Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 174
Filter
1.
Nucleic Acids Res ; 49(2): e9, 2021 01 25.
Article in English | MEDLINE | ID: mdl-33264395

ABSTRACT

There has been a surge of interest towards targeting protein synthesis to treat diseases and extend lifespan. Despite the progress, few options are available to assess translation in live animals, as their complexity limits the repertoire of experimental tools to monitor and manipulate processes within organs and individual cells. It this study, we developed a labeling-free method for measuring organ- and cell-type-specific translation elongation rates in vivo. It is based on time-resolved delivery of translation initiation and elongation inhibitors in live animals followed by ribosome profiling. It also reports translation initiation sites in an organ-specific manner. Using this method, we found that the elongation rates differ more than 50% among mouse organs and determined them to be 6.8, 5.0 and 4.3 amino acids per second for liver, kidney, and skeletal muscle, respectively. We further found that the elongation rate is reduced by 20% between young adulthood and mid-life. Thus, translation, a major metabolic process in cells, is tightly regulated at the level of elongation of nascent polypeptide chains.


Subject(s)
Aging/metabolism , Kidney/metabolism , Liver/metabolism , Muscle, Skeletal/metabolism , Peptide Chain Elongation, Translational , Aging/genetics , Animals , Cluster Analysis , Cranial Sinuses , Cycloheximide/administration & dosage , Cycloheximide/pharmacology , Drug Administration Schedule , Harringtonines/administration & dosage , Harringtonines/pharmacology , High-Throughput Nucleotide Sequencing , Injections, Intravenous , Kinetics , Longevity , Macrolides/administration & dosage , Macrolides/pharmacology , Male , Mice , Mice, Inbred C57BL , Orbit , Organ Specificity , Peptide Chain Elongation, Translational/drug effects , Peptide Chain Initiation, Translational , Piperidones/administration & dosage , Piperidones/pharmacology , Ribosomes/metabolism , Tail , Transcriptome
2.
Arch Pharm Res ; 43(8): 863-874, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32720163

ABSTRACT

Acute myocardial infarction (AMI) results in irreversible cardiac cell damage or death because of decreased blood flow to the heart. Apoptosis plays an important role in the process of tissue damage after myocardial infarction (MI), which has pathological and therapeutic implications. Ferulic acid (FA) is a phenolic acid endowed with strong antioxidative and cytoprotective activities. The present study aimed to investigate whether FA protects cardiomyocytes from apoptosis by regulating autophagy, which is a cellular self-digestion process, and one of the first lines of defense against oxidative stress. Apoptosis was induced by TNF-α (10 ng/mL) and cycloheximide (CHX, 5 µg/mL) in rat H9c2 cardiomyocytes. FA-inhibited TNF-α/CHX-induced apoptosis was determined by the quantification of TUNEL-positive cells, and the effect was associated with decreased ROS production and inhibited caspase3 activation. FA treatment enhanced autophagy and increased autophagy-associated protein expression, leading to an inhibition of mTOR signaling. When co-treated with 3-methyladenine (3-MA), an autophagy inhibitor, the anti-apoptotic effect of FA was attenuated. In an in vivo mouse MI model, FA treatment decreased the apoptotic cell number, reduced infarct size, and improved cardiac performance, as determined by histological and echocardiographic assessments. Taken collectively, these results suggest that FA could protect cardiomyocytes from apoptosis by enhancing autophagy.


Subject(s)
Apoptosis/drug effects , Coumaric Acids/pharmacology , Myocardial Infarction/drug therapy , Myocytes, Cardiac/drug effects , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Autophagy/drug effects , Cell Line , Cycloheximide/administration & dosage , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/physiopathology , Myocytes, Cardiac/pathology , Oxidative Stress/drug effects , Rats , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/administration & dosage
3.
Drug Dev Ind Pharm ; 46(4): 557-565, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32126844

ABSTRACT

Raloxifene hydrochloride (RH) suffers from low oral bioavailability due to its low water-solubility and first-pass metabolism. Therefore, a novel phospholipid complex of RH (RHPC) and a matrix dispersion based on phospholipid complex (RHPC-MD) were successfully prepared and optimized. Several methods were used to validate the formation of RHPC and RHPC-MD, such as differential scanning calorimetry, X-ray diffraction, scanning electron microscopy, transmission electron microscopy, infrared spectroscopy, particle size, and zeta potential, meanwhile, their octanol-water partition coefficient, solubility, and dissolution in vitro were also evaluated. To investigate the absorption mechanism of RHPC in vivo, the RHPC was administered to the chylomicron flow blockage rat model. Interestingly, as we expected, a significant reduction in RHPC absorption (67%) (**p< .01) in presence of cycloheximide (CXI) inhibitor was observed, thus confirming the RHPC could be absorbed by lymphatic transport in vivo. Pharmacokinetic studies revealed that the relative oral bioavailability of RHPC as well as RHPC-MD was 223% and 329%, respectively, when comparing with the commercial RH tablets. These outcomes suggested that the current study provided an attractive formulation to enhance the oral bioavailability of RH and stimulated to further research the absorption mechanism of RHPC in vivo.


