Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Curr Alzheimer Res ; 17(5): 446-459, 2020.
Article in English | MEDLINE | ID: mdl-32579500

ABSTRACT

BACKGROUND: Amyloid Precursor Protein (APP)-Binding Protein 1 (APP-BP1) is a crucial regulator of many key signaling pathways and functions mainly as a scaffold protein to enhance molecular interactions and facilitate catalytic reactions. The interaction of APP-BP1 with Amyloid Precursor Protein (APP) plays a role in cell cycle transit control, which determines the mechanism behind the loss of cell cycle regulation in Alzheimer's Disease (AD). In contrast, neddylation, a posttranslational modification mediated by conjugation of ubiquitin-like protein neural precursor cell expressed developmentally downregulated protein 8 (NEDD8), is activated by a heterodimer composed of APP-BP1 and NEDD8-activating enzyme E1 catalytic subunit (Uba3). NEDD8 controls vital biological events, and along with APP-BP1, its levels are deregulated in AD. OBJECTIVE: The present study investigated the role of melatonin in regulating the APP-BP1 pathway under both physiological and pathological conditions to develop an understanding of the underlying mechanisms. METHODS: Therefore, human SH-SY5Y neuroblastoma cells were treated with various concentrations of Aß42 to induce neurotoxic conditions comparable to AD. RESULTS: The results are the first to demonstrate that melatonin prevents Aß42-induced enhancement of APP-BP1 protein expression and alteration in the cellular localization of NEDD8. Moreover, using MLN4924 (APP-BP1 pathway blocker), we also verified the components of the downstream effector cascade of the APP-BP1 pathway, including tau, APP-cleaving secretases, ß-catenin and p53. CONCLUSION: These findings indicate that melatonin regulates the interplay of molecular signaling associated with the APP-BP1 pathway and might preclude the pathogenic mechanisms occurring during disease development, thus providing a propitious therapeutic strategy for preventing AD.


Subject(s)
Amyloid beta-Peptides/toxicity , Antioxidants/pharmacology , Melatonin/pharmacology , NEDD8 Protein/metabolism , Peptide Fragments/toxicity , Ubiquitin-Activating Enzymes/antagonists & inhibitors , Ubiquitin-Activating Enzymes/biosynthesis , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Neuroblastoma/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology
2.
Curr Mol Med ; 18(6): 376-382, 2018.
Article in English | MEDLINE | ID: mdl-30479214

ABSTRACT

Background: It is now well established that protein sumoylation is an important mechanism to regulate multiple cellular processes including gene transcription, chromatin structure, cell proliferation and differentiation, as well as pathogenesis. Objective: In the vertebrate eye, we and others have previously shown that sumoylation can regulate differentiation of major ocular tissues including retina and lens. However, the expression patterns of the three types of sumoylation enzymes, the activating enzymes SAE1 and UBA2, the conjugating enzyme UBC9, and the ligating enzymes such as RanBP2 and PIAS1 have not been well studied in the ocular tissues. Conclusion: In the present study, using QRT-PCR and western blot analysis, we have determined the differentiatial expression patterns of the above three types of enzymes, and the obtained results lay down a foundation for further exploration of sumoylation functions in vertebrate eye.


Subject(s)
Eye Proteins/biosynthesis , Gene Expression Regulation/physiology , Lens, Crystalline/metabolism , Molecular Chaperones/biosynthesis , Nuclear Pore Complex Proteins/biosynthesis , Protein Inhibitors of Activated STAT/biosynthesis , Retina/metabolism , Sumoylation/physiology , Ubiquitin-Activating Enzymes/biosynthesis , Ubiquitin-Conjugating Enzymes/biosynthesis , Animals , Female , Lens, Crystalline/cytology , Male , Mice , Retina/cytology
3.
Curr Mol Med ; 18(8): 542-549, 2018.
Article in English | MEDLINE | ID: mdl-30636603

ABSTRACT

PURPOSE: Protein sumoylation is a well established regulatory mechanism that regulates chromatin structure and dynamics, cell proliferation and differentiation, stress response and cell apoptosis. In the vertebrate eye, we and others have shown that sumoylation plays an indispensable role in regulating eye development. During stress induction and aging process, the ocular tissues gradually loss their normality and develop major ocular diseases such as cataract and aging-related macular degeneration. We have recently demonstrated that sumoylation actively regulates differentiation of lens cells, whether this process is implicated in lens pathogenesis remains to be investigated. In this study, we have demonstrated that transparent mouse lenses treated with glucose oxidase and UVA irradiation undergo in vitro cataract formation, and associated with this process, the expression patterns of the 3 sumoylation enzymes have been found significantly altered. METHODS: Four-week-old C57BL/6J mice were used in our experiment. Lenses were carefully excised from eyes and cultured in M199 medium (Sigma 3769) for at least 12 hours. Transparent lenses (without surgical damage) were selected for experimentation. The lenses were exposed to UVA for 60 min or treated with 30 mU/mL glucose oxidase (GO, MP Biomedicals, 1673) to induce cataract formation. The mRNA levels were analysed with qRT-PCR. The protein levels were determined with western blot analysis and quantitated with Image J. RESULTS: we have obtained the following results: 1) Both GO treatment and UVA irradiation can induce cataract formation in the in vitro cultured mouse lenses; 2) With GO treatment, the mRNAs and proteins for the 5 sumoylation enzymes were all significantly downregulated; 3) With UVA irradiation, the changes in the expression patterns of the mRNAs and proteins for the SAE1, UBA2 , UBC9 and PIAS1 were opposite, while the mRNAs were upregulated either significantly (for SAE1, UBA2 and UBC9) or slightly (PIAS1), the proteins for all 4 sumoylation enzymes were downregulated; For RanBP2, the UVA induced changes in both mRNA and protein are consist with the GO treatment. CONCLUSION: Under GO and UVA irradiation conditions, the expression levels of both mRNA and protein for the three major sumoylation enzymes were significantly changed. Our results suggest that altered expression patterns of the sumoylation enzymes are associated with oxidative stressinduced cataractogenesis.


