Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Pharm Res ; 38(11): 1807-1820, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34773184

ABSTRACT

PURPOSE: Diabetic nephropathy (DN), one of severe diabetic complications in the diabetes, is the main cause of end stage renal disease (ESRD). Notably, the currently available medications used to treat DN remain limited. Here, we determined whether berberine (BBR) could enhance the anti-diabetic nephropathy activities of metformin (Met) and explored its possible mechanisms. METHOD: The anti-diabetic nephropathy properties were systematically analyzed in the diabetic db/db mice treated with Met, BBR or with combination of Met and BBR. RESULTS: We found that both single Met and BBR treatments, and combination therapy could lower blood glucose, and ameliorate insulin resistance. The improvement of lipids metabolism by co-administration was more evident, as indicated by reduced serum cholesterol and less fat accumulation in the liver. Further, it was found that Met and BBR treatments, and co-administration could attenuate the progression of DN. However, anti-diabetic nephropathy activities of Met were enhanced when combined with BBR, as evidenced by improved renal function and histological abnormalities of diabetic kidney. Mechanistically, BBR enhanced renal-protective effects of Met primarily through potently promoting expression of Trib1, which subsequently downregulated the increased protein levels of CCAAT/enhancer binding protein α (C/EBPα), and eventually inhibited fatty synthesis proteins and nuclear factor kappa-B (NF-κB) signaling. CONCLUSION: Our data provide novel insight that co-administration of BBR and Met exerts a preferable activity of anti-diabetic nephropathy via collectively enhancing lipolysis and inhibiting inflammation. Combination therapy with these two drugs may provide an effective therapeutic strategy for the medical treatment of diabetic nephropathy.


Subject(s)
Berberine/pharmacology , Diabetic Nephropathies/drug therapy , Intracellular Signaling Peptides and Proteins/agonists , Metformin/pharmacology , Nephritis/drug therapy , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Berberine/therapeutic use , Diabetic Nephropathies/genetics , Diabetic Nephropathies/immunology , Disease Models, Animal , Disease Progression , Drug Synergism , Drug Therapy, Combination/methods , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Kidney/drug effects , Kidney/immunology , Kidney/pathology , Male , Metformin/therapeutic use , Mice , Nephritis/genetics , Nephritis/immunology , Nephritis/pathology , Protein Serine-Threonine Kinases/metabolism
2.
Int J Oncol ; 57(4): 967-979, 2020 10.
Article in English | MEDLINE | ID: mdl-32945379

ABSTRACT

Drug resistance is a major obstacle in the therapy of malignant tumors, including non­small cell lung cancer (NSCLC). Long non­coding RNAs (lncRNAs) have been demonstrated to be involved in chemoresistance. The present study aimed to investigate the role of lung cancer­associated transcript 1 (LUCAT1) in cisplatin (DDP) resistance in NSCLC. By using reverse transcription­quantitative polymerase chain reaction (RT­qPCR), it was found that the expression of LUCAT1 was elevated and that of microRNA­514a­3p (miR­514a­3p) was decreased in DDP­resistant NSCLC tissues and cells. Functionally, LUCAT1 upregulation enhanced cisplatin resistance by promoting the viability, autophagy and metastasis, and inhibiting the apoptosis of NSCLC cells, as demonstrated by Cell Counting kit­8 (CCK­8) assay, western blot analysis, Transwell assay and flow cytometric analysis. LUCAT1 was identified as a sponge of miR­514a­3p and uncoordinated­51­like kinase 1 (ULK1) was proven to be a target gene of miR­514a­3p by bioinformatics analysis, dual­luciferase reporter assay and RNA immunoprecipitation (RIP) assay. The enhancing effect of miR­514a­3p on cisplatin sensitivity was reversed by the elevation of LUCAT1. ULK1 knockdown suppressed cisplatin resistance, while this effect was attenuated by miR­514a­3p inhibition. Moreover, LUCAT1 positively regulated ULK1 expression by targeting miR­514a­3p. In addition, LUCAT1 knockdown suppressed tumor growth in vivo. On the whole, the findings of the present study demonstrate that LUCAT1 contributes to the resistance of NSCLC cells to cisplatin by regulating the miR­514a­3p/ULK1 axis, elucidating a novel regulatory network in cisplatin resistance in NSCLC.


Subject(s)
Autophagy-Related Protein-1 Homolog/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Cisplatin/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Lung Neoplasms/metabolism , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , Aged , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , Female , Humans , Intracellular Signaling Peptides and Proteins/agonists , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Mice , Mice, Nude , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Middle Aged , RNA, Long Noncoding/genetics , Signal Transduction , Up-Regulation , Xenograft Model Antitumor Assays
3.
ACS Chem Biol ; 14(9): 1964-1971, 2019 09 20.
Article in English | MEDLINE | ID: mdl-31497942

ABSTRACT

The plant hormone abscisic acid (ABA) regulates the development of various plant organs including seeds, roots, and fruits, and significantly contributes to abiotic stress responses, especially to drought. Since recent climate changes are adversely affecting crop cultivation, enhancement of plant stress tolerance by regulation of ABA signaling would be an important strategy. In the plant genome, ABA receptors are encoded by multiple genes constituting three subfamilies; however, functional differences among them remain unclear. To enhance desired effects of ABA, the biological functions of the receptor family warrant clarification. This study aimed to determine the functional differences among ABA receptors in plants. We screened small-molecule ligands binding to specific receptors, using a chemical array. In vitro evaluation of hit compounds using 11 Arabidopsis ABA receptors revealed that (+)-3'-alkyl ABAs served as agonists for different receptors depending on the length of their 3'-alkyl chains. Combinatorial in vitro and physiological effects of these compounds on the stomata, seeds, and seedlings indicated that, along with subfamily III, receptors of subfamily II are important to induce strong drought responses. Among (+)-3'-alkyl ABAs assessed herein, (+)-3'-butyl ABA induced a transcriptional response and stomatal closure but only slightly inhibited seed germination and growth, suggesting that it enhances drought tolerance. In silico docking simulation and site-directed mutagenesis revealed the amino acid residues contributing to the selective agonist activity of the (+)-3'-alkyl ABAs. These results provide novel insights into the structure and biological effects of 3'-derivatives of ABA and a basis for agrochemical development.