Subject(s)
Bone Density Conservation Agents/administration & dosage , Phospholipids/chemistry , Raloxifene Hydrochloride/administration & dosage , Selective Estrogen Receptor Modulators/administration & dosage , Administration, Oral , Animals , Biological Availability , Bone Density Conservation Agents/chemistry , Bone Density Conservation Agents/pharmacokinetics , Calorimetry, Differential Scanning , Chylomicrons/biosynthesis , Cycloheximide/administration & dosage , Drug Liberation , Female , Humans , Intestinal Absorption/drug effects , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestine, Small/drug effects , Intestine, Small/metabolism , Models, Animal , Osteoporosis, Postmenopausal/drug therapy , Particle Size , Raloxifene Hydrochloride/chemistry , Raloxifene Hydrochloride/pharmacokinetics , Rats , Selective Estrogen Receptor Modulators/chemistry , Selective Estrogen Receptor Modulators/pharmacokinetics , Solubility , Tablets , X-Ray Diffraction
4.
J Biomed Sci ; 26(1): 72, 2019 Oct 09.
Article in English | MEDLINE | ID: mdl-31597574

ABSTRACT

BACKGROUND: Transforming growth factor (TGF) family members play important roles in the regulation of corneal integrity, and the pathogenesis of corneal fibrosis. Currently, there are no effective agents targeting TGF-ß signaling to diminish corneal fibrosis. Glucosamine (GlcN), which is widely used in the treatment of osteoarthritis, abrogates the morphologic effects of TGF-ß2 on retinal pigmented epithelial cells in a mouse disease model. Here, we sought to determine whether GlcN would exert beneficial effects against TGF-ß1-induced corneal fibrosis. METHODS: In human corneal fibroblasts (HCFs) treated with GlcN, the expression of Krüppel-like factor 4 (KLF4) and its downstream signaling effects were determined in the presence and absence of TGF-ß1 using immunoblot analysis. We further explored GlcN inhibition of fibroblast-to-myofibroblast differentiation via KLF4 siRNA. The effect of cycloheximide on KLF4 protein levels with or without GlcN administration was assessed to determine whether GlcN affects the stability of the KLF4 protein. RESULTS: In HCFs, GlcN induced the expression of KLF4, which regulated the maturation and maintenance of the ocular surface. GlcN partially suppressed the TGF-ß1-induced expression of alpha-smooth muscle actin (α-SMA) and reduced the collagen contraction capacity in HCFs, suggesting a decrease in fibroblast-to-myofibroblast differentiation. This effect appeared to be mediated through suppression of Smad2 phosphorylation and ERK-dependent signaling. The levels of KLF4 mRNA were increased by GlcN and decreased by TGF-ß1 and the TGF-ß1-induced α-SMA mRNA expression was upregulated when the KLF4 gene was silenced. GlcN also appeared to stabilize the KLF4 protein, reducing its turnover in corneal fibroblasts. CONCLUSION: These findings shed light on a novel mechanism by which GlcN suppresses TGF-ß1-induced fibroblast-to-myofibroblast differentiation through the upregulation of KLF4 expression. Current strategies for treating corneal fibrosis were not effective. Elevating KLF4 levels through the use of GlcN might provide an effective alternative to alleviate the development and progression of corneal fibrosis.


Subject(s)
Cell Differentiation/drug effects , Corneal Diseases/drug therapy , Fibrosis/drug therapy , Glucosamine/pharmacology , Kruppel-Like Transcription Factors/genetics , Transforming Growth Factor beta1/genetics , Corneal Diseases/etiology , Corneal Diseases/genetics , Cycloheximide/administration & dosage , Fibroblasts/drug effects , Fibrosis/etiology , Fibrosis/genetics , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/metabolism , Myofibroblasts/physiology , Protective Agents/pharmacology , Protein Synthesis Inhibitors/administration & dosage , Signal Transduction/drug effects , Transforming Growth Factor beta1/metabolism
5.
Am J Physiol Regul Integr Comp Physiol ; 317(6): R793-R802, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31596113

ABSTRACT

Compounds described by humans as "bitter" are sensed by a family of type 2 taste receptors (T2Rs). Previous work suggested that diverse bitter stimuli activate distinct receptors, which might allow for perceptually distinct tastes. Alternatively, it has been shown that multiple T2Rs are expressed on the same taste cell, leading to the contrary suggestion that these stimuli produce a unitary perception. Behavioral work done to address this in rodent models is limited to Spector and Kopka (Spector AC, Kopka SL. J Neurosci 22: 1937-1941, 2002), who demonstrated that rats cannot discriminate quinine from denatonium. Supporting this finding, it has been shown that quinine and denatonium activate overlapping T2Rs and neurons in both the mouse and rat nucleus of the solitary tract (NTS). However, cycloheximide and 6-n-propylthiouracil (PROP) do not appear to overlap with quinine in the NTS, suggesting that these stimuli may be discriminable from quinine and the denatonium/quinine comparison is not generalizable. Using the same procedure as Spector and Kopka, we tasked animals with discriminating a range of stimuli (denatonium, cycloheximide, PROP, and sucrose octaacetate) from quinine. We replicated and expanded the findings of Spector and Kopka; rats could not discriminate quinine from denatonium, cycloheximide, or PROP. Rats showed a very weak ability to discriminate between quinine and sucrose octaacetate. All animals succeeded in discriminating quinine from KCl, demonstrating they were capable of the task. These data suggest that rats cannot discriminate this suite of stimuli, although they appear distinct by physiological measures.