Subject(s)
Cataract , Gene Expression Regulation, Enzymologic/immunology , Glucose Oxidase , Lens, Crystalline , Sumoylation , Ubiquitin-Activating Enzymes , Ultraviolet Rays/adverse effects , Animals , Cataract/enzymology , Cataract/immunology , Cataract/pathology , Glucose Oxidase/immunology , Glucose Oxidase/metabolism , Lens, Crystalline/enzymology , Lens, Crystalline/immunology , Lens, Crystalline/pathology , Mice , RNA, Messenger/biosynthesis , RNA, Messenger/immunology , Sumoylation/immunology , Sumoylation/radiation effects , Ubiquitin-Activating Enzymes/biosynthesis , Ubiquitin-Activating Enzymes/immunology
4.
J Pathol ; 242(1): 39-51, 2017 05.
Article in English | MEDLINE | ID: mdl-28054337

ABSTRACT

The process of liver colonization in colorectal cancer remains poorly characterized. Here, we addressed the role of microRNA (miRNA) dysregulation in metastasis. We first compared miRNA expression profiles between colorectal cancer cell lines with different metastatic properties and then identified target proteins of the dysregulated miRNAs to establish their functions and prognostic value. We found that 38 miRNAs were differentially expressed between highly metastatic (KM12SM/SW620) and poorly metastatic (KM12C/SW480) cancer cell lines. After initial validation, we determined that three miRNAs (miR-424-3p, -503, and -1292) were overexpressed in metastatic colorectal cancer cell lines and human samples. Stable transduction of non-metastatic cells with each of the three miRNAs promoted metastatic properties in culture and increased liver colonization in vivo. Moreover, miR-424-3p and miR-1292 were associated with poor prognosis in human patients. A quantitative proteomic analysis of colorectal cancer cells transfected with miR-424-3p, miR-503, or miR-1292 identified alterations in 149, 129, or 121 proteins, respectively, with an extensive overlap of the target proteins of the three miRNAs. Importantly, down-regulation of two of these shared target proteins, CKB and UBA2, increased cell adhesion and proliferation in colorectal cancer cells. The capacity of distinct miRNAs to regulate the same mRNAs boosts the capacity of miRNAs to regulate cancer metastasis and underscores the necessity of targeting multiple miRNAs for effective cancer therapy. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Colorectal Neoplasms/genetics , MicroRNAs/genetics , Neoplasm Metastasis/genetics , RNA, Neoplasm/genetics , Animals , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Creatine Kinase, BB Form/biosynthesis , Creatine Kinase, BB Form/genetics , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/genetics , Heterografts , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/secondary , Mice, Nude , Neoplasm Proteins/genetics , Neoplasm Transplantation , Prognosis , Proteomics/methods , Ubiquitin-Activating Enzymes/biosynthesis , Ubiquitin-Activating Enzymes/genetics
5.
Tumour Biol ; 36(2): 1173-7, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25342595

ABSTRACT

Autophagy is a cellular degradation process for the clearance of damaged or superfluous proteins and organelles, the recycling of which serves as an alternative energy source during periods of metabolic stress to maintain cell homeostasis and viability. The anti-necrotic function of autophagy is critical for tumorigenesis of many tumor cells, including hepatocellular carcinoma (HCC). However, the underlying mechanism is not clarified yet. Ammonium chloride (NH4Cl) is a well-known autophagy inhibitor, whereas its interaction with SMAD2 signaling pathway has not been reported previously. Here, we show that NH4Cl significantly inhibited rapamycin-induced autophagy in HCC cells through decreasing the levels of Beclin-1, autophagy-related protein 7 (ATG7), p62, and autophagosome marker LC3 and significantly decreased the level of phosphorylated SMAD2 in rapamycin-treated HCC cells. In order to find out whether NH4Cl may inhibit the autophagy in rapamycin-treated HCC cells through inhibition of SMAD2 signaling, we used transforming growth factor ß1 (TGFß1) to induce phosphorylation of SMAD2 in HCC cells. We found that induction of SMAD2 in HCC cells completely abolished the inhibitory effect of NH4Cl on rapamycin-induced autophagy in HCC cells, suggesting that NH4Cl inhibits autophagy of HCC cells through inhibiting SMAD2 signaling.


Subject(s)
Ammonium Chloride/administration & dosage , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Smad2 Protein/genetics , Apoptosis/drug effects , Apoptosis Regulatory Proteins , Autophagy/drug effects , Autophagy-Related Protein 7 , Beclin-1 , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Membrane Proteins , RNA-Binding Proteins/biosynthesis , Signal Transduction/drug effects , Sirolimus/administration & dosage , Smad2 Protein/biosynthesis , Ubiquitin-Activating Enzymes/biosynthesis
6.
Biosci Biotechnol Biochem ; 78(9): 1542-9, 2014.
Article in English | MEDLINE | ID: mdl-25209502

ABSTRACT

Ubiquitin-activating enzyme (E1) is a key regulator in protein ubiquitination, which lies on the upstream of the ubiquitin-related pathways and determines the activation of the downstream enzyme cascade. Thus far, no structural information about the human ubiquitin-activating enzyme has been reported. We expressed and purified the N-terminal domains of human E1 and determined their crystal structures, which contain inactive adenylation domain (IAD) and the first catalytic cysteine half-domain (FCCH). This study presents the crystal structure of human E1 fragment for the first time. The main structure of both IAD and FCCH superimposed well with their corresponding domains in yeast Uba1, but their relative positions vary significantly. This work provides new structural insights in understanding the mechanisms of ubiquitin activation in humans.