Subject(s)
Abscisic Acid/analogs & derivatives , Arabidopsis Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Transport Proteins/metabolism , Receptors, Cell Surface/metabolism , Abscisic Acid/metabolism , Arabidopsis/chemistry , Arabidopsis/metabolism , Arabidopsis Proteins/agonists , Arabidopsis Proteins/antagonists & inhibitors , Arabidopsis Proteins/genetics , Germination/drug effects , Intracellular Signaling Peptides and Proteins/agonists , Membrane Transport Proteins/agonists , Molecular Docking Simulation , Molecular Structure , Mutagenesis, Site-Directed , Mutation , Phosphoprotein Phosphatases/antagonists & inhibitors , Plant Leaves/metabolism , Protein Binding , RNA, Messenger/metabolism , Receptors, Cell Surface/agonists , Receptors, Cell Surface/genetics , Structure-Activity Relationship
4.
Trends Pharmacol Sci ; 40(10): 725-734, 2019 10.
Article in English | MEDLINE | ID: mdl-31500846

ABSTRACT

The urotensinergic system, comprised of a G protein-coupled receptor (UT) and two endogenous ligands named urotensin II (UII) and urotensin II-related peptide (URP), has garnered significant attention due to its involvement in the initiation and/or the evolution of various diseases. Accordingly, multiple studies using animal models have demonstrated that UT antagonists may have utility as potential therapeutic agents for treating atherosclerosis, pulmonary arterial hypertension, heart failure, and cancer. Unfortunately, clinical investigations of UT antagonist candidates showed limited efficacy in humans. This system, which has yet to be effectively targeted, therefore remains to be therapeutically exploited. Here, we discuss various hypotheses that could explain the in vivo failure of UT antagonists.


Subject(s)
Peptide Hormones/agonists , Peptide Hormones/antagonists & inhibitors , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Urotensins/agonists , Urotensins/antagonists & inhibitors , Animals , Drug Delivery Systems , Humans , Intracellular Signaling Peptides and Proteins/agonists , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/metabolism , Ligands , Peptide Hormones/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Urotensins/metabolism
5.
Biochem Biophys Res Commun ; 506(1): 114-121, 2018 11 17.
Article in English | MEDLINE | ID: mdl-30340826

ABSTRACT

Di-2-pyridylketone-4,4-dimethyl-3-thiosemicarbazone (Dp44mT), as the novel iron chelator, has been reported to inhibit the tumorigenesis and progression of various cancer cells. However, whether Dp44mT has anticancer effects in colon cancer cells is still unknown. Here, we investigated the antitumor action of Dp44mT in colon cancer and its underlying mechanisms, and the connections between Dp44mT and N-myc downstream-regulated genes 1(Ndrg1). We used cell viability, migration and invasion assay, flow cytometry, western blot and qRT-PCR to examine the anticancer effects of Dp44mT and Ndrg1. We found that Dp44mT suppressed cell viability, migration, invasion and induced apoptosis of colon cancer cells and over-expression of Ndrg1 also suppressed cell viability, migration, invasion and induced apoptosis of colon cancer cells. Dp44mT attenuated the TGF-ß1-induced EMT in colon cancer cells, and Dp44mT could up-regulate Ndrg1 expression level. Overexpression of Ndrg1 attenuates the TGF-ß1-induced EMT, Dp44mT and Ndrg1 suppressed EMT through activation of Wnt/ß catenin signaling pathway. In conclusion, our data demonstrated that Dp44mT/Ndrg1 have effective anticancer capability in colon cancer cells and that may represent a promising treatment strategy for human colon cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Cycle Proteins/genetics , Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation, Neoplastic , Intracellular Signaling Peptides and Proteins/genetics , Iron Chelating Agents/pharmacology , Thiosemicarbazones/pharmacology , Wnt Signaling Pathway/drug effects , Apoptosis/drug effects , Apoptosis/genetics , Cell Cycle Proteins/agonists , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cyclin D1/antagonists & inhibitors , Cyclin D1/genetics , Cyclin D1/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/genetics , HCT116 Cells , Humans , Intracellular Signaling Peptides and Proteins/agonists , Intracellular Signaling Peptides and Proteins/metabolism , Transforming Growth Factor beta1/antagonists & inhibitors , Transforming Growth Factor beta1/pharmacology , Wnt Signaling Pathway/genetics , Wnt3A Protein/antagonists & inhibitors , Wnt3A Protein/genetics , Wnt3A Protein/metabolism , beta Catenin/antagonists & inhibitors , beta Catenin/genetics , beta Catenin/metabolism
6.
J Diabetes Complications ; 32(9): 813-818, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29996974

ABSTRACT

AIM: The impact of BL11282, an imidazoline receptor (NISCH) agonist, on potentiation of glucose-stimulated insulin secretion (GSIS) from isolated human non-diabetic (ND) and type 2 diabetic (T2D) islets was investigated. METHODS: Analysis of mRNA was performed by RNA-sequencing and qPCR. Insulin and cAMP by RIA and ELISA respectively. RESULTS: RNA-sequencing data revealed that NISCH is highly expressed in fat tissues, islets, liver and muscles, with eight detectable splice variants of transcripts in islets. NISCH had a positive correlation with GLP-1 (GLP1R) and GIP (GIPR) receptor transcripts. The expression of NISCH was confirmed by qPCR in human islets. NISCH and GLP1R were comparably higher expressed in mouse islets compared to human islets. GSIS was dose-dependently potentiated by BL11282 from incubated islets of ND and T2D human islet donors. The insulinotropic action of BL11282 was associated with increased cAMP. While the harmful effect of high glucose on reductive capacity of islet cells was enhanced by glibenclamide during long-term culture, it was counteracted by BL11282 or Bt2-cAMP. BL11282 also increased proliferation of INS-1 cells during long-time culture. CONCLUSION: Our data suggest that BL11282 potentiates GSIS by an action involving cAMP/PKA system and BL11282 could be an attractive insulinotropic and ß-cell protective agent.