Subject(s)
Quinine/pharmacology , Taste , Animals , Cycloheximide/administration & dosage , Cycloheximide/pharmacology , Dose-Response Relationship, Drug , Male , Propylthiouracil/administration & dosage , Propylthiouracil/pharmacology , Quaternary Ammonium Compounds/administration & dosage , Quaternary Ammonium Compounds/pharmacology , Quinine/administration & dosage , Random Allocation , Rats , Rats, Long-Evans , Stimulation, Chemical , Sucrose/administration & dosage , Sucrose/analogs & derivatives , Sucrose/pharmacology
6.
Behav Neurosci ; 132(2): 99-105, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29672107

ABSTRACT

Mixed evidence exists regarding the role of N-methyl-D-aspartate (NMDA) receptors in memory reconsolidation. We provide no evidence that NMDA receptors are involved with memory reconsolidation, but instead demonstrate that prereactivation systemic MK-801 injection, combined with postreactivation intrabasolateral amygdala (BLA) cycloheximide infusion, produces a delayed potentiation of extinction learning. These data suggest that an interaction between NMDA antagonism and protein synthesis inhibition may enhance extinction by exerting effects outside of the intended reconsolidation manipulation window. The present work demonstrates a novel pharmacological enhancement of extinction, and underscores the importance of employing proper control procedures in reconsolidation research. (PsycINFO Database Record


Subject(s)
Cycloheximide/administration & dosage , Discrimination, Psychological/drug effects , Dizocilpine Maleate/administration & dosage , Extinction, Psychological/drug effects , Memory/drug effects , Psychotropic Drugs/administration & dosage , Amygdala/drug effects , Amygdala/metabolism , Animals , Discrimination, Psychological/physiology , Excitatory Amino Acid Antagonists/administration & dosage , Extinction, Psychological/physiology , Fear/drug effects , Fear/physiology , Freezing Reaction, Cataleptic/drug effects , Freezing Reaction, Cataleptic/physiology , Male , Memory/physiology , Protein Synthesis Inhibitors/administration & dosage , Rats, Long-Evans , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Time Factors
7.
Cell Reprogram ; 20(3): 187-195, 2018 06.
Article in English | MEDLINE | ID: mdl-29688743

ABSTRACT

Unlike oocytes of many other mammalian species, parthenogenetically activated hamster oocytes have not been reported to develop beyond the two-cell stage. This study investigated the in vitro development into blastocysts of parthenogenetic embryos of Golden Syrian hamsters. We observed that hamster oocytes could easily be artificially activated (AA) by treatment with ionomycin plus 6-dimethylaminopurine + cycloheximide + cytochalasin B as assessed by embryo cleavage in HECM-9 (63.15%) or HECM-10 (63.82%). None of the cleaved embryos developed beyond the two-cell stage when cultured in either of the two media. However, some of the embryos overcame the two-cell block and developed to the blastocyst stage (26.45%) when they were first cultured in HECM-10 for 24 hours and then in HECM-9 (serial culture media HECM-10-9) for 72 hours. Blastocyst development was further significantly (66.2%) improved when embryos were cultured in HECM-10 supplemented with ethylenediaminetetraacetic acid for 24 hours, then in HECM-9 supplemented with glucose for 72 hours (serial culture media HECM-11a-b). Hamster oocytes activated with ionomycin, ethanol, or a combination of the two treatments would develop to the blastocyst stage in serial culture media HECM-11a-b, whereas none of the spontaneously activated oocytes cleaved (0% vs. 86.93%, p < 0.05). DNA and microtubule configurations of spontaneously activated and AA oocytes were assessed by immunocytochemical staining and fluorescence microscopy. The results indicate that serial culture and the method of activation are critical for overcoming the in vitro developmental block of hamster parthenogenetic embryos. This study is the first to report blastocyst development from parthenogenetically activated hamster oocytes.


Subject(s)
Blastocyst/physiology , Embryo Culture Techniques/veterinary , Oocytes/physiology , Parthenogenesis , Adenine/administration & dosage , Adenine/analogs & derivatives , Animals , Blastocyst/drug effects , Culture Media , Cycloheximide/administration & dosage , Cytochalasin B/administration & dosage , Embryo Culture Techniques/methods , Embryonic Development/drug effects , Female , Ionomycin/administration & dosage , Mesocricetus , Oocytes/drug effects
8.
Mol Cancer Res ; 15(7): 905-914, 2017 07.
Article in English | MEDLINE | ID: mdl-28242811

ABSTRACT

KRAS is frequently mutated in lung cancers and is associated with aggressive biology and chemotherapy resistance. Therefore, innovative approaches are needed to treat these lung cancers. Prior work implicated the IFN-stimulated gene 15 (ISG15) deubiquitinase (DUB) USP18 as having antineoplastic activity by regulating lung cancer growth and oncoprotein stability. This study demonstrates that USP18 affects the stability of the KRAS oncoprotein. Interestingly, loss of USP18 reduced KRAS expression, and engineered gain of USP18 expression increased KRAS protein levels in lung cancer cells. Using the protein synthesis inhibitor cycloheximide, USP18 knockdown significantly reduced the half-life of KRAS, but gain of USP18 expression significantly increased its stability. Intriguingly, loss of USP18 altered KRAS subcellular localization by mislocalizing KRAS from the plasma membrane. To explore the biologic consequences, immunohistochemical (IHC) expression profiles of USP18 were compared in lung cancers of KrasLA2/+ versus cyclin E engineered mouse models. USP18 expression was higher in Kras-driven murine lung cancers, indicating a link between KRAS and USP18 expression in vivo To solidify this association, loss of Usp18 in KrasLA2/+ /Usp18-/- mice was found to significantly reduce lung cancers as compared with parental KrasLA2/+ mice. Finally, translational relevance was confirmed in a human lung cancer panel by showing that USP18 IHC expression was significantly higher in KRAS-mutant versus wild-type lung adenocarcinomas.Implications: Taken together, this study highlights a new way to combat the oncogenic consequences of activated KRAS in lung cancer by inhibiting the DUB USP18. Mol Cancer Res; 15(7); 905-14. ©2017 AACR.