Subject(s)
Models, Molecular , Ubiquitin-Activating Enzymes/biosynthesis , Ubiquitin-Activating Enzymes/chemistry , Ubiquitin/chemistry , Amino Acid Sequence , Catalysis , Catalytic Domain , Crystallography, X-Ray , Cysteine , Gene Expression Regulation, Enzymologic , Humans , Protein Structure, Tertiary , Ubiquitin/metabolism , Ubiquitin-Activating Enzymes/isolation & purification
7.
Oncol Rep ; 31(5): 2321-7, 2014 May.
Article in English | MEDLINE | ID: mdl-24626574

ABSTRACT

Autophagy, type II nonapoptotic cell death, is characterized by the formation of double-membrane cytosolic vesicles, the recycling of damaged cytoplasmic content and the maintenance of genetic stability and cellular homeostasis, under conditions of nutrient starvation, hypoxia or other therapeutic stress. In the present study, we comprehensively discuss its indispensable role in the formation of vasculogenic mimicry (VM), capillary-like tubes consisting of cells from the tumor itself instead of vascular endothelial cells. A short hairpin RNA (shRNA) to silence beclin1, an autophagy-specific gene, was designed, synthesized and subcloned into a vector to establish an autophagy-inhibited group, while negative control and blank groups were also established using human gastric cancer SGC7901 cells. We then investigated the VM formation ability of these three groups and detected changes in gene expression, survival and invasion correspondingly. The results showed that, following the formation of VM, the expression of pluripotent genes (c-myc, oct3/4, sox-2) and autophagy-specific genes (beclin1, ATG5, ATG7) were increased, which was consistent with the negative control cell group. However, the autophagy inhibited cell group did not form VM, and the expression of pluripotent genes was decreased. Moreover, the inhibition of autophagy reduced the survival and invasive ability of cancer cells under stress. We suggest that during the formation of VM, the stable expression of genes and the maintenance of survival and invasion are indispensable. Under a stress environment, autophagy is activated to maintain the stability of gene expression, maintain survival and invasive ability and facilitate VM formation, which can provide nutrients, oxygen and invasive channels to tumors, facilitating survival and development under stress.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Membrane Proteins/genetics , Neoplasm Invasiveness/genetics , Neovascularization, Pathologic/genetics , Stomach Neoplasms/blood supply , Autophagy , Autophagy-Related Protein 5 , Autophagy-Related Protein 7 , Beclin-1 , Cell Line, Tumor , Cell Survival/genetics , Humans , Microtubule-Associated Proteins/biosynthesis , Octamer Transcription Factor-3/biosynthesis , Proto-Oncogene Proteins c-myc/biosynthesis , RNA Interference , RNA, Small Interfering , SOXB1 Transcription Factors/biosynthesis , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Ubiquitin-Activating Enzymes/biosynthesis
8.
Mol Biol (Mosk) ; 48(1): 107-16, 2014.
Article in Russian | MEDLINE | ID: mdl-25842831

ABSTRACT

Deregulated expression of proteins involved in the SUMOylation pathway has been detected in several tumors. SUMO1-activating enzyme subunit 1 (SAE1) plays an important role in this process. We found that SAE1 was highly expressed in the colon cancer cell line RKO and used lentivirus-mediated siRNA to suppress SAE1 expressionin RKO cells. RNA-interference efficiently and specifically downregulated the target gene expression in RKO on both mRNA and protein levels. Silencing of SAE1 inhibited proliferation and reduced colony formation of RKO cells. Furthermore, as it has been shown by flow cytometry analysis, specific knockdown of SAE1 slowed down the cell population at G0/G1 phase and induced apoptosis of RKO cells. On the base of results obtained, one can suppose the biological significance of SAE1 in colon tumorigenesis and a use of this protein as a novel molecular target for colon cancer therapy.


Subject(s)
Apoptosis , Cell Cycle Checkpoints , Colonic Neoplasms/enzymology , Gene Expression Regulation, Neoplastic , Gene Targeting , Lentivirus , Neoplasm Proteins/biosynthesis , RNA, Small Interfering/biosynthesis , Ubiquitin-Activating Enzymes/biosynthesis , Cell Line, Tumor , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Down-Regulation/genetics , Gene Expression Regulation, Enzymologic/genetics , HEK293 Cells , Humans , Neoplasm Proteins/genetics , RNA, Small Interfering/genetics , Ubiquitin-Activating Enzymes/genetics
9.
Cancer Biol Ther ; 14(7): 574-86, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23792642