Subject(s)
Imidazoles/pharmacology , Imidazoline Receptors/agonists , Insulin-Secreting Cells/drug effects , Islets of Langerhans/drug effects , Animals , Cells, Cultured , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Gene Expression/drug effects , Humans , Imidazoline Receptors/genetics , Imidazoline Receptors/metabolism , Insulin/metabolism , Insulin Secretion/drug effects , Insulin Secretion/genetics , Insulin-Secreting Cells/pathology , Insulin-Secreting Cells/physiology , Intracellular Signaling Peptides and Proteins/agonists , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Mice , Signal Transduction/drug effects , Signal Transduction/genetics
7.
Stem Cell Res Ther ; 9(1): 53, 2018 03 07.
Article in English | MEDLINE | ID: mdl-29514703

ABSTRACT

BACKGROUND: Adipose-derived stem cells (ADSCs) are an attractive cell source for bone tissue engineering and have great potential for bone regeneration and defect repair. The transcriptional coactivator with PDZ-binding motif (TAZ) has been demonstrated to modulate osteogenic and adipogenic differentiation of mesenchymal stem cells. However, its roles during ADSC differentiation and therapeutic potentials for bone regeneration have as yet not been well established. METHODS: TAZ expression was measured during osteogenic differentiation of ADSCs in vitro. Both loss-of-function and gain-of-function approaches by TAZ knockdown or enforced overexpression were utilized to determine its functions during osteogenic differentiation of ADSCs. TM-25659, a chemical activator of TAZ, was used to determine whether pharmacological activation of TAZ in ADSCs enhanced osteogenic differentiation in vitro and bone formation in animal models. The molecular mechanisms underlying TAZ in promoting osteogenesis of ADSCs were also explored. RESULTS: Increased TAZ expression was observed during osteogenic differentiation of human ADSCs. TAZ knockdown resulted in compromised osteogenic differentiation and enhanced adipogenic differentiation of ADSCs. In contrast, enforced TAZ overexpression yielded increased osteogenic differentiation and bone regeneration in vivo, and impaired adipogenic differentiation of ADSCs. Pharmacological activation of TAZ by its chemical activator TM-25659 facilitated osteogenic differentiation of ADSCs. Noticeably, transient treatment of ADSCs with TM-25659 or intraperitoneal injection of TM-25659 significantly enhanced bone regeneration of ADSCs loaded with porous ß-TCP in vivo. Mechanistically, TM-25659 exposure significantly promoted TAZ phosphorylation and nuclear translocation, and potentiated the assembly of the TAZ-Runx2 complex. Subsequently, the TAZ-Runx2 complex was further recruited to the promoter of osteocalcin and in turn enhanced its transcription. CONCLUSIONS: Our findings indicate that TAZ is a key mediator that promotes ADSC commitment to the osteoblast lineage. Pharmacological activation of TAZ in ADSCs might become a feasible and promising approach to enhance bone regeneration and repair.


Subject(s)
Intracellular Signaling Peptides and Proteins/agonists , Mesenchymal Stem Cells/cytology , Osteogenesis , Adipose Tissue/cytology , Adult , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Differentiation , Cells, Cultured , Female , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Mice, Nude , Tetrazoles/pharmacology , Trans-Activators , Transcription Factors , Transcriptional Coactivator with PDZ-Binding Motif Proteins
8.
Oncotarget ; 7(35): 56253-56265, 2016 Aug 30.
Article in English | MEDLINE | ID: mdl-27494845

ABSTRACT

Bcl2 and IAP families are anti-apoptotic proteins deregulated in multiple myeloma (MM) cells. Pharmacological inhibition of each of these families has shown significant activity only in subgroups of MM patients. Here, we have examined a broad-spectrum Bcl2 family inhibitor Obatoclax (OBX) in combination with a Smac mimetic LCL161 in MM cell lines and patient cells. LCL161/OBX combination induced synergistic cytotoxicity and anti-proliferative effects on a broad range of human MM cell lines. The cytotoxicity was mediated through inhibition of the IAPs, activation of caspases and up regulation of the pro-apoptotic proteins Bid, Bim, Puma and Noxa by the drug combination. In addition, we observed that OBX caused ER stress and activated the Unfolded Protein Response (UPR) leading to drug resistance. LCL161, however inhibited spliced Xbp-1, a pro-survival factor. In addition, we observed that OBX increased GRP78 localization to the cell surface, which then induced PI3K dependent Akt activation and resistance to cell death. LCL161 was able to block OBX induced Akt activation contributing to synergistic cell death. Our results support clinical evaluation of this combination strategy in relapsed refractory MM patients.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Endoplasmic Reticulum Stress/drug effects , Multiple Myeloma/drug therapy , Neoplasm Recurrence, Local/drug therapy , Pyrroles/pharmacology , Thiazoles/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Apoptosis Regulatory Proteins , Caspases/metabolism , Cell Line, Tumor , Down-Regulation , Drug Resistance, Neoplasm , Drug Synergism , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/metabolism , Humans , Indoles , Intracellular Signaling Peptides and Proteins/agonists , Mitochondrial Proteins/agonists , Multiple Myeloma/pathology , Neoplasm Recurrence, Local/pathology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/metabolism , Pyrroles/therapeutic use , Thiazoles/therapeutic use , Unfolded Protein Response/drug effects , Up-Regulation , X-Box Binding Protein 1/metabolism
9.
Sci Rep ; 6: 23768, 2016 Mar 31.
Article in English | MEDLINE | ID: mdl-27029555