Subject(s)
Adenocarcinoma/genetics , Endopeptidases/genetics , Lung Neoplasms/genetics , Protein Biosynthesis/drug effects , Proto-Oncogene Proteins p21(ras)/genetics , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Animals , Carcinogenesis/genetics , Cell Line, Tumor , Cell Membrane/genetics , Cyclin E/genetics , Cycloheximide/administration & dosage , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Male , Mice, Knockout , Mutation , Ubiquitin Thiolesterase
9.
Int J Oncol ; 50(5): 1857-1867, 2017 May.
Article in English | MEDLINE | ID: mdl-28358429

ABSTRACT

Previous studies described that the expression of aquaporin 5 (AQP5) was altered in tumors of various organs. AQP5 is attracting attention as a new cancer therapeutic target. In the present study, heat shock-induced changes in AQP5 expression were evaluated by immunofluorescent staining (IF) and western blotting (WB) of liver cancer cells. AQP5 knockdown experiments or a heat shock treatment were conducted, and their effects on cell volume, proliferation, cell cycle, the activity of apoptosis and migration/invasion were compared. Cycloheximide (CHX) chase experiments and double IF of AQP5 and light chain 3B (LC3B) were performed to investigate the mechanisms underlying changes in AQP5 expression. The results showed that IF and WB revealed decrease in AQP5 expression on cellular membranes and in the cytoplasm of heated cells. AQP5 knockdown and heat shock similarly decreased cell volume, suppressed migration/invasion and proliferation, and induced early apoptosis and partial G0/G1 arrest. CHX chase experiments revealed that heat shock accelerated the degradation of AQP5, which was rescued under CHX and the autophagy inhibitor, bafilomycin A1 (BafA1). Double IF showed the co-localization of AQP5 and LC3B on BafA1-treated heated cells. In conclusion, we demonstrated that heat shock decreased AQP5 on cellular membranes and in the cytoplasm by activating autophagic degradation, and heat shock and AQP5 knockdown exerted similar anticancer effects, suggesting that heat shock exerts anticancer effects via the autophagic degradation of AQP5.


Subject(s)
Aquaporin 5/genetics , Autophagy/drug effects , Heat-Shock Response/genetics , Liver Neoplasms/drug therapy , Microtubule-Associated Proteins/genetics , Apoptosis/genetics , Aquaporin 5/biosynthesis , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cycloheximide/administration & dosage , Cytoplasm/genetics , Gene Expression Regulation, Neoplastic/genetics , Gene Knockdown Techniques , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Macrolides/administration & dosage , Microtubule-Associated Proteins/biosynthesis , Proteolysis
10.
J Zhejiang Univ Sci B ; 17(9): 672-82, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27604859

ABSTRACT

OBJECTIVE: To explore the efficacy of ursolic acid in sensitizing colon cancer cells to chemotherapy under hypoxia and its underlying mechanisms. METHODS: Three colon cancer cell lines (RKO, LoVo, and SW480) were used as in vitro models. 5-Fluorouracil (5-FU) and oxaliplatin were used as chemotherapeutic drugs. Cell viability and apoptosis were tested to evaluate the sensitivity of colon cancer cells to chemotherapy. The transcription and expression levels of hypoxia-inducible factor-1α (HIF-1α), multidrug resistance gene 1 (MDR1), and vascular endothelial growth factors (VEGF) were assessed by quantitative real-time polymerase chain reaction (qRT-PCR) and immunoblotting. Cycloheximide and MG132 were used to inhibit protein synthesis and degradation, respectively. In vitro tube formation assay was used to evaluate angiogenesis. RESULTS: We demonstrated the chemosensitizing effects of ursolic acid with 5-FU and oxaliplatin in three colon cancer cell lines under hypoxia. This effect was correlated to its inhibition of MDR1 through HIF-1α. Moreover, ursolic acid was capable of inhibiting HIF-1α accumulation with little effects on its constitutional expression in normoxia. In addition, ursolic acid also down-regulated VEGF and inhibited tumor angiogenesis. CONCLUSIONS: Ursolic acid exerted chemosensitizing effects in colon cancer cells under hypoxia by inhibiting HIF-1α accumulation and the subsequent expression of the MDR1 and VEGF.