ABSTRACT

ATG7 is a key autophagy-promoting gene that plays a critical role in the regulation of cell death and survival of various cell types. We report here that microRNAs (miRNAs), a class of endogenous 22-24 nucleotide noncoding RNA molecules able to affect stability and translation of mRNA, may represent a novel mechanism for regulating ATG7 expression and therefore autophagy. We demonstrated that ATG7 is a potential target for miR-17, and this miRNA could negatively regulate ATG7 expression, resulting in a modulation of the autophagic status in T98G glioblastoma cells. Treatment of these tumor cells with the miR-17 mimic decreased, and with the antagomir increased, the expression of ATG7 protein. Dual luciferase reporter assay confirmed that a specific miR-17 binding sequence in the 3'-UTR of ATG7 contributed to the modulation of the expression of the gene by miR-17. Interestingly, our results showed that anti-miR-17 administration activated autophagy through autophagosome formation, as resulted by LC3B and ATG7 protein expression increase, and by the analysis of GFP-LC3 positive autophagosome vesicles in living cells. Furthermore, the autophagy activation by anti-miR-17 resulted in a decrease of the threshold resistance at temozolomide doses in T98G cells, while miR-17 modulation in U373-MG glioblastoma cells resulted in a sensitization to low ionizing radiation doses. Our study of the role of miR-17 in regulating ATG7 expression and autophagy reveals a novel function for this miRNA sequence in a critical cellular event with significant impacts in cancer development, progression and treatment.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Dacarbazine/analogs & derivatives , Glioblastoma/genetics , Glioblastoma/therapy , MicroRNAs/metabolism , Ubiquitin-Activating Enzymes/biosynthesis , Autophagy/drug effects , Autophagy/genetics , Autophagy/radiation effects , Autophagy-Related Protein 7 , Cell Line, Tumor , Dacarbazine/pharmacology , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , MicroRNAs/genetics , Radiation, Ionizing , Temozolomide , Ubiquitin-Activating Enzymes/genetics , Ubiquitin-Activating Enzymes/metabolism
10.
Int J Oncol ; 42(3): 993-1000, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23354080

ABSTRACT

Baicalin is one of the major compounds in the traditional Chinese medicinal herb from Scutellaria baicalensis Georgi. We investigated the molecular mechanisms of cell autophagy induced by baicalin in human bladder cancer T24 cells. Baicalin inhibited cell survival as shown by MTT assay and increased cell death by trypan blue exclusion assay in a concentration-dependent manner. Baicalin did not induce apoptotic cell death in T24 cells by TUNEL and caspase-3 activity assay. Baicalin induced the acidic vesicular organelle cell autophagy marker, manifested by acridine orange (AO) and monodansylcadaverine (MDC) staining and cleavage of microtubule-associated protein 1 light chain 3 (LC3). The protein expression levels of the Atg 5, Atg 7, Atg 12, Beclin-1 and LC3-II were upregulated in T24 cells after baicalin treatment. Inhibition of autophagy by 3-methyl-adenine (an inhibitor of class III phosphatidylinositol-3 kinase; 3-MA) reduced the cleavage of LC3 in T24 cells after baicalin treatment. Furthermore, protein expression levels of phospho-AKT (Ser473) and enzyme activity of AKT were downregulated in T24 cells after baicalin treatment. In conclusion, baicalin triggered cell autophagy through the AKT signaling pathway in T24 cells.


Subject(s)
Autophagy/drug effects , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Urinary Bladder Neoplasms/drug therapy , Acridine Orange , Adenine/analogs & derivatives , Adenine/pharmacology , Apoptosis Regulatory Proteins/biosynthesis , Autophagy-Related Protein 12 , Autophagy-Related Protein 5 , Autophagy-Related Protein 7 , Beclin-1 , Cadaverine/analogs & derivatives , Caspase 3 , Cell Line, Tumor , Cell Survival/drug effects , Humans , Medicine, Chinese Traditional , Membrane Proteins/biosynthesis , Microtubule-Associated Proteins/biosynthesis , Microtubule-Associated Proteins/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/biosynthesis , Signal Transduction/drug effects , Small Ubiquitin-Related Modifier Proteins/biosynthesis , Transglutaminases/antagonists & inhibitors , Ubiquitin-Activating Enzymes/biosynthesis
11.
Age (Dordr) ; 35(5): 1531-44, 2013 Oct.
Article in English | MEDLINE | ID: mdl-22843415

ABSTRACT

Type 2 diabetes is characterized by a deficit in ß-cell function and mass, and its incidence increases with age. Autophagy is a highly regulated intracellular process for degrading cytoplasmic components, particularly protein aggregates and damaged organelles. Impaired or deficient autophagy is believed to cause or contribute to aging and age-related disease. Autophagy may be necessary to maintain structure, mass, and function of pancreatic ß-cells. In this study, we investigated the effects of age on ß-cell function and autophagy in pancreatic islets of 4-month-old (young), 14-month-old (adult), and 24-month-old (old) male Wistar rats. We found that islet ß-cell function decreased gradually with age. Protein expression of the autophagy markers LC3/Atg8 and Atg7 exhibited a marked decline in aged islets. The expression of Lamp-2, a good indicator of autophagic degradation rate, was significantly reduced in the islets of old rats, suggesting that autophagic degradation is decreased in the islets of aged rats. However, protein expression of beclin-1/Atg6, which plays an important role in the induction and formation of the pre-autophagosome structure by associating with a multimeric complex of autophagy regulatory proteins (Atg14, Vps34/class 3 PI3 kinase, and Vps15), was most prominent in the islets of adult rats, and was higher in 24-month-old islets than in 4-month-old islets. The levels of p62/SQSTM1 and polyubiquitin aggregates, representing the functions of autophagy and proteasomal degradation, were increased in aging islets. 8-Hydroxydeoxyguanosine, a marker of mitochondrial and nuclear DNA oxidative damage, exhibited strong immunostaining in old islets. Analysis by electron microscopy demonstrated swelling and disintegration of cristae in the mitochondria of aged islets. These results suggest that ß-cell and autophagic function in islets decline simultaneously with increasing age in Wistar rats, and that impaired autophagy in the islets of older rats may cause accumulation of misfolded and aggregated proteins and reduce the removal of abnormal mitochondria in ß-cells, leading to reduced ß-cell function. Dysfunctional autophagy in islets during the aging process may be an important mechanism leading to the development of type 2 diabetes.