ABSTRACT

Robust and sensitive detection systems are a crucial asset for risk management of chemicals, which are produced in increasing number and diversity. To establish an in vivo biosensor system with quantitative readout for potential toxicant effects on motor function, we generated a transgenic zebrafish line TgBAC(hspb11:GFP) which expresses a GFP reporter under the control of regulatory elements of the small heat shock protein hspb11. Spatiotemporal hspb11 transgene expression in the musculature and the notochord matched closely that of endogenous hspb11 expression. Exposure to substances that interfere with motor function induced a dose-dependent increase of GFP intensity beginning at sub-micromolar concentrations, while washout of the chemicals reduced the level of hspb11 transgene expression. Simultaneously, these toxicants induced muscle hyperactivity with increased calcium spike height and frequency. The hspb11 transgene up-regulation induced by either chemicals or heat shock was eliminated after co-application of the anaesthetic MS-222. TgBAC(hspb11:GFP) zebrafish embryos provide a quantitative measure of muscle hyperactivity and represent a robust whole organism system for detecting chemicals that affect motor function.


Subject(s)
Biosensing Techniques/methods , Green Fluorescent Proteins/genetics , Intracellular Signaling Peptides and Proteins/genetics , Motor Activity/drug effects , Muscles/drug effects , Mutant Chimeric Proteins/genetics , Animals , Animals, Genetically Modified , Azinphosmethyl/analysis , Azinphosmethyl/toxicity , Dose-Response Relationship, Drug , Founder Effect , Galantamine/analysis , Galantamine/toxicity , Gene Expression Regulation , Green Fluorescent Proteins/agonists , Green Fluorescent Proteins/antagonists & inhibitors , Green Fluorescent Proteins/metabolism , Intracellular Signaling Peptides and Proteins/agonists , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/metabolism , Muscles/metabolism , Mutant Chimeric Proteins/agonists , Mutant Chimeric Proteins/antagonists & inhibitors , Mutant Chimeric Proteins/metabolism , Notochord/drug effects , Notochord/metabolism , Pesticides/analysis , Pesticides/toxicity , Promoter Regions, Genetic , Propoxur/analysis , Propoxur/toxicity , Zebrafish
10.
J Biol Chem ; 291(3): 1029-52, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26534963

ABSTRACT

N-MYC downstream-regulated gene-1 (NDRG1) is a potent growth and metastasis suppressor that acts through its inhibitory effects on a wide variety of cellular signaling pathways, including the TGF-ß pathway, protein kinase B (AKT)/PI3K pathway, RAS, etc. To investigate the hypothesis that its multiple effects could be regulated by a common upstream effector, the role of NDRG1 on the epidermal growth factor receptor (EGFR) and other members of the ErbB family, namely human epidermal growth factor receptor 2 (HER2) and human epidermal growth factor receptor 3 (HER3), was examined. We demonstrate that NDRG1 markedly decreased the expression and activation of EGFR, HER2, and HER3 in response to the epidermal growth factor (EGF) ligand, while also inhibiting formation of the EGFR/HER2 and HER2/HER3 heterodimers. In addition, NDRG1 also decreased activation of the downstream MAPKK in response to EGF. Moreover, novel anti-tumor agents of the di-2-pyridylketone class of thiosemicarbazones, namely di-2-pyridylketone 4,4-dimethyl-3-thiosemicarbazone and di-2-pyridylketone 4-cyclohexyl-4-methyl-3-thiosemicarbazone, which markedly up-regulate NDRG1, were found to inhibit EGFR, HER2, and HER3 expression and phosphorylation in cancer cells. However, the mechanism involved appeared dependent on NDRG1 for di-2-pyridylketone 4,4-dimethyl-3-thiosemicarbazone, but was independent of this metastasis suppressor for di-2-pyridylketone 4-cyclohexyl-4-methyl-3-thiosemicarbazone. This observation demonstrates that small structural changes in thiosemicarbazones result in marked alterations in molecular targeting. Collectively, these results reveal a mechanism for the extensive downstream effects on cellular signaling attributed to NDRG1. Furthermore, this study identifies a novel approach for the treatment of tumors resistant to traditional EGFR inhibitors.


Subject(s)
Cell Cycle Proteins/metabolism , Colonic Neoplasms/metabolism , ErbB Receptors/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/metabolism , MAP Kinase Signaling System , Pancreatic Neoplasms/metabolism , Pyridines/therapeutic use , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-3/antagonists & inhibitors , Thiosemicarbazones/therapeutic use , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Cycle Proteins/agonists , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Epidermal Growth Factor/antagonists & inhibitors , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , ErbB Receptors/agonists , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Intracellular Signaling Peptides and Proteins/agonists , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , MAP Kinase Signaling System/drug effects , Mice, Nude , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Pyridines/pharmacology , RNA Interference , Random Allocation , Receptor, ErbB-2/agonists , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/agonists , Receptor, ErbB-3/genetics , Receptor, ErbB-3/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Thiosemicarbazones/pharmacology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
11.
FEBS J ; 282(24): 4672-8, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26432171

