Family Kinases/metabolism , Animals , Apoptosis/drug effects , Azoxymethane/toxicity , Blotting, Western , CSK Tyrosine-Protein Kinase , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Colorectal Neoplasms/chemically induced , Colorectal Neoplasms/metabolism , Dextran Sulfate , Humans , Immunoenzyme Techniques , Male , Mice , Protein Transport , Quercetin/pharmacology , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , beta Catenin/genetics , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/genetics
18.
Carcinogenesis ; 34(6): 1403-10, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23393226

ABSTRACT

Sulfiredoxin (Srx) is the enzyme that reduces the hyperoxidized inactive form of peroxiredoxins. To study the function of Srx in carcinogenesis in vivo, we tested whether loss of Srx protects mice from cancer development. Srx null mice were generated and colon carcinogenesis was induced by an azoxymethane (AOM) and dextran sulfate sodium (DSS) protocol. Compared with either wild-type (Wt) or heterozygotes, Srx(-/-) mice had significantly reduced rates in both tumor multiplicity and volume. Mechanistic studies reveal that loss of Srx did not alter tumor cell proliferation; however, increased apoptosis and decreased inflammatory cell infiltration were obvious in tumors from Srx null mice compared with those from Wt control. In addition to the AOM/DSS model, examination of Srx expression in human reveals a tissue-specific expression pattern. Srx expression was also demonstrated in tumors from colorectal cancer patients and the levels of expression were associated with patients' clinic stages. These data provide the first in vivo evidence that loss of Srx renders mice resistant to AOM/DSS-induced colon carcinogenesis, suggesting that Srx has a critical oncogenic role in cancer development, and Srx may be used as a marker for human colon cancer pathogenicity.


Subject(s)
Cell Transformation, Neoplastic , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Oxidoreductases Acting on Sulfur Group Donors/metabolism , Animals , Apoptosis , Azoxymethane , Cell Line, Tumor , Cell Proliferation , Colonic Neoplasms/chemically induced , Dextran Sulfate , Genotype , Humans , Lung Neoplasms , Macrophages/immunology , Mice , Mice, Knockout , Oxidoreductases Acting on Sulfur Group Donors/genetics , Peroxiredoxins/metabolism
19.
J Cell Physiol ; 228(7): 1536-50, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23280476

ABSTRACT

Recent studies have suggested that changes in serum phosphate levels influence pathological states associated with aging such as cancer, bone metabolism, and cardiovascular function, even in individuals with normal renal function. The causes are only beginning to be elucidated but are likely a combination of endocrine, paracrine, autocrine, and cell autonomous effects. We have used an integrated quantitative biology approach, combining transcriptomics and proteomics to define a multi-phase, extracellular phosphate-induced, signaling network in pre-osteoblasts as well as primary human and mouse mesenchymal stromal cells. We identified a rapid mitogenic response stimulated by elevated phosphate that results in the induction of immediate early genes including c-fos. The mechanism of activation requires FGF receptor signaling followed by stimulation of N-Ras and activation of AP-1 and serum response elements. A distinct long-term response also requires FGF receptor signaling and results in N-Ras activation and expression of genes and secretion of proteins involved in matrix regulation, calcification, and angiogenesis. The late response is synergistically enhanced by addition of FGF23 peptide. The intermediate phase results in increased oxidative phosphorylation and ATP production and is necessary for the late response providing a functional link between the phases. Collectively, the results define elevated phosphate, as a mitogen and define specific mechanisms by which phosphate stimulates proliferation and matrix regulation. Our approach provides a comprehensive understanding of the cellular response to elevated extracellular phosphate, functionally connecting temporally coordinated signaling, transcriptional, and metabolic events with changes in long-term cell behavior.


Subject(s)
Mesenchymal Stem Cells/metabolism , Phosphates/metabolism , Signal Transduction/physiology , 3T3 Cells , Adenosine Triphosphate/biosynthesis , Animals , Cells, Cultured , Computational Biology , Extracellular Space/metabolism , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/metabolism , GTP-Binding Proteins/metabolism , Gene Expression , Genes, Immediate-Early , Genes, fos , Genes, ras , Humans , Mice , Neovascularization, Physiologic , Osteoblasts/metabolism , Promoter Regions, Genetic , Proteins/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Transcription Factor AP-1/metabolism , Transcription Factors/metabolism
20.
Gut ; 62(8): 1179-86, 2013 Aug.
Article in English | MEDLINE | ID: mdl-22698650

ABSTRACT

OBJECTIVES: Screening colonoscopy to monitor for early colitis-associated colon cancer (CAC) is difficult due to the aberrant mucosal patterns associated with long-standing colitis. The aim of this study was to develop a rapid fluorescent detection method for use during colonoscopy for improving the detection of CAC utilising a topically applied enzymatically activatable probe (gGlu-HMRG) which fluoresces in the presence of γ-glutamyltranspeptidase (GGT), an enzyme associated with cancer. METHODS: Expression of GGT in colon cell lines was examined with fluorescence microscopy and flow cytometry. A mouse model (azoxymethane/dextran sulphate sodium) of CAC was used and mice were examined with white light and fluorescence colonoscopy before and after topical gGlu-HMRG administration. RESULTS: Expression of GGT, although variable, was higher in human colon cancer cells than normal human colon cells. Using fluorescence colonoscopy in mice, gGlu-HMRG fluorescent lesions were detected 5 min after topical administration and fluorescence persisted for at least 30 min. Fluorescence guided biopsy revealed all fluorescent lesions that contained cancer or dysplasia (n=16), whereas three out of 12 non-fluorescent lesions contained low grade dysplasia and others did not contain neoplastic histology. Microscopic inflammatory infiltration also had variable fluorescence but in general was much lower (∼10-fold) in signal than cancer. Repeat fluorescence endoscopy allowed individual tumours to be monitored. CONCLUSION: These results suggest that gGlu-HMRG can improve endoscopic detection of CAC with a higher target to background ratio than conventional white light colonoscopy. This could be of benefit to patients with long-standing colitis who must undergo repeated screening colonoscopies.


Subject(s)
Colitis/complications , Colonic Neoplasms/diagnosis , Colonic Neoplasms/etiology , Adenocarcinoma/diagnosis , Adenocarcinoma/enzymology , Adenocarcinoma/etiology , Administration, Topical , Animals , Biomarkers, Tumor/metabolism , Biopsy , Colon/enzymology , Colonic Neoplasms/enzymology , Colonoscopy/methods , Disease Models, Animal , Early Detection of Cancer/methods , Fluorescent Dyes/administration & dosage , Humans , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence/methods , Precancerous Conditions/diagnosis , Precancerous Conditions/enzymology , Precancerous Conditions/etiology , Tumor Cells, Cultured , gamma-Glutamyltransferase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...