Subject(s)
Aging/metabolism , Autophagy , Diabetes Mellitus, Experimental/metabolism , Insulin-Secreting Cells/metabolism , Aging/pathology , Animals , Autophagy-Related Protein 7 , Blotting, Western , Diabetes Mellitus, Experimental/pathology , Disease Progression , Immunohistochemistry , Insulin-Secreting Cells/ultrastructure , Male , Microscopy, Electron , Microtubule-Associated Proteins/biosynthesis , Rats , Rats, Wistar , Ubiquitin-Activating Enzymes/biosynthesis
12.
J Biol Chem ; 287(42): 35576-35588, 2012 Oct 12.
Article in English | MEDLINE | ID: mdl-22927446

ABSTRACT

Tetrandrine, a bisbenzylisoquinoline alkaloid isolated from the broadly used Chinese medicinal herb Stephaniae tetrandrae, exhibits potent antitumor effects and has the potential to be used as a cancer chemotherapeutic agent. We previously reported that high concentrations of tetrandrine induce apoptosis in liver cancer cells. Here, we found that in human hepatocellular carcinoma (HCC) cells, a low dose of tetrandrine (5 µm) induced the expression of LC3-II, resulted in the formation of acidic autophagolysosome vacuoles (AVOs), and caused a punctate fluorescence pattern with the GFP-LC3 protein, which all are markers for cellular autophagy. Tetrandrine induced the production of intracellular reactive oxygen species (ROS), and treatment with ROS scavengers significantly abrogated the tetrandrine-induced autophagy. These results suggest that the generation of ROS plays an important role in promoting tetrandrine-induced autophagy. Tetrandrine-induced mitochondrial dysfunction resulted in ROS accumulation and autophagy. ROS generation activated the ERK MAP kinase, and the ERK signaling pathway at least partially contributed to tetrandrine-induced autophagy in HCC cells. Moreover, we found that tetrandrine transcriptionally regulated the expression of autophagy related gene 7 (ATG7), which promoted tetrandrine-induced autophagy. In addition to in vitro studies, similar results were also observed in vivo, where tetrandrine caused the accumulation of ROS and induced cell autophagy in a tumor xenograft model. Interestingly, tetrandrine treatment also induced autophagy in a ROS-dependent manner in C. elegans muscle cells. Therefore, these findings suggest that tetrandrine is a potent autophagy agonist and may be a promising clinical chemotherapeutic agent.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacokinetics , Autophagy/drug effects , Benzylisoquinolines/pharmacology , Carcinoma, Hepatocellular/drug therapy , Gene Expression Regulation, Neoplastic/drug effects , Liver Neoplasms/drug therapy , MAP Kinase Signaling System/drug effects , Neoplasm Proteins/metabolism , Reactive Oxygen Species/metabolism , Ubiquitin-Activating Enzymes/biosynthesis , Animals , Antineoplastic Agents, Phytogenic/chemistry , Autophagy/genetics , Autophagy-Related Protein 7 , Benzylisoquinolines/chemistry , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Dose-Response Relationship, Drug , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation, Neoplastic/genetics , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , MAP Kinase Signaling System/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Microtubule-Associated Proteins/biosynthesis , Microtubule-Associated Proteins/genetics , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , Neoplasm Proteins/genetics , Stephania tetrandra/chemistry , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , Ubiquitin-Activating Enzymes/genetics , Xenograft Model Antitumor Assays
13.
Biochem Pharmacol ; 84(9): 1154-63, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22902833

ABSTRACT

Multiple lines of evidence support the idea that autophagy plays an essential role in the development of drug resistance, self-renewal, differentiation, and tumorigenic potentials of cancer stem cells (CSCs). Rottlerin (ROT) is widely used as a protein kinase C-delta (PKC-δ) inhibitor. Recent studies revealed that ROT induces apoptosis through engagement of mitochondria. However, it is not known whether ROT-induced apoptosis is associated with other mechanisms such as autophagy. Here we found that ROT induced autophagy followed by induction of apoptosis via inhibition of PI3K/Akt/mTOR pathway and activation of caspase cascade in human pancreatic CSCs. ROT induced a dose- and time-dependent inhibition of cell survival and induction of cytoplasmic vacuolations. The conversion of microtubule-associated protein LC3-I to LC3-II, and increased accumulations of Atg7 and Beclin-1 were also observed in CSCs treated with ROT. Prolonged exposure of CSCs to ROT eventually caused apoptosis which was associated with the suppression of phosphorylated Akt (Ser473) and mTOR (Ser2448), downregulation of XIAP, cIAP-1, Bcl-2 and Bcl-X(L), induction of Bax, activation of caspase-3 and -9, and concomitant degradation of PARP. ROT-induced apoptosis was enhanced by dominant negative AKT, Akt1/2 inhibitor, and rapamycin. Our study also demonstrates that gene silencing of Atg7 and Beclin1, or cotreatment of the autophagosome inhibitor, 3-methyladenine, inhibited ROT-induced autophagy and accelerated ROT-induced apoptosis. The knockdown of PKC-δ did not block ROT-induced autophagy and cell death, suggesting these effects of ROT were exerted through PKC-δ-independent pathway. In summary, our data demonstrate that ROT can induce autophagy which leads to cell death in pancreatic CSCs.