ABSTRACT

p62/SQSTM1 is a stress-inducible cellular protein that is conserved among metazoans but not in plants and fungi. p62/SQSTM1 has multiple domains that mediate its interactions with various binding partners and it serves as a signaling hub for diverse cellular events such as amino acid sensing and the oxidative stress response. In addition, p62/SQSTM1 functions as a selective autophagy receptor for degradation of ubiqutinated substrates. In the present review, we describe the current knowledge about p62 with regard to mammalian target of rapamycin complex 1 activation, the Keap1-Nrf2 pathway and selective autophagy.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Autophagy , Models, Biological , Models, Molecular , Signal Transduction , Adaptor Proteins, Signal Transducing/chemistry , Animals , Enzyme Activation , Humans , Intracellular Signaling Peptides and Proteins/agonists , Intracellular Signaling Peptides and Proteins/metabolism , Kelch-Like ECH-Associated Protein 1 , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes/agonists , Multiprotein Complexes/metabolism , NF-E2-Related Factor 2/agonists , NF-E2-Related Factor 2/metabolism , Protein Interaction Domains and Motifs , Sequestosome-1 Protein , TOR Serine-Threonine Kinases/metabolism
12.
Pharmacol Ther ; 156: 69-74, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26432617

ABSTRACT

This review aims at presenting a new concept pertaining to the development of antioxidants, namely, to evolve from disease-oriented therapy to mechanism-oriented therapy. Using as our illustrative example is DJ-1, a homodimeric protein that is ubiquitously expressed in a variety of mammalian tissues, including the brain, and is found in the matrix and the intermembrane space of the mitochondria. DJ-1 is known to be an endogenous antioxidant against cancer, neurodegeneration and cardiovascular diseases, of which oxidative stress plays a causal role. Interestingly, the mechanistic targets of DJ-1 as an antioxidant, including Daxx, Nrf2, thioredoxin, glutathione, α-synuclein, PTEN/PI3K/Akt, and Pink/Parkin are also associated with those oxidative stress-related diseases. Furthermore, activators of DJ-1 are available in the form of mortalin, phenylbutyrate and NAD(P)H: quinone oxidoreductase 1. It follows that activation of DJ-1 as a common endogenous antioxidant provides a new strategy against cancer, neurodegeneration and cardiovascular diseases. Since clinical trials on exogenous application of the known antioxidants have basically failed, an alternative approach would logically be to activate the endogenous antioxidants that are already present in the appropriate cellular locale where elevated oxidative stress is the culprit for the disease. At the same time, since oxidative stress is a common denominator among cancer, neurodegeneration and cardiovascular diseases, development of antioxidant therapy should target the reduction in reactive oxygen species. Instead of focusing on disease-oriented therapy, pharmaceutical companies should concentrate on developing agents and dosing schemes for effective activation of the endogenous antioxidants that are associated with a multitude of oxidative stress-related diseases (mechanism-oriented therapy).


Subject(s)
Antioxidants/metabolism , Cardiovascular Diseases/metabolism , Intracellular Signaling Peptides and Proteins/agonists , Intracellular Signaling Peptides and Proteins/biosynthesis , Neoplasms/metabolism , Neurodegenerative Diseases/metabolism , Oncogene Proteins/agonists , Oncogene Proteins/biosynthesis , Humans , Parkinson Disease/metabolism , Protein Deglycase DJ-1 , Reactive Oxygen Species/metabolism
13.
J Nutr Biochem ; 26(9): 921-8, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26007286

ABSTRACT

The activation of sterol regulatory element binding proteins (SREBPs) is regulated by insulin-induced genes 1 and 2 (Insig-1 and Insig-2) and SCAP. We previously reported that feeding R-α-lipoic acid (LA) to Zucker diabetic fatty (ZDF) rats improves severe hypertriglyceridemia. In this study, we investigated the role of cyclic AMP-responsive element binding protein H (CREBH) in the lipid-lowering mechanism of LA and its involvement in the SREBP-1c and Insig pathway. Incubation of McA cells with LA (0.2 mM) or glucose (6 mM) stimulated activation of CREBH. LA treatment further induced mRNA expression of Insig-1 and Insig-2a, but not Insig-2b, in glucose-treated cells. In vivo, feeding LA to obesity-induced hyperlipidemic ZDF rats activated hepatic CREBH and stimulated transcription and translation of Insig-1 and Insig-2a. Activation of CREBH and Insigs induced by LA suppressed processing of SREBP-1c precursor into nuclear SREBP-1c, which subsequently inhibited expression of genes involved in fatty acid synthesis, including FASN, ACC and SCD-1, and reduced triglyceride (TG) contents in both glucose-treated cells and ZDF rat livers. Additionally, LA treatment also decreased abundances of very low density lipoprotein (VLDL)-associated apolipoproteins, apoB100 and apoE, in glucose-treated cells and livers of ZDF rats, leading to decreased secretion of VLDL and improvement of hypertriglyceridemia. This study unveils a novel molecular mechanism whereby LA lowers TG via activation of hepatic CREBH and increased expression of Insig-1 and Insig-2a to inhibit de novo lipogenesis and VLDL secretion. These findings provide novel insight into the therapeutic potential of LA as an anti-hypertriglyceridemia dietary molecule.


Subject(s)
Cyclic AMP Response Element-Binding Protein/agonists , Dietary Supplements , Hepatocytes/enzymology , Hypertriglyceridemia/diet therapy , Hypolipidemic Agents/therapeutic use , Intracellular Signaling Peptides and Proteins/agonists , Membrane Proteins/agonists , Thioctic Acid/therapeutic use , Alternative Splicing , Animals , Cell Line, Tumor , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Hepatocytes/metabolism , Hypertriglyceridemia/blood , Hypertriglyceridemia/metabolism , Hypolipidemic Agents/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lipoproteins, VLDL/blood , Lipoproteins, VLDL/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Protein Processing, Post-Translational , Random Allocation , Rats, Inbred BUF , Rats, Zucker , Signal Transduction , Sterol Regulatory Element Binding Protein 1/antagonists & inhibitors , Sterol Regulatory Element Binding Protein 1/metabolism , Thioctic Acid/metabolism , Up-Regulation
14.
J Biochem ; 158(5): 413-23, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25979969