Subject(s)
Antineoplastic Agents/chemistry , Colonic Neoplasms/drug therapy , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Triterpenes/chemistry , ATP Binding Cassette Transporter, Subfamily B/metabolism , Apoptosis , Cell Hypoxia , Cell Line, Tumor , Cell Proliferation , Cell Survival , Cycloheximide/administration & dosage , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Fluorouracil/administration & dosage , Human Umbilical Vein Endothelial Cells , Humans , Neovascularization, Pathologic , Organoplatinum Compounds/administration & dosage , Oxaliplatin , RNA, Small Interfering/metabolism , Real-Time Polymerase Chain Reaction , Vascular Endothelial Growth Factor A/metabolism , Ursolic Acid
11.
Learn Mem ; 23(8): 391-8, 2016 08.
Article in English | MEDLINE | ID: mdl-27421890

ABSTRACT

Previously consolidated memories have the potential to enter a state of lability upon memory recall, during which time the memory can be altered before undergoing an additional consolidation-like process and being stored again as a long-term memory. Blocking reconsolidation of aberrant memories has been proposed as a potential treatment for psychiatric disorders including addiction. Here we investigated of the effect of systemically administering the protein synthesis inhibitor cycloheximide or the ß-adrenergic antagonist propranolol on reconsolidation. Rats were trained to self-administer cocaine, during which each lever press resulted in the presentation of a cue paired with an intravenous infusion of cocaine. After undergoing lever press extinction to reduce operant responding, the cue memory was reactivated and rats were administered systemic injections of propranolol, cycloheximide, or vehicle. Post-reactivation cycloheximide, but not propranolol, resulted in a reactivation-dependent decrease in cue-induced reinstatement, indicative of reconsolidation blockade by protein synthesis inhibition. The present data indicate that systemically targeting protein synthesis as opposed to the ß-adrenergic system may more effectively attenuate the reconsolidation of a drug-related memory and decrease drug-seeking behavior.


Subject(s)
Cocaine/administration & dosage , Memory Consolidation/physiology , Mental Recall/physiology , Protein Synthesis Inhibitors/administration & dosage , Receptors, Adrenergic, beta/physiology , Adrenergic beta-Antagonists/administration & dosage , Animals , Conditioning, Operant/drug effects , Cues , Cycloheximide/administration & dosage , Male , Memory Consolidation/drug effects , Mental Recall/drug effects , Propranolol/administration & dosage , Rats, Sprague-Dawley
12.
BMC Cancer ; 16: 239, 2016 Mar 19.
Article in English | MEDLINE | ID: mdl-26993100

ABSTRACT

BACKGROUND: Carbonic anhydrase IX (CA IX) is a tumor-associated, highly active, transmembrane carbonic anhydrase isoform regulated by hypoxia and implicated in pH control and adhesion-migration-invasion. CA IX ectodomain (ECD) is shed from the tumor cell surface to serum/plasma of patients, where it can signify cancer prognosis. We previously showed that the CA IX ECD release is mediated by disintegrin and metalloproteinase ADAM17. Here we investigated the CA IX ECD shedding in tumor cells undergoing apoptosis in response to cytotoxic drugs, including cycloheximide and doxorubicin. METHODS: Presence of cell surface CA IX was correlated to the extent of apoptosis by flow cytometry in cell lines with natural or ectopic CA IX expression. CA IX ECD level was assessed by ELISA using CA IX-specific monoclonal antibodies. Effect of recombinant CA IX ECD on the activation of molecular pathways was evaluated using the cell-based dual-luciferase reporter assay. RESULTS: We found a significantly lower occurrence of apoptosis in the CA IX-positive cell subpopulation than in the CA IX-negative one. We also demonstrated that the cell-surface CA IX level dropped during the death progress due to an increased ECD shedding, which required a functional ADAM17. Inhibitors of metalloproteinases reduced CA IX ECD shedding, but not apoptosis. The CA IX ECD release induced by cytotoxic drugs was connected to elevated expression of CA IX in the surviving fraction of cells. Moreover, an externally added recombinant CA IX ECD activated a pathway driven by the Nanog transcription factor implicated in epithelial-mesenchymal transition and stemness. CONCLUSIONS: These findings imply that the increased level of the circulating CA IX ECD might be useful as an indicator of an effective antitumor chemotherapy. Conversely, elevated CA IX ECD might generate unwanted effects through autocrine/paracrine signaling potentially contributing to resistance and tumor progression.


Subject(s)
ADAM17 Protein/genetics , Carbonic Anhydrase IX/genetics , Epithelial-Mesenchymal Transition/genetics , Neoplasms/genetics , ADAM17 Protein/metabolism , Antibodies, Monoclonal/administration & dosage , Apoptosis/drug effects , Apoptosis/genetics , Carbonic Anhydrase IX/administration & dosage , Carbonic Anhydrase IX/metabolism , Cell Hypoxia/genetics , Cycloheximide/administration & dosage , Female , HeLa Cells , Humans , Male , Neoplasms/pathology
13.
Brain Res ; 1642: 104-113, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27017957

ABSTRACT

Memory reconsolidation processes and protein kinase Mzeta (PKMzeta) activity in memory maintenance and reorganization are poorly understood. Therefore, we examined memory reconsolidation and PKMzeta activity during the maintenance and reorganization of a conditioned food aversion memory among snails. These processes were specifically evaluated after administration of a serotonin receptor antagonist (methiothepin), NMDA glutamate receptor antagonist (MK-801), protein synthesis inhibitor (cycloheximide; CYH), or PKMzeta inhibitor (zeta inhibitory peptide; ZIP) either 2 or 10 days after aversion training. Two days post-training, injections of MK-801 or CYH, combined with a conditioned stimulus reminder, caused amnesia development, and a second training 11 days after this induction did not lead to long-term memory formation. Interestingly, MK-801 or CYH injections and the reminder 10 days after training did not affect memory retrieval. Methiothepin and the reminder, or ZIP without the reminder, at 2 and 10 days after training led to memory impairment, while a second training 11 days after amnesia induction resulted in memory formation. These results suggest that the maintenance of a conditioned food aversion involves two different components with variable dynamics. One component could be characterized by memory strengthening over time and involve N-methyl-D-aspartate receptors and protein synthesis reconsolidation at early, but not late, training stages. The other memory component could involve serotonin-dependent reconsolidation and Mzeta-like kinase activity at both early and late stages after learning. Deficiencies within these two components led to various forms of memory impairment, which differed in terms of the formation of a conditioned food aversion during the second training.