Subject(s)
Acetophenones/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Benzopyrans/pharmacology , Neoplastic Stem Cells/drug effects , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , TOR Serine-Threonine Kinases/physiology , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/genetics , Autophagy/drug effects , Autophagy-Related Protein 7 , Beclin-1 , Caspase 3/metabolism , Caspase 9/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Enzyme Activation , Humans , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Pancreatic Neoplasms , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , RNA, Small Interfering/genetics , Signal Transduction , Ubiquitin-Activating Enzymes/biosynthesis , Ubiquitin-Activating Enzymes/genetics
14.
J Cell Biochem ; 113(6): 2027-39, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22275115

ABSTRACT

Umbilical cord mesenchymal stem cells (MSCs) have been shown to inhibit breast cancer cell growth but it is not known whether this effect is specific to only breast cancer cells. We compared the effects of human Wharton's jelly stem cell (hWJSC) extracts [conditioned medium (hWJSC-CM) and cell lysate (hWJSC-CL)] on breast adenocarcinoma (MDA-MB-231), ovarian carcinoma (TOV-112D), and osteosarcoma (MG-63) cells. The cells were treated with either hWJSC-CM (50%) or hWJSC-CL (15 µg/ml) for 48-72 h and changes in cell morphology, proliferation, cycle, gene expression, migration, and cell death studied. All three cancer cell lines showed cell shrinkage, blebbing, and vacuolations with hWJSC-CL and hWJSC-CM compared to controls. MTT and BrdU assays showed inhibition of cell growth by 2-6% and 30-60%, while Transwell migration assay showed inhibition by 20-26% and 31-46% for hWJSC-CM and hWJSC-CL, respectively, for all three cancer cell lines. Cell cycle assays showed increases in sub-G1 and G2/M phases for all three cancer cell lines suggestive of apoptosis and metaphase arrest. AnnexinV-FITC and TUNEL positive cells seen in TOV-112D and MDA-MB-231 suggested that inhibition was via apoptosis while the presence of anti-BECLIN1 and anti-LC3B antibodies seen with MG-63 indicated autophagy. Upregulation of pro-apoptotic BAX and downregulation of anti-apoptotic BCL2 and SURVIVIN genes were observed in all three cancer cell lines and additionally the autophagy genes (ATG5, ATG7, and BECLIN1) were upregulated in MG-63 cells. hWJSCs possess tumor inhibitory properties that are not specific to breast cancer cells alone and these effects are mediated via agents in its extracts.


Subject(s)
Cell Extracts/pharmacology , Mesenchymal Stem Cells/cytology , Neoplasms/drug therapy , Umbilical Cord/cytology , Umbilical Cord/metabolism , Wharton Jelly/cytology , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Apoptosis/drug effects , Apoptosis Regulatory Proteins/immunology , Autophagy-Related Protein 5 , Autophagy-Related Protein 7 , Beclin-1 , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Cycle/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Culture Media, Conditioned , Female , Gene Expression/drug effects , Humans , Inhibitor of Apoptosis Proteins/biosynthesis , Membrane Proteins/immunology , Mesenchymal Stem Cells/metabolism , Microtubule-Associated Proteins/biosynthesis , Microtubule-Associated Proteins/immunology , Neoplasms/genetics , Neoplasms/pathology , Osteosarcoma/drug therapy , Osteosarcoma/genetics , Osteosarcoma/pathology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Survivin , Ubiquitin-Activating Enzymes/biosynthesis , bcl-2-Associated X Protein/biosynthesis , bcl-Associated Death Protein/biosynthesis
15.
Mol Biotechnol ; 51(3): 254-61, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22012022

ABSTRACT

Research in the ubiquitin field requires large amounts of ubiquitin-activating enzyme (E1) for in vitro ubiquitination assays. Typically, the mammalian enzyme is either isolated from natural sources or produced recombinantly using baculovirus/insect cell protein expression systems. Escherichia coli is seldom used to produce mammalian E1 probably due to the instability and insolubility of this high-molecular mass protein. In this report, we show that 5-10 mg of histidine-tagged mouse E1 can be easily obtained from a 1 l E. coli culture. A low temperature during the protein induction step was found to be critical to obtain an active enzyme.


Subject(s)
Escherichia coli/chemistry , Escherichia coli/enzymology , Recombinant Fusion Proteins/isolation & purification , Ubiquitin-Activating Enzymes/isolation & purification , Animals , Chromatography, High Pressure Liquid , Enzyme Assays , Escherichia coli/genetics , Histidine/analogs & derivatives , Histidine/chemistry , Histidine/genetics , Liver/chemistry , Liver/cytology , Mice , Peroxisome-Targeting Signal 1 Receptor , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Temperature , Ubiquitin-Activating Enzymes/biosynthesis , Ubiquitin-Activating Enzymes/genetics
16.
Int J Cancer ; 131(3): 548-57, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-21858812