ABSTRACT

Transcriptional co-activator with PSD-95/Dlg-A/ZO-1 (PDZ)-binding motif (TAZ) regulates in cell proliferation and differentiation. In mesenchymal stem cells it promotes osteogenesis and myogenesis, and suppresses adipogenesis. TAZ activators are expected to prevent osteoporosis, obesity and muscle atrophy. TAZ activation induces epithelial-mesenchymal transition, confers stemness to cancer cells and leads to poor clinical prognosis in cancer patients. In this point of view, TAZ inhibitors should contribute to cancer therapy. Thus, TAZ attracts attention as a two-faced drug target. We screened for TAZ modulators by using human lung cancer A549 cells expressing the fluorescent reporter. Through this assay, we obtained TAZ activator candidates. We unexpectedly found that ethacridine, a widely used antiseptic and abortifacient, enhances the interaction of TAZ and protein phosphatases and increases unphosphorylated and nuclear TAZ. Ethacridine inhibits adipogenesis in mesenchymal C3H10T1/2 cells through the activation of TAZ. This finding suggests that ethacridine is a bona fide TAZ activator and supports that our assay is useful to discover TAZ activators.


Subject(s)
Adipogenesis/drug effects , Anti-Obesity Agents/pharmacology , Ethacridine/pharmacology , Intracellular Signaling Peptides and Proteins/agonists , Mesenchymal Stem Cells/drug effects , Protein Phosphatase 1/metabolism , Protein Phosphatase 2/metabolism , Active Transport, Cell Nucleus/drug effects , Adaptor Proteins, Signal Transducing/agonists , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Cell Line, Tumor , Drug Evaluation, Preclinical , Genes, Reporter/drug effects , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Phosphoproteins/agonists , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation/drug effects , Promoter Regions, Genetic/drug effects , Protein Phosphatase 1/chemistry , Protein Phosphatase 1/genetics , Protein Phosphatase 2/chemistry , Protein Phosphatase 2/genetics , Protein Processing, Post-Translational/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Trans-Activators , Transcription Factors , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , YAP-Signaling Proteins
15.
J Immunol ; 195(1): 126-33, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26002975

ABSTRACT

NF-κB plays a central role in pathogenesis of inflammation and cancer. Many phytochemicals, including γ-tocotrienol (γTE), a natural form of vitamin E, have been shown to inhibit NF-κB activation, but the underlying mechanism has not been identified. In this study, we show that γTE inhibited cytokine-triggered activation of NF-κB and its upstream regulator TGF-ß-activated kinase-1 in murine RAW 264.7 macrophages and primary bone marrow-derived macrophages. In these cells, γTE induced upregulation of A20, an inhibitor of NF-κB. Knockout of A20 partially diminished γTE's anti-NF-κB effect, but γTE increased another NF-κB inhibitor, Cezanne, in A20(-/-) cells. In search of the reason for A20 upregulation, we found that γTE treatment increased phosphorylation of translation initiation factor 2, IκBα, and JNK, indicating induction of endoplasmic reticulum stress. Liquid chromatography-tandem mass spectrometry analyses revealed that γTE modulated sphingolipids, including enhancement of intracellular dihydroceramides, sphingoid bases in de novo synthesis of the sphingolipid pathway. Chemical inhibition of de novo sphingolipid synthesis partially reversed γTE's induction of A20 and the anti-NF-κB effect. The importance of dihydroceramide increase is further supported by the observation that C8-dihydroceramide mimicked γTE in upregulating A20, enhancing endoplasmic reticulum stress, and attenuating TNF-triggered NF-κB activation. Our study identifies a novel anti-NF-κB mechanism where A20 is induced by stress-induced adaptive response as a result of modulation of sphingolipids, and it demonstrates an immunomodulatory role of dihydrocermides.


Subject(s)
Adaptation, Physiological , Intracellular Signaling Peptides and Proteins/agonists , NF-kappa B/antagonists & inhibitors , Sphingolipids/immunology , gamma-Tocopherol/pharmacology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Cell Line , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/immunology , Endopeptidases/genetics , Endopeptidases/immunology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/immunology , Gene Expression Regulation , I-kappa B Proteins/genetics , I-kappa B Proteins/immunology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/immunology , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/immunology , Macrophages/cytology , Macrophages/drug effects , Macrophages/immunology , Mice , Mice, Inbred C57BL , NF-KappaB Inhibitor alpha , NF-kappa B/genetics , NF-kappa B/immunology , Primary Cell Culture , Signal Transduction , Sphingolipids/metabolism , Stress, Physiological/drug effects , Stress, Physiological/immunology , Tumor Necrosis Factor alpha-Induced Protein 3
16.
Nature ; 521(7552): 357-61, 2015 May 21.
Article in English | MEDLINE | ID: mdl-25799995

ABSTRACT

B cells are selected for an intermediate level of B-cell antigen receptor (BCR) signalling strength: attenuation below minimum (for example, non-functional BCR) or hyperactivation above maximum (for example, self-reactive BCR) thresholds of signalling strength causes negative selection. In ∼25% of cases, acute lymphoblastic leukaemia (ALL) cells carry the oncogenic BCR-ABL1 tyrosine kinase (Philadelphia chromosome positive), which mimics constitutively active pre-BCR signalling. Current therapeutic approaches are largely focused on the development of more potent tyrosine kinase inhibitors to suppress oncogenic signalling below a minimum threshold for survival. We tested the hypothesis that targeted hyperactivation--above a maximum threshold--will engage a deletional checkpoint for removal of self-reactive B cells and selectively kill ALL cells. Here we find, by testing various components of proximal pre-BCR signalling in mouse BCR-ABL1 cells, that an incremental increase of Syk tyrosine kinase activity was required and sufficient to induce cell death. Hyperactive Syk was functionally equivalent to acute activation of a self-reactive BCR on ALL cells. Despite oncogenic transformation, this basic mechanism of negative selection was still functional in ALL cells. Unlike normal pre-B cells, patient-derived ALL cells express the inhibitory receptors PECAM1, CD300A and LAIR1 at high levels. Genetic studies revealed that Pecam1, Cd300a and Lair1 are critical to calibrate oncogenic signalling strength through recruitment of the inhibitory phosphatases Ptpn6 (ref. 7) and Inpp5d (ref. 8). Using a novel small-molecule inhibitor of INPP5D (also known as SHIP1), we demonstrated that pharmacological hyperactivation of SYK and engagement of negative B-cell selection represents a promising new strategy to overcome drug resistance in human ALL.