Subject(s)
Conditioning, Classical/physiology , Food Preferences/physiology , Memory Consolidation/physiology , Protein Biosynthesis , Protein Kinase C/physiology , Receptors, Serotonin/physiology , Animals , Conditioning, Classical/drug effects , Cycloheximide/administration & dosage , Dizocilpine Maleate/administration & dosage , Excitatory Amino Acid Antagonists/administration & dosage , Food Preferences/drug effects , Helix, Snails , Memory Consolidation/drug effects , Methiothepin/administration & dosage , Protein Biosynthesis/drug effects , Protein Kinase C/antagonists & inhibitors , Protein Synthesis Inhibitors , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/physiology , Serotonin Antagonists/administration & dosage
14.
PLoS One ; 9(4): e94395, 2014.
Article in English | MEDLINE | ID: mdl-24722177

ABSTRACT

Multiple myeloma (MM) is characterized by advanced osteolytic lesions resulting from the activation of osteoclasts (OCs) and inhibition of osteoblasts (OBs). OBs are derived from mesenchymal stem cells (MSCs) from the bone marrow (BM), however the pool and function of BMMSCs in MM patients (MM-BMMSCs) are reduced by myeloma cells (MCs) and cytokines secreted from MCs and related anti-MM treatment. Such reduction in MM-BMMSCs currently cannot be restored by any means. Recently, genetic aberrations of MM-BMMSCs have been noted, which further impaired their differentiation toward OBs. We hypothesize that the MSCs derived from adipose tissue (ADMSCs) can be used as alternative MSC sources to enhance the pool and function of OBs. Therefore, the purpose of this study was to compare the osteogenesis ability of paired ADMSCs and BMMSCs in MM patients who had completed intensive therapy. Fifteen MM patients who had received bortezomib-based induction and autologous transplantation were enrolled. At the third month after the transplant, the paired ADMSCs and BMMSCs were obtained and cultured. Compared with the BMMSCs, the ADMSCs exhibited a significantly higher expansion capacity (100% vs 13%, respectively; P = .001) and shorter doubling time (28 hours vs 115 hours, respectively; P = .019). After inducing osteogenic differentiation, although the ALP activity did not differ between the ADMSCs and BMMSCs (0.78 U/µg vs 0.74±0.14 U/µg, respectively; P = .834), the ADMSCs still exhibited higher calcium mineralization, which was determined using Alizarin red S (1029 nmole vs 341 nmole, respectively; P = .001) and von Kossa staining (2.6 E+05 µm2 vs 5 E+04 µm2, respectively; P = .042), than the BMMSCs did. Our results suggested that ADMSCs are a feasible MSC source for enhancing the pool and function of OBs in MM patients who have received intensive therapy.


Subject(s)
Adipose Tissue/cytology , Antineoplastic Combined Chemotherapy Protocols , Hematopoietic Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Multiple Myeloma/therapy , Osteoblasts/cytology , Adult , Aged , Antineoplastic Agents/therapeutic use , Bone Marrow Cells/pathology , Boronic Acids/therapeutic use , Bortezomib , Cell Differentiation , Cell Proliferation , Cells, Cultured , Cycloheximide/administration & dosage , Dexamethasone/administration & dosage , Female , Humans , Male , Middle Aged , Multiple Myeloma/immunology , Multiple Myeloma/pathology , Osteoblasts/transplantation , Osteogenesis , Pyrazines/therapeutic use , Remission Induction , Thalidomide/administration & dosage , Transplantation, Autologous
15.
Biotechniques ; 55(4): 198-203, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24107251

ABSTRACT

Clinical approaches for tumor treatment often rely on combination therapy where a DNA damaging agent is used in combination with a DNA repair protein inhibitor. For this reason, great efforts have been made during the last decade to identify inhibitors of DNA repair proteins or, alternatively, small molecules that specifically alter protein stability or trafficking. Unfortunately, when studying these drug candidates, classical biochemical approaches are prone to artifacts. The apurinic/apyrimidinic endonuclease (APE1) protein is an essential component of the base excision repair (BER) pathway that is responsible for repairing DNA damage caused by oxidative and alkylating agents. In this work, we combined conditional gene expression knockdown of APE1 protein by RNA interference (RNAi) technology with re-expression of an ectopic recombinant form of APE1 fused with the photoconvertible fluorescent protein (PCFP) Dendra2. Dendra2 did not alter the subcellular localization or endonuclease activity of APE1. We calculated APE1 half-life and compared these results with the classical biochemical approach, which is based on cycloheximide (CHX) treatment. In conclusion, we combined RNAi and in vivo confocal microscopy to study a DNA repair protein demonstrating the feasibility and the advantage of this approach for the study of the cellular dynamic of a DNA repair protein.