ABSTRACT

Multikinase inhibitor sorafenib inhibits proliferation and angiogenesis of tumors by suppressing the Raf/MEK/ERK signaling pathway and VEGF receptor tyrosine kinase. It significantly prolongs median survival of patients with advanced hepatocellular carcinoma (HCC) but the response is disease-stabilizing and cytostatic rather than one of tumor regression. To examine the mechanisms underlying the relative resistance in HCC, we investigated the role of autophagy, an evolutionarily conserved self-digestion pathway, in hepatoma cells in vitro and in vivo. Sorafenib treatment led to accumulation of autophagosomes as evidenced by conversion from LC3-I to LC3-II observed by immunoblot in Huh7, HLF and PLC/PRF/5 cells. This induction was due to activation of autophagic flux, as there was further increase in LC3-II expression upon treatment with lysosomal inhibitors, clear decline of the autophagy substrate p62, and an mRFP-GFP-LC3 fluorescence change in sorafenib-treated hepatoma cells. Sorafenib inhibited the mammalian target of rapamycin complex 1 and its inhibition led to accumulation of LC3-II. Pharmacological inhibition of autophagic flux by chloroquine increased apoptosis and decreased cell viability in hepatoma cells. siRNA-mediated knockdown of the ATG7 gene also sensitized hepatoma cells to sorafenib. Finally, sorafenib induced autophagy in Huh7 xenograft tumors in nude mice and coadministration with chloroquine significantly suppressed tumor growth compared with sorafenib alone. In conclusion, sorafenib administration induced autophagosome formation and enhanced autophagic activity, which conferred a survival advantage to hepatoma cells. Concomitant inhibition of autophagy may be an attractive strategy for unlocking the antitumor potential of sorafenib in HCC.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/drug effects , Benzenesulfonates/pharmacology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Pyridines/pharmacology , Adaptor Proteins, Signal Transducing/biosynthesis , Animals , Apoptosis/drug effects , Autophagy-Related Protein 7 , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Chloroquine/pharmacology , Drug Resistance, Neoplasm , Humans , Liver Neoplasms/metabolism , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Nude , Microtubule-Associated Proteins/metabolism , Multiprotein Complexes , Niacinamide/analogs & derivatives , Phenylurea Compounds , Proteins/antagonists & inhibitors , Proteins/metabolism , RNA Interference , RNA, Small Interfering , Sequestosome-1 Protein , Sorafenib , TOR Serine-Threonine Kinases , Ubiquitin-Activating Enzymes/biosynthesis , Ubiquitin-Activating Enzymes/genetics , Xenograft Model Antitumor Assays
17.
J Pharmacol Exp Ther ; 338(3): 829-40, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21685314

ABSTRACT

NAD is an essential coenzyme involved in numerous metabolic pathways. Its principal role is in redox reactions, and as such it is not heavily "consumed" by cells. Yet a number of signaling pathways that bring about its consumption have recently emerged. This has brought about the hypothesis that the enzymes that lead to its biosynthesis may be targets for anticancer therapy. In particular, inhibition of the enzyme nicotinamide phosphoribosyl transferase has been shown to be an effective treatment in a number of preclinical studies, and two lead molecules [N-[4-(1-benzoyl-4-piperidinyl)butyl]-3-(3-pyridinyl)-2E-propenamide (FK866) and (E)-1-[6-(4-chlorophenoxy)hexyl]-2-cyano-3-(pyridin-4-yl)guanidine (CHS 828)] have now entered preclinical trials. Yet, the full potential of these drugs is still unclear. In the present study we have investigated the role of FK866 in neuroblastoma cell lines. We now confirm that FK866 alone in neuroblastoma cells induces autophagy, and its effects are potentiated by chloroquine and antagonized by 3-methyladenine or by down-regulating autophagy-related protein 7. Autophagy, in this model, seems to be crucial for FK866-induced cell death. On the other hand, a striking potentiation of the effects of cisplatin and etoposide is given by cotreatment of cells with ineffective concentrations of FK866 (1 nM). The effect of etoposide on DNA damage is potentiated by FK866 treatment, whereas the effect of FK866 on cytosolic NAD depletion is potentiated by etoposide. Even more strikingly, cotreatment with etoposide/cisplatin and FK866 unmasks an effect on mitochondrial NAD depletion.


Subject(s)
Acrylamides/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Etoposide/pharmacology , Neuroblastoma/drug therapy , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Piperidines/pharmacology , Acrylamides/antagonists & inhibitors , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine Triphosphate/metabolism , Annexin A5/metabolism , Autophagy/drug effects , Autophagy-Related Protein 7 , Cell Death/drug effects , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Survival/drug effects , Chloroquine/pharmacology , Comet Assay , DNA Damage , Down-Regulation/drug effects , Drug Synergism , Humans , Immunohistochemistry , L-Lactate Dehydrogenase/metabolism , Neuroblastoma/pathology , Piperidines/antagonists & inhibitors , Ubiquitin-Activating Enzymes/biosynthesis , Ubiquitin-Activating Enzymes/genetics
18.
J Biol Chem ; 286(15): 12924-32, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21300795

ABSTRACT

Autophagy has recently been implicated in both the prevention and progression of cancer. However, the molecular basis for the relationship between autophagy induction and the initial acquisition of malignancy is currently unknown. Here, we provide the first evidence that autophagy is essential for oncogenic K-Ras (K-Ras(V12))-induced malignant cell transformation. Retroviral expression of K-Ras(V12) induced autophagic vacuole formation and malignant transformation in human breast epithelial cells. Interestingly, pharmacological inhibition of autophagy completely blocked K-Ras(V12)-induced, anchorage-independent cell growth on soft agar. Both mRNA and protein levels of ATG5 and ATG7 (autophagy-specific genes 5 and 7, respectively) were increased in cells overexpressing K-Ras(V12). Targeted suppression of ATG5 or ATG7 expression by short hairpin (sh) RNA inhibited cell growth on soft agar and tumor formation in nude mice. Moreover, inhibition of reactive oxygen species (ROS) with antioxidants clearly attenuated K-Ras(V12)-induced ATG5 and ATG7 induction, autophagy, and malignant cell transformation. MAPK pathway components were activated in cells overexpressing K-Ras(V12), and inhibition of JNK blunted induction of ATG5 and ATG7 and subsequent autophagy. In addition, pretreatment with antioxidants completely inhibited K-Ras(V12)-induced JNK activation. Our results provide novel evidence that autophagy is critically involved in malignant transformation by oncogenic K-Ras and show that reactive oxygen species-mediated JNK activation plays a causal role in autophagy induction through up-regulation of ATG5 and ATG7.