Subject(s)
B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Signal Transduction , Amino Acid Motifs/genetics , Animals , Antigens, CD/metabolism , B-Lymphocytes/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Transformation, Neoplastic , Disease Models, Animal , Drug Resistance, Neoplasm/drug effects , Enzyme Activation/drug effects , Female , Fusion Proteins, bcr-abl/genetics , Gene Deletion , Humans , Inositol Polyphosphate 5-Phosphatases , Intracellular Signaling Peptides and Proteins/agonists , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Phosphoric Monoester Hydrolases/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cells, B-Lymphoid/drug effects , Precursor Cells, B-Lymphoid/metabolism , Precursor Cells, B-Lymphoid/pathology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/deficiency , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Protein-Tyrosine Kinases/metabolism , Receptors, Antigen, B-Cell/deficiency , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, B-Cell/metabolism , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Signal Transduction/drug effects , Syk Kinase , Tyrosine/metabolism , Xenograft Model Antitumor Assays
17.
Mol Med Rep ; 12(1): 1145-50, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25776802

ABSTRACT

Previous studies have revealed that high levels of serum homocysteine (Hcy) are closely associated with the development of juvenile and age-related cataracts. An increased concentration of Hcy is likely to induce gene specific demethylation in DNA promoter regions. The aim of the present study was to prevent this demethylation by administering acetyl-l-carnitine (ALCAR) to human lens epithelial cells (HLECs). Different concentrations of Hcy were used to treat HLECs for 3, 6, 12 and 24 h and the findings were used to determine the optimum dose to induce endoplasmic reticulum (ER) stress. Similarly, the concentration of ALCAR was standardized. The production of reactive oxygen species (ROS) and the percentage of cells undergoing cell death were measured. The levels of antioxidants, ER stress-associated proteins, mRNA levels of nuclear factor erythroid-2-related factor 2 (Nrf2), Kelch-like ECH-associated protein 1 (Keap1) and promoter DNA methylation of the Keap1 gene were also assessed. Hcy was observed to induce ER stress, produce ROS and lead to cell death. However, administration of ALCAR prevented these effects to a significant degree. Additionally, western blot analysis revealed that ALCAR increased the levels of antioxidant proteins, including catalase, superoxide dismutase, glutathione peroxidase, Nrf2, Keap1 and glutathione. Similarly, the reverse transcription-quantitative polymerase chain reaction experiments on Nrf2 and Keap1, as well as the bisulfite genomic DNA sequencing analysis revealed a preventive effect of ALCAR against Hcy-induced ER stress. The ER stress-induced activation of the unfolded protein response is responsible for demethylation of Keap1 promoter DNA to activate the expression of the Keap1 protein, which then increases the targeting of Nrf2 for proteosomal degradation. This decrease in Nrf2 activity represses the transcription of numerous antioxidant enzyme genes and alters the redox-balance towards lens oxidation. However, treatment with ALCAR led to significant protection from these effects. The present results suggested that ALCAR either prevents or ameliorates the actions of the antioxidant system in HLECs at the level of the protein and the gene. Further advanced studies are required for the development of ALCAR as an anti-cataract agent.


Subject(s)
Acetylcarnitine/pharmacology , Antioxidants/pharmacology , Homocysteine/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Lens, Crystalline/drug effects , NF-E2-Related Factor 2/genetics , Animals , Catalase/genetics , Catalase/metabolism , Cell Death/drug effects , Cell Line , DNA Methylation/drug effects , Endoplasmic Reticulum Stress/drug effects , Epigenesis, Genetic , Glutathione/metabolism , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Homocysteine/pharmacology , Humans , Intracellular Signaling Peptides and Proteins/agonists , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/metabolism , Kelch-Like ECH-Associated Protein 1 , Lens, Crystalline/cytology , Lens, Crystalline/metabolism , NF-E2-Related Factor 2/agonists , NF-E2-Related Factor 2/antagonists & inhibitors , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Promoter Regions, Genetic/drug effects , Proteasome Endopeptidase Complex/drug effects , Proteasome Endopeptidase Complex/metabolism , Proteolysis/drug effects , Reactive Oxygen Species/agonists , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Signal Transduction , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Transcription, Genetic
18.
Apoptosis ; 20(4): 551-61, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25666707