Subject(s)
DNA Repair/drug effects , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Luminescent Proteins/genetics , Microscopy, Confocal , Neoplasms/therapy , Cycloheximide/administration & dosage , DNA Damage/drug effects , DNA Repair/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Neoplasms/genetics , Neoplasms/pathology , Oxidation-Reduction/drug effects , RNA Interference
16.
PLoS One ; 8(9): e74533, 2013.
Article in English | MEDLINE | ID: mdl-24040273

ABSTRACT

Despite the subjective nature of pain experience with cognitive and affective dimensions, preclinical pain research has largely focused on its sensory dimension. Here, we examined the relationship between learning/memory and nociceptive behavior in rats with combined learning impairment and persistent nociception. Learning impairment was induced by bilateral hippocampal injection of a mixed Aß solution, whereas persistent nociception produced in these rats by complete Freund's adjuvant-induced ankle inflammation. Those rats with learning impairment showed a diminished development of thermal hyperalgesia and mechanical allodynia and a shorter time course of nociceptive behavior without alteration of their baseline nociceptive threshold. In rats with pre-established hyperalgesia and allodynia due to ankle inflammation, bilateral intra-hippocampal injection of cycloheximide (a protein synthesis inhibitor) promoted the earlier recovery of nociceptive behavior. Moreover, expression of Aß, NR1 subunit of the N-methyl-D-aspartate receptor, and protein kinase Cγ was upregulated, whereas the choline acetyl transferase expression was downregulated, in the hippocampus, thalamus, amygdala, and/or spinal cord of rats with combined learning impairment and persistent nociception. The data indicate that learning impairment could disrupt the response to a state of persistent nociception, suggesting an important role for cognitive maladaptation in the mechanisms of chronic pain. These results also suggest that a preclinical model of combined learning impairment and persistent nociception may be useful to explore the brain mechanisms underlying the transition from acute to chronic pain.


Subject(s)
Cognition Disorders/physiopathology , Hyperalgesia/physiopathology , Learning , Nociception , Pain Threshold/physiology , Pain/physiopathology , Amyloid beta-Peptides/administration & dosage , Animals , Ankle/physiopathology , Behavior, Animal , Cognition Disorders/chemically induced , Cognition Disorders/metabolism , Cycloheximide/administration & dosage , Freund's Adjuvant/administration & dosage , Gene Expression , Hot Temperature , Hyperalgesia/metabolism , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/physiopathology , Male , Pain/metabolism , Pain Measurement , Protein Kinase C/genetics , Protein Kinase C/metabolism , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism
17.
Neuropsychopharmacology ; 38(7): 1308-21, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23348064

ABSTRACT

Antagonism of group I metabotropic glutamate receptors (mGluR1 and mGluR5) reduces behavioral effects of drugs of abuse, including cocaine. However, the underlying mechanisms remain poorly understood. Activation of mGluR5 increases protein synthesis at synapses. Although mGluR5-induced excessive protein synthesis has been implicated in the pathology of fragile X syndrome, it remains unknown whether group I mGluR-mediated protein synthesis is involved in any behavioral effects of drugs of abuse. We report that group I mGluR agonist DHPG induced more pronounced initial depression of inhibitory postsynaptic currents (IPSCs) followed by modest long-term depression (I-LTD) in dopamine neurons of rat ventral tegmental area (VTA) through the activation of mGluR1. The early component of DHPG-induced depression of IPSCs was mediated by the cannabinoid CB1 receptors, while DHPG-induced I-LTD was dependent on protein synthesis. Western blotting analysis indicates that mGluR1 was coupled to extracellular signal-regulated kinase (ERK) and mammalian target of rapamycin (mTOR) signaling pathways to increase translation. We also show that cocaine conditioning activated translation machinery in the VTA via an mGluR1-dependent mechanism. Furthermore, intra-VTA microinjections of mGluR1 antagonist JNJ16259685 and protein synthesis inhibitor cycloheximide significantly attenuated or blocked the acquisition of cocaine-induced conditioned place preference (CPP) and activation of translation elongation factors. Taken together, these results suggest that mGluR1 antagonism inhibits de novo protein synthesis; this effect may block the formation of cocaine-cue associations and thus provide a mechanism for the reduction in CPP to cocaine.


Subject(s)
Cocaine/pharmacology , Conditioning, Psychological/physiology , Nerve Tissue Proteins/physiology , Receptors, Metabotropic Glutamate/physiology , Animals , Cocaine/antagonists & inhibitors , Conditioning, Psychological/drug effects , Cycloheximide/administration & dosage , Cycloheximide/pharmacology , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/physiology , Drug Interactions , Extracellular Signal-Regulated MAP Kinases/metabolism , Glycine/analogs & derivatives , Glycine/pharmacology , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/physiology , Long-Term Synaptic Depression/drug effects , Long-Term Synaptic Depression/physiology , Male , Microinjections , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/drug effects , Peptide Chain Elongation, Translational/drug effects , Protein Synthesis Inhibitors/administration & dosage , Protein Synthesis Inhibitors/pharmacology , Quinolines/administration & dosage , Quinolines/pharmacology , Rats , Receptor, Cannabinoid, CB1/physiology , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Receptors, Metabotropic Glutamate/metabolism , Resorcinols/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Synapses/drug effects , Synapses/metabolism , TOR Serine-Threonine Kinases/metabolism , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/physiology
18.
Bull Exp Biol Med ; 153(5): 738-41, 2012 Sep.
Article in English | MEDLINE | ID: mdl-23113273

ABSTRACT

Possible mechanisms of recovery of the memory impaired during consolidation process were investigated. In mice, amnesia was induced by intraperitoneal cycloheximide (100 mg/kg) administration 20 min before exposure to tone signal combined with footshock (2 sec, 0.5 mA). Reminder by the footshock (2 sec, 0.5 mA) 24 h after the learning procedure resulted in recovery of impaired memory in amnestic animals up to the level of control animals. Analysis of c-Fos expression in response to reminder indicated increased number of c-Fos-positive cells in prelimbic cortex in the animals with unaffected memory in comparison with corresponding parameter in amnestic animals. These findings are indicative of impairment in prelimbic cortex activity in experimental amnesia as well as for reminder ability to recover the memory impaired in that way.


Subject(s)
Amnesia/physiopathology , Prefrontal Cortex/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Recovery of Function/physiology , Acoustic Stimulation , Animals , Cycloheximide/administration & dosage , Cycloheximide/toxicity , Electric Stimulation , Immunohistochemistry , Injections, Intraperitoneal , Male , Mice , Mice, Inbred C57BL
19.
Curr Pharm Des ; 18(28): 4414-25, 2012.
Article in English | MEDLINE | ID: mdl-22721505

ABSTRACT

In the mouse postoperative ileus model, we have shown an increase in oxidative stress after intestinal manipulation occurring earlier in the mucosa than in the muscular layer, which might contribute to epithelial barrier dysfunction. To address these findings in vitro, we assessed TNF-α/cycloheximide (CHX)-induced oxidative stress and apoptosis in a mouse intestinal epithelial cell line, MODE-K. The influence of heme oxygenase (HO)-1-related products and agents known to reduce reactive oxygen species (ROS) production on TNF-α/CHX-induced oxidative stress and apoptosis were investigated. MODE-K cells were exposed to different concentrations of TNF- α/CHX in the absence/presence of the test agents. Cell viability, caspase-3/7 activity, apoptosis, reduced glutathione level (GSH) and intracellular ROS production were measured. TNF-α/CHX decreased cell viability, increased caspase-3/7 activity, induced apoptosis, reduced the GSH level and increased ROS production in a concentration-dependent manner in MODE-K cells. All these effects of TNF- α/CHX were partially prevented by pretreatment with a carbon monoxide-releasing agent (CORM-A1) and nitrite. The antioxidant resveratrol abolished TNF-α/CHX-induced increase in ROS production and caspase-3/7 activity, but apoptosis was only partially prevented. MODE-K cells are sensitive to TNF-α-induced apoptosis in the presence of CHX, which is associated with increased intracellular ROS production and caspase-3/7 activation. The effects were partially mitigated by CORM-A1, nitrite and resveratrol. Thus, these agents could be of potential use in protecting the epithelial barrier against oxidative stress during intestinal ischemia/reperfusion injury.


Subject(s)
Apoptosis/drug effects , Cycloheximide/pharmacology , Oxidative Stress/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Animals , Antioxidants/pharmacology , Boranes/pharmacology , Carbonates/pharmacology , Caspase 3/metabolism , Caspase 7/metabolism , Cell Line , Cell Survival/drug effects , Cycloheximide/administration & dosage , Dose-Response Relationship, Drug , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Heme Oxygenase-1/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Mice , Reactive Oxygen Species/metabolism , Resveratrol , Sodium Nitrite/pharmacology , Stilbenes/pharmacology , Tumor Necrosis Factor-alpha/administration & dosage , Tumor Necrosis Factor-alpha/metabolism
20.
Epilepsia ; 53(7): 1225-32, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22642664

ABSTRACT

PURPOSE: Mossy fiber sprouting (MFS) is a frequent finding following status epilepticus (SE). The present study aimed to test the feasibility of using manganese-enhanced magnetic resonance imaging (MEMRI) to detect MFS in the chronic phase of the well-established pilocarpine (Pilo) rat model of temporal lobe epilepsy (TLE). METHODS: To modulate MFS, cycloheximide (CHX), a protein synthesis inhibitor, was coadministered with Pilo in a subgroup of animals. In vivo MEMRI was performed 3 months after induction of SE and compared to the neo-Timm histologic labeling of zinc mossy fiber terminals in the dentate gyrus (DG). KEY FINDINGS: Chronically epileptic rats displaying MFS as detected by neo-Timm histology had a hyperintense MEMRI signal in the DG, whereas chronically epileptic animals that did not display MFS had minimal MEMRI signal enhancement compared to nonepileptic control animals. A strong correlation (r = 0.81, p < 0.001) was found between MEMRI signal enhancement and MFS. SIGNIFICANCE: This study shows that MEMRI is an attractive noninvasive method for detection of mossy fiber sprouting in vivo and can be used as an evaluation tool in testing therapeutic approaches to manage chronic epilepsy.


Subject(s)
Epilepsy/pathology , Hippocampus/pathology , Magnetic Resonance Imaging , Manganese , Mossy Fibers, Hippocampal/drug effects , Animals , Anticonvulsants/pharmacology , Anticonvulsants/therapeutic use , Cycloheximide/administration & dosage , Disease Models, Animal , Drug Interactions , Epilepsy/chemically induced , Epilepsy/drug therapy , Hippocampus/drug effects , Image Processing, Computer-Assisted , Male , Mossy Fibers, Hippocampal/pathology , Muscarinic Agonists/toxicity , Pilocarpine/toxicity , Protein Synthesis Inhibitors/administration & dosage , Rats , Rats, Wistar , Statistics, Nonparametric , Thiopental/pharmacology , Thiopental/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...