Subject(s)
Autophagy , Cell Transformation, Neoplastic/metabolism , Epithelial Cells/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Animals , Autophagy-Related Protein 5 , Autophagy-Related Protein 7 , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Epithelial Cells/pathology , Epithelial Cells/transplantation , Female , Humans , Mice , Mice, Nude , Microtubule-Associated Proteins/biosynthesis , Microtubule-Associated Proteins/genetics , Proto-Oncogene Proteins p21(ras)/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Transplantation, Heterologous , Ubiquitin-Activating Enzymes/biosynthesis , Ubiquitin-Activating Enzymes/genetics , Up-Regulation/genetics
19.
Int J Mol Med ; 27(3): 337-44, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21225224

ABSTRACT

Autophagy is an evolutionarily conserved process that maintains cellular homeostasis via synthesis, degradation, and subsequent recycling of cellular products under various physiological conditions. However, the link between autophagy and the innate immune system remains unknown. In the present study, we evaluated Toll-like receptor (TLR)-mediated autophagy induction in intestinal epithelial cells (IECs) and its relationship to interleukin (IL)-8 production. IEC-6, HCT-15, RAW264.7, and THP-1 cells were cultured with or without various TLR ligands, followed by evaluation of the expressions of pro-inflammatory cytokines [IL-8, cytokine-induced neutrophil chemoattractants (CINC)-2ß, macrophage inflammatory protein (MIP)-2] by real-time PCR and ELISA. To reveal the status of autophagy in IECs and macrophages, light chain 3 (LC3)-II expression was examined using Western blotting and immunofluorescence with confocal microscopy. Also, to evaluate the influence of TLR ligands on autophagy-mediated innate-immune responses, autophagy-related gene (Atg)7 specific siRNA was transfected into intestinal epithelial cells and IL-8 expression was determined following exposure to various TLR ligands. Cells treated with the TLR ligands produced considerable amounts of pro-inflammatory cytokines (IL-8, CINC-2ß, MIP-2). Furthermore, the basal levels of LC3-II were markedly higher in IECs as compared to those in macrophages. Our findings indicated that autophagy induction following TLR ligand stimulation was not significantly evident in IECs as compared to macrophages. In addition, Atg7 gene expression silencingled to down-regulation of TLR-mediated IL-8 expression in IECs, which indicates a potential role of autophagy in generating innate-immune responses. In conclusion, autophagy may be an important intracellular machinery for inducing the innate immune system in IECs.


Subject(s)
Autophagy/physiology , Epithelial Cells/metabolism , Gene Expression Regulation/physiology , Interleukin-8/biosynthesis , Intestinal Mucosa/metabolism , Toll-Like Receptors/metabolism , Animals , Autophagy-Related Protein 7 , Cell Line , Chemokine CXCL2/biosynthesis , Chemokine CXCL2/immunology , Chemokines, CXC/biosynthesis , Chemokines, CXC/immunology , Epithelial Cells/immunology , Humans , Immunity, Innate/physiology , Interleukin-8/immunology , Intestinal Mucosa/immunology , Macrophages/immunology , Macrophages/metabolism , Mice , Microtubule-Associated Proteins/biosynthesis , Microtubule-Associated Proteins/immunology , Rats , Toll-Like Receptors/immunology , Ubiquitin-Activating Enzymes/biosynthesis , Ubiquitin-Activating Enzymes/immunology
20.
Mol Biol Cell ; 22(2): 165-78, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21119005

ABSTRACT

The protumorigenic functions for autophagy are largely attributed to its ability to promote cancer cell survival in response to diverse stresses. Here we demonstrate an unexpected connection between autophagy and glucose metabolism that facilitates adhesion-independent transformation driven by a strong oncogenic insult-mutationally active Ras. In cells ectopically expressing oncogenic H-Ras as well as human cancer cell lines harboring endogenous K-Ras mutations, autophagy is induced following extracellular matrix detachment. Inhibiting autophagy due to the genetic deletion or RNA interference-mediated depletion of multiple autophagy regulators attenuates Ras-mediated adhesion-independent transformation and proliferation as well as reduces glycolytic capacity. Furthermore, in contrast to autophagy-competent cells, both proliferation and transformation in autophagy-deficient cells expressing oncogenic Ras are insensitive to reductions in glucose availability. Overall, increased glycolysis in autophagy-competent cells facilitates Ras-mediated adhesion-independent transformation, suggesting a unique mechanism by which autophagy may promote Ras-driven tumor growth in specific metabolic contexts.


Subject(s)
Autophagy , Cell Transformation, Neoplastic/metabolism , ras Proteins/physiology , Animals , Anoikis , Autophagy-Related Protein 12 , Autophagy-Related Protein 7 , Cell Adhesion , Cell Line, Transformed , Cell Proliferation , Cell Transformation, Neoplastic/pathology , Female , Glycolysis , Humans , Mice , Proto-Oncogene Proteins c-bcl-2/biosynthesis , RNA Interference , Small Ubiquitin-Related Modifier Proteins/biosynthesis , Small Ubiquitin-Related Modifier Proteins/genetics , Tumor Cells, Cultured , Ubiquitin-Activating Enzymes/biosynthesis , Ubiquitin-Activating Enzymes/genetics , ras Proteins/biosynthesis , ras Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...