ABSTRACT

Human (HN) prevents stress-induced apoptosis in many cells/tissues. In this study we showed that HN ameliorated chemotherapy [cyclophosphamide (CP) and Doxorubicin (DOX)]-induced male germ cell apoptosis both ex vivo in seminiferous tubule cultures and in vivo in the testis. HN acts by several putative mechanisms via binding to: an IL-12 like trimeric membrane receptor; BAX; or insulin-like growth factor binding protein-3 (IGFBP-3, a proapoptotic factor). To understand the mechanisms of HN on male germ cell apoptosis, we studied five HN analogues including: HNG (HN-S14G, a potent agonist), HNG-F6A (no binding to IGFBP-3), HN-S7A (no self-dimerization), HN-C8P (no binding to BAX), and HN-L12A (a HN antagonist) on CP-induced male germ cell apoptosis in mice. CP-induced germ cell apoptosis was inhibited by HN, HNG, HNG-F6A, HN-S7A, and HN-C8P (less effective); but not by HN-L12A. HN-L12A, but not HN-S7A or HN-C8P, blocked the protective effect of HN against CP-induced male germ cell apoptosis. HN, HN-S7A, and HN-C8P restored CP-suppressed STAT3 phosphorylation. These results suggest that HN: (1) decreases DOX (ex vivo) and CP (in vivo) induced male germ cell apoptosis; (2) action is mediated by the membrane receptor/STAT3 with minor contribution by BAX-binding pathway; (3) self-dimerization or binding to IGFBP-3 may not be involved in HN's effect in testis. HN is an important molecule in the regulation of germ cell homeostasis after injury and agonistic analogues may be developed for treating male infertility or protection against chemotherapy side effects.


Subject(s)
Antineoplastic Agents/toxicity , Apoptosis/drug effects , Germ Cells/cytology , Intracellular Signaling Peptides and Proteins/metabolism , Animals , Cyclophosphamide/toxicity , Doxorubicin/toxicity , Germ Cells/drug effects , Germ Cells/metabolism , Intracellular Signaling Peptides and Proteins/agonists , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Male , Mice , Testis/cytology , Testis/drug effects , Testis/metabolism
19.
Expert Opin Ther Targets ; 18(12): 1435-47, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25200207

ABSTRACT

INTRODUCTION: Filamin A interacting protein 1-like (FILIP1L) is a novel tumor suppressor-like protein that has its expression downregulated in various cancers through promoter hypermethylation. When overexpressed, FILIP1L inhibits cancer cell invasion and metastasis through the inhibition of canonical WNT signaling. AREAS COVERED: This review gives an overview of the structure and isoforms, gene expression and cellular location of FILIP1L, and how FILIP1L inhibits cancer invasion and metastasis. Furthermore, the review discusses the potential mechanism by which FILIP1L inhibits cancer metastasis through inhibiting canonical WNT signaling and thus blocking downstream ß-catenin transcriptional targets. EXPERT OPINION: By inhibiting ß-catenin, the key transcriptional factor of the canonical WNT signaling pathway, FILIP1L could block various downstream pathways that are regulated by ß-catenin transcriptional targets. FILIP1L could therefore have great potential as a novel cancer therapeutic target. However, in order to fulfill its therapeutic potential, its precise mechanism of action of antimetastatic activity has to be identified. In addition, the physiological role of FILIP1L and its relationship with other isoforms needs to be characterized.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Delivery Systems/methods , Intracellular Signaling Peptides and Proteins/biosynthesis , Neoplasms/drug therapy , Neoplasms/metabolism , Animals , Drug Delivery Systems/trends , Gene Expression Regulation, Neoplastic , Humans , Intracellular Signaling Peptides and Proteins/agonists
20.
Am J Physiol Endocrinol Metab ; 307(9): E773-83, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25159329

ABSTRACT

The steroid receptor coactivator 1 (SRC1) regulates key metabolic pathways, including glucose homeostasis. SRC1(-/-) mice have decreased hepatic expression of gluconeogenic enzymes and a reduction in the rate of endogenous glucose production (EGP). We sought to determine whether decreasing hepatic and adipose SRC1 expression in normal adult rats would alter glucose homeostasis and insulin action. Regular chow-fed and high-fat-fed male Sprage-Dawley rats were treated with an antisense oligonucleotide (ASO) against SRC1 or a control ASO for 4 wk, followed by metabolic assessments. SRC1 ASO did not alter basal EGP or expression of gluconeogenic enzymes. Instead, SRC1 ASO increased insulin-stimulated whole body glucose disposal by ~30%, which was attributable largely to an increase in insulin-stimulated muscle glucose uptake. This was associated with an approximately sevenfold increase in adipose expression of lipocalin-type prostaglandin D2 synthase, a previously reported regulator of insulin sensitivity, and an approximately 70% increase in plasma PGD2 concentration. Muscle insulin signaling, AMPK activation, and tissue perfusion were unchanged. Although GLUT4 content was unchanged, SRC1 ASO increased the cleavage of tether-containing UBX domain for GLUT4, a regulator of GLUT4 translocation. These studies point to a novel role of adipose SRC1 as a regulator of insulin-stimulated muscle glucose uptake.


Subject(s)
Enzyme Inhibitors/therapeutic use , Glucose Intolerance/drug therapy , Insulin Resistance , Muscle, Skeletal/drug effects , Nuclear Receptor Coactivator 1/antagonists & inhibitors , Oligodeoxyribonucleotides, Antisense/therapeutic use , Adipose Tissue/drug effects , Adipose Tissue/enzymology , Adipose Tissue/metabolism , Animals , Biological Transport/drug effects , Diet, High-Fat/adverse effects , Gene Expression Regulation, Enzymologic/drug effects , Glucose Intolerance/etiology , Glucose Intolerance/metabolism , Glucose Transporter Type 4/agonists , Glucose Transporter Type 4/chemistry , Glucose Transporter Type 4/metabolism , Intracellular Signaling Peptides and Proteins/agonists , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Lipocalins/agonists , Lipocalins/genetics , Lipocalins/metabolism , Liver/drug effects , Liver/enzymology , Liver/metabolism , Male , Muscle, Skeletal/metabolism , Nuclear Receptor Coactivator 1/genetics , Nuclear Receptor Coactivator 1/metabolism , Phosphoenolpyruvate Carboxykinase (GTP)/genetics , Phosphoenolpyruvate Carboxykinase (GTP)/metabolism , Prostaglandin D2/blood , Prostaglandin D2/metabolism , Protein Interaction Domains and Motifs , Proteolysis/drug effects , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL