Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 76
1.
Int J Mol Sci ; 23(14)2022 Jul 13.
Article En | MEDLINE | ID: mdl-35887068

Nonalcoholic fatty liver disease (NAFLD) is characterized by the accumulation of lipids in the liver. Given the high prevalence of NAFLD, its evolution to nonalcoholic steatohepatitis (NASH) and hepatocellular carcinoma (HCC) is of global concern. Therapies for managing NASH-driven HCC can benefit from targeting factors that play a continuous role in NAFLD evolution to HCC. Recent work has shown that postprandial liver translation exacerbates lipid accumulation through the activity of a translation factor, eukaryotic initiation factor 6 (eIF6). Here, we test the effect of eIF6 inhibition on the progression of HCC. Mice heterozygous for eIF6 express half the level of eIF6 compared to wt mice and are resistant to the formation of HCC nodules upon exposure to a high fat/high sugar diet combined with liver damage. Histology showed that nodules in eIF6 het mice were smaller with reduced proliferation compared to wt nodules. By using an in vitro model of human HCC, we confirm that eIF6 depletion reduces the growth of HCC spheroids. We also tested three pharmacological inhibitors of eIF6 activity-eIFsixty-1, eIFsixty-4, and eIFsixty-6-and all three reduced eIF6 binding to 60S ribosomes and limited the growth of HCC spheroids. Thus, inhibition of eIF6 activity is feasible and limits HCC formation.


Carcinoma, Hepatocellular , Liver Neoplasms , Non-alcoholic Fatty Liver Disease , Animals , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Eukaryotic Initiation Factors/antagonists & inhibitors , Eukaryotic Initiation Factors/genetics , Eukaryotic Initiation Factors/metabolism , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Mice , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Peptide Initiation Factors/antagonists & inhibitors , Peptide Initiation Factors/genetics , Peptide Initiation Factors/metabolism
2.
Nat Commun ; 12(1): 4878, 2021 08 12.
Article En | MEDLINE | ID: mdl-34385447

A postprandial increase of translation mediated by eukaryotic Initiation Factor 6 (eIF6) occurs in the liver. Its contribution to steatosis and disease is unknown. In this study we address whether eIF6-driven translation contributes to disease progression. eIF6 levels increase throughout the progression from Non-Alcoholic Fatty Liver Disease (NAFLD) to hepatocellular carcinoma. Reduction of eIF6 levels protects the liver from disease progression. eIF6 depletion blunts lipid accumulation, increases fatty acid oxidation (FAO) and reduces oncogenic transformation in vitro. In addition, eIF6 depletion delays the progression from NAFLD to hepatocellular carcinoma, in vivo. Mechanistically, eIF6 depletion reduces the translation of transcription factor C/EBPß, leading to a drop in biomarkers associated with NAFLD progression to hepatocellular carcinoma and preserves mitochondrial respiration due to the maintenance of an alternative mTORC1-eIF4F translational branch that increases the expression of transcription factor YY1. We provide proof-of-concept that in vitro pharmacological inhibition of eIF6 activity recapitulates the protective effects of eIF6 depletion. We hypothesize the existence of a targetable, evolutionarily conserved translation circuit optimized for lipid accumulation and tumor progression.


Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Non-alcoholic Fatty Liver Disease/genetics , Peptide Initiation Factors/genetics , Protein Biosynthesis/genetics , Animals , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , Carcinoma, Hepatocellular/metabolism , Cell Line , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Clofazimine/pharmacology , Diet, High-Fat/adverse effects , Disease Progression , Gene Silencing , Humans , Lipogenesis/drug effects , Lipogenesis/genetics , Liver Neoplasms/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease/metabolism , Obesity/etiology , Obesity/genetics , Obesity/metabolism , Peptide Initiation Factors/antagonists & inhibitors , Peptide Initiation Factors/metabolism
3.
Biochem Biophys Res Commun ; 519(4): 838-845, 2019 11 19.
Article En | MEDLINE | ID: mdl-31558321

Metastasis is the leading cause of mortality in patients with solid tumors. In this regard, we previously reported that Pseudopodium-Enriched Atypical Kinase One (PEAK1) is necessary for non-canonical Transforming Growth Factor ß (TGFß) signaling and TGFß/fibronectin-induced metastasis. Here, we demonstrate that inhibition of DHPS-dependent eIF5A1/2 hypusination blocks PEAK1 and E-Cadherin expression, breast cancer cell viability and TGFß/fibronectin-induced PEAK1-dependent breast cancer metastasis. Interestingly, TGFß stimulation of high-grade metastatic breast cancer cells increases and sustains eIF5A1/2 hypusination. We used a suite of bioinformatics platforms to search biochemical/functional interactions and clinical databases for additional control points in eIF5A1/2 and PEAK1-Epithelial to Mesenchymal Transition (EPE) pathways. This effort revealed that interacting EPE genes were enriched for TP53 transcriptional targets and were commonly co-amplified in breast cancer patients harboring inactivating TP53 mutations. Taken together, these results suggest that combinatorial therapies targeting DHPS and protein activities elevated in TP53-mutant breast cancers may reduce systemic tumor burden and improve patient outcomes.


Breast Neoplasms/metabolism , Fibronectins/metabolism , Oxidoreductases Acting on CH-NH Group Donors/metabolism , Peptide Initiation Factors/metabolism , RNA-Binding Proteins/metabolism , Transforming Growth Factor beta/metabolism , Tumor Suppressor Protein p53/genetics , Breast Neoplasms/pathology , Cadherins/antagonists & inhibitors , Cadherins/genetics , Cadherins/metabolism , Female , Guanine/analogs & derivatives , Guanine/pharmacology , Humans , Oxidoreductases Acting on CH-NH Group Donors/antagonists & inhibitors , Peptide Initiation Factors/antagonists & inhibitors , Prognosis , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , RNA-Binding Proteins/antagonists & inhibitors , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism , Eukaryotic Translation Initiation Factor 5A
4.
Sci Rep ; 9(1): 1533, 2019 02 07.
Article En | MEDLINE | ID: mdl-30733517

We have developed a transgenic mouse model of Type 1 Diabetes (T1D) in which human GAD65 is expressed in pancreatic ß-cells, and human MHC-II is expressed on antigen presenting cells. Induced GAD65 antigen presentation activates T-cells, which initiates the downstream events leading to diabetes. In our humanized mice, we have shown downregulation of eukaryotic translation initiation factor 5 A (elF5A), expressed only in actively dividing mammalian cells. In-vivo inhibition of elF5A hypusination by deoxyhypusine synthase (DHS) inhibitor "GC7" was studied; DHS inhibitor alters the pathophysiology in our mouse model by catalyzing the crucial hypusination and the rate-limiting step of elF5A activation. In our mouse model, we have shown that inhibition of eIF5A resets the pro-inflammatory bias in the pancreatic microenvironment. There was: (a) reduction of Th1/Th17 response, (b) an increase in Treg numbers, (c) debase in IL17 and IL21 cytokines levels in serum, (d) lowering of anti-GAD65 antibodies, and (e) ablation of the ER stress that improved functionality of the ß-cells, but minimal effect on the cytotoxic CD8 T-cell (CTL) mediated response. Conclusively, immune modulation, in the case of T1D, may help to manipulate inflammatory responses, decreasing disease severity, and may help manage T1D in early stages of disease. Our study also demonstrates that without manipulating the CTLs mediated response extensively, it is difficult to treat T1D.


Enzyme Inhibitors/chemistry , Glutamate Decarboxylase/genetics , Peptide Initiation Factors/metabolism , RNA-Binding Proteins/metabolism , T-Lymphocytes/metabolism , Animals , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Down-Regulation/drug effects , Endoplasmic Reticulum Stress/drug effects , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation/drug effects , Glutamate Decarboxylase/metabolism , Heptanes/chemistry , Heptanes/metabolism , Heptanes/pharmacology , Humans , Insulin-Secreting Cells/metabolism , Interleukins/blood , Male , Mice , Mice, Transgenic , Oxidoreductases Acting on CH-NH Group Donors/antagonists & inhibitors , Oxidoreductases Acting on CH-NH Group Donors/metabolism , Peptide Initiation Factors/antagonists & inhibitors , Peptide Initiation Factors/genetics , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , T-Lymphocytes/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Eukaryotic Translation Initiation Factor 5A
5.
J Cell Mol Med ; 23(4): 2678-2688, 2019 04.
Article En | MEDLINE | ID: mdl-30761741

BACKGROUND: Pancreatic cancer (PC) has a very poor prognosis and comparatively short survival. Eukaryotic translation initiation factor 5A (EIF5A) promotes cancer metastasis. Here, we exploited the biological role of EIF5A in PC chemoresistance. METHODS: Expression of EIF5A was analysed in PC cells and tissues by real-time PCR, Western blotting, immunohistochemistry and immunofluorescent. EIF5A expression was specifically suppressed by transfection, and subsequently the alterations of growth behaviour and resistance to anticancer treatment were tested in an orthotopic tumour model. RESULTS: The results showed EIF5A was increased in human PC tissues and PC cells. We found EIF5A knockdown reduced the PC proliferation ability in vivo and in vitro. In addition, sonic hedgehog (sHH) signalling pathway may be a downstream of EIF5A in PC cells. Inhibition of EIF5A and sHH signalling pathway could suppress PC cells proliferation and tumour growth. Importantly, EIF5A played an important role in gemcitabine sensitivity for PC. CONCLUSION: Taken together, our results revealed that EIF5A regulated the proliferation of PC through the sHH signalling pathway and decreased the Gem sensitivity in PC, which provided a novel therapeutic strategy for PC patients.


Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Hedgehog Proteins/genetics , Pancreatic Neoplasms/genetics , Peptide Initiation Factors/genetics , RNA-Binding Proteins/genetics , Animals , Antimetabolites, Antineoplastic/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Hedgehog Proteins/metabolism , Humans , Mice , Mice, Nude , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Peptide Initiation Factors/antagonists & inhibitors , Peptide Initiation Factors/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/metabolism , Signal Transduction , Tumor Burden/drug effects , Xenograft Model Antitumor Assays , Zinc Finger Protein GLI1/genetics , Zinc Finger Protein GLI1/metabolism , Gemcitabine , Eukaryotic Translation Initiation Factor 5A
6.
Biomed Pharmacother ; 100: 168-175, 2018 Apr.
Article En | MEDLINE | ID: mdl-29428664

Epithelial ovarian cancer (EOC) is one of the most common gynecological cancers and has the highest mortality rate thereof. We found abundant eukaryotic translation initiation factor 5A1 (EIF5A1) in 54 EOC tissues, and high EIF5A1 levels predicted poor survival. EIF5A1 ectopic expression enhanced EOC cell proliferative, migration, and invasive capabilities, while EIF5A1 knockdown suppressed them. Most importantly, GC7 (N1-guanyl-1,7-diaminoheptane, an EIF5A1 hypusination inhibitor) could reverse the effect of EIF5A1 upregulation on EOC cell proliferation, migration, and invasion and mutant type EIF5A1K50A plasmid [bearing a single point mutation (K50 → A50) that prevents hypusination] had no effects on these malignant behaviors. Our findings imply that EIF5A1 is a vital regulator of EOC proliferation and progression and is a potential prognostic marker and therapeutic target in EOC.


Cell Movement/genetics , Cell Proliferation/genetics , Epithelial Cells/pathology , Neoplasms, Glandular and Epithelial/metabolism , Ovarian Neoplasms/metabolism , Ovary/pathology , Peptide Initiation Factors/metabolism , RNA-Binding Proteins/metabolism , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Epithelial Cells/metabolism , Female , Guanine/analogs & derivatives , Guanine/pharmacology , Humans , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasms, Glandular and Epithelial/genetics , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Ovary/metabolism , Peptide Initiation Factors/antagonists & inhibitors , Peptide Initiation Factors/genetics , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , Up-Regulation , Eukaryotic Translation Initiation Factor 5A
7.
Biochem J ; 475(2): 531-545, 2018 01 31.
Article En | MEDLINE | ID: mdl-29295892

The eukaryotic initiation factor 5A (eIF5A), which contributes to several crucial processes during protein translation, is the only protein that requires activation by a unique post-translational hypusine modification. eIF5A hypusination controls cell proliferation and has been linked to cancer. eIF5A hypusination requires the enzymes deoxyhypusine synthase (DHPS) and deoxyhypusine hydroxylase and uniquely depends on the polyamine (PA) spermidine as the sole substrate. Ornithine decarboxylase (ODC) is the rate-limiting enzyme in PA biosynthesis. Both ODC and PAs control cell proliferation and are frequently dysregulated in cancer. Since only spermidine can activate eIF5A, we chose the hypusine-PA nexus as a rational target to identify new drug combinations with synergistic antiproliferative effects. We show that elevated mRNA levels of the two target enzymes DHPS and ODC correlate with poor prognosis in a large cohort of neuroblastoma (NB) tumors. The DHPS inhibitor GC7 (N1-guanyl-1,7-diaminoheptane) and the ODC inhibitor α-difluoromethylornithine (DFMO) are target-specific and in combination induced synergistic effects in NB at concentrations that were not individually cytotoxic. Strikingly, while each drug alone at higher concentrations is known to induce p21/Rb- or p27/Rb-mediated G1 cell cycle arrest, we found that the drug combination induced caspase 3/7/9, but not caspase 8-mediated apoptosis, in NB cells. Hypusinated eIF5A levels and intracellular spermidine levels correlated directly with drug treatments, signifying specific drug targeting effects. This two-pronged GC7/DFMO combination approach specifically inhibits both spermidine biosynthesis and post-translational, spermidine-dependent hypusine-eIF5A activation, offering an exciting clue for improved NB drug therapy.


Antineoplastic Combined Chemotherapy Protocols , Eflornithine/pharmacology , Gene Expression Regulation, Neoplastic , Guanine/analogs & derivatives , Nervous System Neoplasms/genetics , Neuroblastoma/genetics , Peptide Initiation Factors/genetics , RNA-Binding Proteins/genetics , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Drug Synergism , Guanine/pharmacology , Humans , Lysine/analogs & derivatives , Lysine/metabolism , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/metabolism , Nervous System Neoplasms/metabolism , Nervous System Neoplasms/mortality , Nervous System Neoplasms/pathology , Neuroblastoma/metabolism , Neuroblastoma/mortality , Neuroblastoma/pathology , Ornithine Decarboxylase/genetics , Ornithine Decarboxylase/metabolism , Oxidoreductases Acting on CH-NH Group Donors/genetics , Oxidoreductases Acting on CH-NH Group Donors/metabolism , Peptide Initiation Factors/antagonists & inhibitors , Peptide Initiation Factors/metabolism , Prognosis , Protein Processing, Post-Translational , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/metabolism , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Signal Transduction , Spermidine/metabolism , Eukaryotic Translation Initiation Factor 5A
8.
Oncol Rep ; 39(3): 1283-1291, 2018 Mar.
Article En | MEDLINE | ID: mdl-29286162

Eukaryotic initiation factor 5A2 (eIF5A2), a newly identified oncogene, promotes cell survival, proliferation and motility in tumorigenesis. Drug resistance and dose-related adverse side-effects greatly reduce the efficiency and safety of cisplatin-based chemotherapy in advanced or recurrent oral squamous cell carcinoma (OSCC) patients. The present study investigated the effect of eIF5A2 combined with N1-guanyl-1,7-diaminoheptane (GC7, a novel eIF5A2 inhibitor) or siRNA. We found that low concentrations of GC7 (≤5 µM) had little effect on OSCC cell viability, but significantly enhanced cisplatin cytotoxicity. Compared with cisplatin, GC7/cisplatin had little effect on cisplatin-promoted mesenchymal-epithelial transition in mesenchymal phenotype Tca8113 and HN30 cells, or on cisplatin-induced epithelial-mesenchymal transition (EMT) in epithelial phenotype Cal27 and HN4 cells. Further research revealed that the upregulation of p-STAT3 and c-Myc which was induced by the single treatment with either cisplatin or GC7 was significantly reversed by the GC7/cisplatin combination in mesenchymal phenotype Tca8113 and HN30 cells. In in vivo treatment, we revealed that the GC7/cisplatin combination presented significant tumor volume reduction without distinct body weight loss. In conclusion, our data indicated that eIF5A2 is a potent therapeutic target in OSCC treatment. Our results revealed a novel mechanism by which GC7/cisplatin combination therapy may offer an efficient and safe therapeutic alternative to advanced or recurrent OSCC patients.


Cisplatin/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Guanine/analogs & derivatives , Mouth Neoplasms/drug therapy , Mouth Neoplasms/pathology , Peptide Initiation Factors/antagonists & inhibitors , RNA-Binding Proteins/antagonists & inhibitors , STAT3 Transcription Factor/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Biomarkers, Tumor , Cell Proliferation/drug effects , Drug Synergism , Guanine/pharmacology , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Mouth Neoplasms/metabolism , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Eukaryotic Translation Initiation Factor 5A
9.
Anticancer Drugs ; 28(10): 1097-1105, 2017 11.
Article En | MEDLINE | ID: mdl-28885268

N1-guanyl-1,7-diaminoheptane (GC7), a deoxyhypusine synthase inhibitor, has been shown to exert antiproliferation effects in many solid tumors by regulating eukaryotic translation initiation factor 5a2 (eif5a-2). However, little is known about the role of GC7 and eif5a-2 in drug resistance in acute lymphoblastic leukemia (ALL). In the present study, we investigated the effect of GC7 on drug-resistant ALL and its potential mechanism. We found that using the CCK-8 assay that combined treatment with GC7 and vincristine (VCR) significantly inhibited the cell viability of two ALL cell lines. Using EdU incorporation assays and flow cytometry, we also showed that GC7 could markedly enhance the VCR sensitivity of ALL cells by suppressing cell proliferation and promoting apoptosis. Furthermore, we showed that GC7 could downregulate eif5a-2 and myeloid cell leukemia-1 (Mcl-1) expression. Knockdown of eif5a-2 inhibited the expression of Mcl-1 and significantly enhanced the VCR sensitivity. Moreover, eif5a-2 knockdown decreased the regulatory role of GC7 in increasing VCR sensitivity. Thus, our findings indicate that combined treatment with GC7 could enhance VCR sensitivity of ALL cells by regulating the eif5a-2/Mcl-1 axis. Together, our results highlight the potential clinical application of GC7 in VCR-based chemotherapy for the treatment of ALL.


Antineoplastic Combined Chemotherapy Protocols/pharmacology , Guanine/analogs & derivatives , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Vincristine/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Drug Resistance, Neoplasm , Drug Synergism , Gene Knockdown Techniques , Guanine/administration & dosage , Guanine/pharmacology , Humans , Jurkat Cells , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Peptide Initiation Factors/antagonists & inhibitors , Peptide Initiation Factors/genetics , Peptide Initiation Factors/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Vincristine/administration & dosage , Eukaryotic Translation Initiation Factor 5A
10.
Sci Rep ; 5: 16911, 2015 Nov 23.
Article En | MEDLINE | ID: mdl-26593060

Spinal cord injury (SCI) is frequently accompanied by a degree of spontaneous functional recovery. The underlying mechanisms through which such recovery is generated remain elusive. In this study, we observed a significant spontaneous motor function recovery 14 to 28 days after spinal cord transection (SCT) in rats. Using a comparative proteomics approach, caudal to the injury, we detected difference in 20 proteins. Two of these proteins, are eukaryotic translation initiation factor 5A1 (eIF5A1) that is involved in cell survival and proliferation, and Rho GDP dissociation inhibitor alpha (RhoGDIα), a member of Rho GDI family that is involved in cytoskeletal reorganization. After confirming the changes in expression levels of these two proteins following SCT, we showed that in vivo eIF5A1 up-regulation and down-regulation significantly increased and decreased, respectively, motor function recovery. In vitro, eIF5A1 overexpression in primary neurons increased cell survival and elongated neurite length while eIF5A1 knockdown reversed these results. We found that RhoGDIα up-regulation and down-regulation rescues the effect of eIF5A1 down-regulation and up-regulation both in vivo and in vitro. Therefore, we have identified eIF5A1/RhoGDIα pathway as a new therapeutic target for treatment of spinal cord injured patients.


Peptide Initiation Factors/genetics , RNA-Binding Proteins/genetics , Recovery of Function/physiology , Signal Transduction/genetics , Spinal Cord Injuries/genetics , rho Guanine Nucleotide Dissociation Inhibitor alpha/genetics , Amino Acid Sequence , Animals , Cell Line , Cell Proliferation/genetics , Cell Survival/genetics , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Female , Gene Expression Regulation , Humans , Molecular Sequence Data , Motor Activity/physiology , Neurons/metabolism , Neurons/ultrastructure , Peptide Initiation Factors/antagonists & inhibitors , Peptide Initiation Factors/metabolism , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/metabolism , Rats , Rats, Sprague-Dawley , Remission, Spontaneous , Spinal Cord/metabolism , Spinal Cord/pathology , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , rho Guanine Nucleotide Dissociation Inhibitor alpha/antagonists & inhibitors , rho Guanine Nucleotide Dissociation Inhibitor alpha/metabolism , Eukaryotic Translation Initiation Factor 5A
11.
Sci Rep ; 5: 13737, 2015 Sep 08.
Article En | MEDLINE | ID: mdl-26348594

Oxidative stress plays a critical role in ischemia/reperfusion-injury, atherosclerosis, and aging. It causes cell damage that leads to apoptosis via uncertain mechanisms. Because conditioned medium from cardiac myocytes subjected to hypoxia/reoxygenation induces extensive apoptosis of cardiac myocytes under normoxia, we hypothesized that a humoral factor released from the hypoxic/reoxygenated cardiac myocytes mediates apoptosis. We identified an apoptosis-inducing humoral factor in the hypoxia/reoxygenation-conditioned medium. Here, we found that eIF5A undergoes tyrosine sulfation in the trans-Golgi and is rapidly secreted from cardiac myocytes in response to hypoxia/reoxygenation; then, eIF5A induces apoptosis by acting as a pro-apoptotic ligand. The apoptosis of cardiac myocytes induced by hypoxia/reoxygenation or ultraviolet irradiation was suppressed by anti-eIF5A neutralizing monoclonal antibodies (mAbs) in vitro. Myocardial ischemia/reperfusion (but not ischemia alone) markedly increased the plasma levels of eIF5A, and treatment with anti-eIF5A neutralizing mAbs significantly reduced myocardial injury. These results identify an important, novel specific biomarker and a critical therapeutic target for oxidative stress-induced cell injury.


Apoptosis , Myocytes, Cardiac/metabolism , Oxidative Stress , Peptide Initiation Factors/metabolism , RNA-Binding Proteins/metabolism , Tyrosine/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Apoptosis/genetics , Disease Models, Animal , Golgi Apparatus/metabolism , Humans , Hypoxia/metabolism , Male , Models, Biological , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Oxidative Stress/genetics , Oxygen/metabolism , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Peptide Initiation Factors/antagonists & inhibitors , Peptide Initiation Factors/genetics , Protein Transport , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , Rats , Signal Transduction , trans-Golgi Network/metabolism , Eukaryotic Translation Initiation Factor 5A
12.
Toxicol Sci ; 147(2): 326-38, 2015 Oct.
Article En | MEDLINE | ID: mdl-26141394

Chronic exposure to trichothecenes is known to disturb insulin-like growth factor 1 and signaling of insulin and leptin hormones and causes considerable growth retardation in animals. However, limited information was available on mechanisms underlying trichothecene-induced growth retardation. In this study, we employed an integrated transcriptomics, proteomics, and RNA interference (RNAi) approach to study the molecular mechanisms underlying trichothecene cytotoxicity in rat pituitary adenoma GH3 cells. Our results showed that trichothecenes suppressed the synthesis of growth hormone 1 (Gh1) and inhibited the eukaryotic transcription and translation initiation by suppressing aminoacyl-tRNA synthetases transcription, inducing eukaryotic translation initiation factor 2-alpha kinase 2 (EIF2AK2) and reducing eukaryotic translation initiation factor 5 a. The sulfhydryl oxidases , protein disulfide isomerase,and heat shock protein 90 (were greatly reduced, which resulted in adverse regulation of protein processing and folding. Differential genes and proteins associated with a decline in energy metabolism and cell cycle arrest were also found in our study. However, use of RNAi to interfere with hemopoietic cell kinase (Hck) and EIF2AK2 transcriptions or use of chemical inhibitors of MAPK, p38, Ras, and JNK partially reversed the reduction of Gh1 levels induced by trichothecenes. It indicated that the activation of MAPKs, Hck, and EIF2AK2 were important for trichothecene-induced growth hormone suppression. Considering the potential hazards of exposure to trichothecenes, our findings could help to improve our understanding regarding human and animal health implications.


Gene Expression Profiling , Growth Hormone/antagonists & inhibitors , Proteomics , T-2 Toxin/analogs & derivatives , Amino Acyl-tRNA Synthetases/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Line , Cell Survival/drug effects , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Oxidoreductases/antagonists & inhibitors , Peptide Initiation Factors/antagonists & inhibitors , Protein Biosynthesis/drug effects , Protein Disulfide-Isomerases/antagonists & inhibitors , RNA Interference/drug effects , RNA-Binding Proteins/antagonists & inhibitors , Rats , T-2 Toxin/pharmacology , Transcription, Genetic/drug effects , eIF-2 Kinase/antagonists & inhibitors , Eukaryotic Translation Initiation Factor 5A
13.
J Biol Chem ; 290(32): 19987-98, 2015 Aug 07.
Article En | MEDLINE | ID: mdl-26082486

The eukaryotic protozoan parasite Trypanosoma brucei is the causative agent of human African trypanosomiasis. Polyamine biosynthesis is essential in T. brucei, and the polyamine spermidine is required for synthesis of a novel cofactor called trypanothione and for deoxyhypusine modification of eukaryotic translation initiation factor 5A (eIF5A). eIF5A promotes translation of proteins containing polyprolyl tracts in mammals and yeast. To evaluate the function of eIF5A in T. brucei, we used RNA interference (RNAi) to knock down eIF5A levels and found that it is essential for T. brucei growth. The RNAi-induced growth defect was complemented by expression of wild-type human eIF5A but not by a Lys-50 mutant that blocks modification by deoxyhypusine. Bioinformatics analysis showed that 15% of the T. brucei proteome contains 3 or more consecutive prolines and that actin-related proteins and cysteine proteases were highly enriched in the group. Steady-state protein levels of representative proteins containing 9 consecutive prolines that are involved in actin assembly (formin and CAP/Srv2p) were significantly reduced by knockdown of eIF5A. Several T. brucei polyprolyl proteins are involved in flagellar assembly. Knockdown of TbeIF5A led to abnormal cell morphologies and detached flagella, suggesting that eIF5A is important for translation of proteins needed for these processes. Potential specialized functions for eIF5A in T. brucei in translation of variable surface glycoproteins were also uncovered. Inhibitors of deoxyhypusination would be expected to cause a pleomorphic effect on multiple cell processes, suggesting that deoxyhypusine/hypusine biosynthesis could be a promising drug target in not just T. brucei but in other eukaryotic pathogens.


Lysine/analogs & derivatives , Peptide Initiation Factors/metabolism , Protein Processing, Post-Translational , Protozoan Proteins/metabolism , RNA, Messenger/metabolism , RNA, Protozoan/metabolism , RNA-Binding Proteins/metabolism , Trypanosoma brucei brucei/metabolism , Amino Acid Sequence , Animals , Escherichia coli/genetics , Escherichia coli/metabolism , Flagella/genetics , Flagella/metabolism , Flagella/ultrastructure , Gene Knockdown Techniques , Humans , Lysine/metabolism , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Molecular Sequence Data , Peptide Initiation Factors/antagonists & inhibitors , Peptide Initiation Factors/genetics , Peptides/metabolism , Proteome/genetics , Proteome/metabolism , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/genetics , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Protozoan/antagonists & inhibitors , RNA, Protozoan/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Trypanosoma brucei brucei/genetics , Trypanosoma brucei brucei/ultrastructure , Eukaryotic Translation Initiation Factor 5A
14.
BMC Pulm Med ; 14: 174, 2014 Nov 07.
Article En | MEDLINE | ID: mdl-25380840

BACKGROUND: Epithelial-mesenchymal transition (EMT) has been believed to be related with chemotherapy resistance in non-small cell lung cancer (NSCLC). Recent studies have suggested eIF5A-2 may function as a proliferation-related oncogene in tumorigenic processes. METHODS: We used cell viability assays, western blotting, immunofluorescence, transwell-matrigel invasion assay, wound-healing assay combined with GC7 (a novel eIF5A-2 inhibitor) treatment or siRNA interference to investigate the role of eIF5A-2 playing in NSCLC chemotherapy. RESULTS: We found low concentrations of GC7 have little effect on NSCLC viability, but could enhance cisplatin cytotoxicity in NSCLC cells. GC7 also could reverse mesenchymal phenotype in NCI-H1299 and prevented A549 cells undergoing EMT after TGF-ß1 inducement. eIF5A-2 knockdown resulted in EMT inhibition. CONCLUSION: Our data indicated GC7 enhances cisplatin cytotoxicity and prevents the EMT in NSCLC cells by inhibiting eIF5A-2.


Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Cisplatin/pharmacology , Enzyme Inhibitors/pharmacology , Guanine/analogs & derivatives , Lung Neoplasms/drug therapy , Peptide Initiation Factors/antagonists & inhibitors , RNA-Binding Proteins/antagonists & inhibitors , Cadherins/metabolism , Cell Line, Tumor , Cell Movement , Cell Survival/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Epithelial-Mesenchymal Transition/drug effects , Gene Silencing , Guanine/pharmacology , Humans , Inhibitory Concentration 50 , Oxidoreductases Acting on CH-NH Group Donors/antagonists & inhibitors , Peptide Initiation Factors/genetics , RNA, Small Interfering , RNA-Binding Proteins/genetics , Vimentin/metabolism , Eukaryotic Translation Initiation Factor 5A
15.
Mol Ther ; 22(9): 1643-52, 2014 Sep.
Article En | MEDLINE | ID: mdl-24569836

The high rates of recurrence and low median survival in many B-cell cancers highlight a need for new targeted therapeutic modalities. In dividing cells, eukaryotic translation initiation factor 5A (eIF5A) is hypusinated and involved in regulation of protein synthesis and proliferation, whereas the non-hypusinated form of eIF5A is a potent inducer of cell death in malignant cells. Here, we demonstrate the potential of modulating eIF5A expression as a novel approach to treating B-cell cancers. SNS01-T is a nonviral polyethylenimine-based nanoparticle, designed to induce apoptosis selectively in B-cell cancers by small interfering RNA-mediated suppression of hypusinated eIF5A and plasmid-based overexpression of a non-hypusinable eIF5A mutant. In this study, we show that SNS01-T is preferentially taken up by malignant B cells, inhibits tumor growth in multiple animal models of B-cell cancers without damaging normal tissues, and synergizes with the current therapies bortezomib and lenalidomide to inhibit tumor progression. The results collectively demonstrate the potential of SNS01-T as a novel therapeutic for treatment of a diverse range of B-cell malignancies.


Boronic Acids/therapeutic use , Lymphoproliferative Disorders/therapy , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Peptide Initiation Factors/antagonists & inhibitors , Pyrazines/therapeutic use , RNA, Small Interfering/administration & dosage , RNA-Binding Proteins/antagonists & inhibitors , Thalidomide/analogs & derivatives , Animals , Bortezomib , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Synergism , Humans , Lenalidomide , Lymphoproliferative Disorders/genetics , Lymphoproliferative Disorders/pathology , Mice , Nanoparticles/therapeutic use , Neoplasm Transplantation , Polyethyleneimine/chemistry , RNA, Small Interfering/therapeutic use , Thalidomide/therapeutic use , Xenograft Model Antitumor Assays , Eukaryotic Translation Initiation Factor 5A
16.
Am J Physiol Endocrinol Metab ; 306(7): E791-8, 2014 Apr 01.
Article En | MEDLINE | ID: mdl-24496311

Therapeutic options for treatment of type 1 diabetes (T1D) are still missing. New avenues for immune modulation need to be developed. Here we attempted at altering the diabetes outcome of our humanized model of T1D by inhibiting translation-initiation factor eIF5A hypusination in vivo. Double-transgenic (DQ8-GAD65) mice were immunized with adenoviral vectors carrying GAD65 for diabetes induction. Animals were subsequently treated with deoxyhypusine synthase (DHS) inhibitor GC7 and monitored for diabetes development over time. On one hand, helper CD4(+) T cells were clearly affected by the downregulation of the eIF5A not just at the pancreas level but overall. On the other hand, the T regulatory cell component of CD4 responded with activation and proliferation significantly higher than in the non-GC7-treated controls. Female mice seemed to be more susceptible to these effects. All together, our results show for the first time that downregulation of eIF5A through inhibition of DHS altered the physiopathology and observed immune outcome of diabetes in an animal model that closely resembles human T1D. Although the development of diabetes could not be abrogated by DHS inhibition, the immunomodulatory capacity of this approach may supplement other interventions directed at increasing regulation of autoreactive T cells in T1D.


Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 1/drug therapy , Enzyme Inhibitors/therapeutic use , Guanine/analogs & derivatives , Immunity, Innate/drug effects , Peptide Initiation Factors/antagonists & inhibitors , RNA-Binding Proteins/antagonists & inhibitors , Animals , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Type 1/immunology , Female , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/immunology , Guanine/therapeutic use , HLA-DQ alpha-Chains/genetics , HLA-DQ alpha-Chains/immunology , HLA-DQ beta-Chains/genetics , HLA-DQ beta-Chains/immunology , Humans , Immunity, Innate/genetics , Lysine/analogs & derivatives , Lysine/metabolism , Mice , Mice, Transgenic , Oxidoreductases Acting on CH-NH Group Donors/antagonists & inhibitors , Peptide Initiation Factors/metabolism , RNA-Binding Proteins/metabolism , Eukaryotic Translation Initiation Factor 5A
17.
Am J Reprod Immunol ; 71(3): 229-40, 2014 Mar.
Article En | MEDLINE | ID: mdl-24382123

PROBLEM: The role of eukaryotic initiation factor 5A (eIF5A) in feto-maternal immunotolerance is poorly understood. METHODS OF STUDY: The effects of N1-guanyl-1,7-diaminoheptane (GC7), an inhibitor of eIF5A, on the proportion and function of natural killer (NK) cell subsets were investigated using flow cytometry, immunofluorescence, CCK8 assay, TUNEL assay, DNA fragmentation analysis, mitochondrial membrane potential assay, and Western blotting. RESULTS: Inhibition of eIF5A by GC7 increased embryo loss and reduced the percentage of NK cells in the uterus and spleen. GC7 treatment caused inhibition of NK cell proliferation in a time- and dose-dependent manner. GC7 also induced apoptosis of NK cells. GC7 treatment increased the protein levels of FasL, bax, p53, and cleaved caspase-3. Moreover, GC7 caused loss of mitochondrial membrane potential in NK cells. CONCLUSION: Inhibition of eIF5A results in aberrant NK cell function and increased embryo loss.


Embryo Loss/immunology , Guanine/analogs & derivatives , Killer Cells, Natural/immunology , Peptide Initiation Factors/metabolism , RNA-Binding Proteins/metabolism , Uterus/immunology , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Guanine/administration & dosage , Guanine/pharmacology , Immune Tolerance , Killer Cells, Natural/drug effects , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Peptide Initiation Factors/antagonists & inhibitors , Peptide Initiation Factors/genetics , Pregnancy , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , Spleen/drug effects , Spleen/immunology , Uterus/drug effects , Eukaryotic Translation Initiation Factor 5A
18.
Cancer Sci ; 105(2): 219-27, 2014 Feb.
Article En | MEDLINE | ID: mdl-24262005

Drug resistance greatly reduces the efficacy of doxorubicin-based chemotherapy in bladder cancer treatment; however, the underlying mechanisms are poorly understood. We aimed to investigate whether N1-guanyl-1,7-diaminoheptane (GC7), which inhibits eukaryotic translation initiation factor 5A2 (eIF5A2) activation, exerts synergistic cytotoxicity with doxorubicin in bladder cancer, and whether eIF5A2 is involved in chemoresistance to doxorubicin-based bladder cancer treatment. BIU-87, J82, and UM-UC-3 bladder cancer cells were transfected with eIF5A2 siRNA or negative control siRNA before incubation with doxorubicin alone or doxorubicin plus GC7 for 48 h. Doxorubicin cytotoxicity was enhanced by GC7 in BIU-87, J82, and UM-UC-3 cells. It significantly inhibited activity of eIF5A2, suppressed doxorubicin-induced epithelial-mesenchymal transition in BIU-87 cells, and promoted mesenchymal-epithelial transition in J82 and UM-UC-3 cells. Knockdown of eIF5A2 sensitized bladder cancer cells to doxorubicin, prevented doxorubicin-induced EMT in BIU-87 cells, and encouraged mesenchymal-epithelial transition in J82 and UM-UC-3 cells. Combination therapy with GC7 may enhance the therapeutic efficacy of doxorubicin in bladder cancer by inhibiting eIF5A2 activation and preventing epithelial-mesenchymal transition.


Antineoplastic Combined Chemotherapy Protocols/pharmacology , Doxorubicin/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Guanine/analogs & derivatives , Peptide Initiation Factors/antagonists & inhibitors , RNA-Binding Proteins/antagonists & inhibitors , Urinary Bladder Neoplasms/drug therapy , Cell Survival/drug effects , Cell Survival/genetics , Doxorubicin/administration & dosage , Drug Synergism , Epithelial-Mesenchymal Transition/genetics , Guanine/administration & dosage , Guanine/pharmacology , Humans , Peptide Initiation Factors/genetics , RNA-Binding Proteins/genetics , Tumor Cells, Cultured , Urinary Bladder Neoplasms/genetics , Eukaryotic Translation Initiation Factor 5A
19.
Cancer Res ; 74(2): 552-62, 2014 Jan 15.
Article En | MEDLINE | ID: mdl-24220243

Cancer etiology is influenced by alterations in protein synthesis that are not fully understood. In this study, we took a novel approach to investigate the role of the eukaryotic translation initiation factor eIF5A in human cervical cancers, where it is widely overexpressed. eIF5A contains the distinctive amino acid hypusine, which is formed by a posttranslational modification event requiring deoxyhypusine hydroxylase (DOHH), an enzyme that can be inhibited by the drugs ciclopirox and deferiprone. We found that proliferation of cervical cancer cells can be blocked by DOHH inhibition with either of these pharmacologic agents, as well as by RNA interference-mediated silencing of eIF5A, DOHH, or another enzyme in the hypusine pathway. Proteomic and RNA analyses in HeLa cervical cancer cells identified two groups of proteins in addition to eIF5A that were coordinately affected by ciclopirox and deferiprone. Group 1 proteins (Hsp27, NM23, and DJ-1) were downregulated at the translational level, whereas group 2 proteins (TrpRS and PRDX2) were upregulated at the mRNA level. Further investigations confirmed that eIF5A and DOHH are required for Hsp27 expression in cervical cancer cells and for regulation of its key target IκB and hence NF-κB. Our results argue that mature eIF5A controls a translational network of cancer-driving genes, termed the eIF5A regulon, at the levels of mRNA abundance and translation. In coordinating cell proliferation, the eIF5A regulon can be modulated by drugs such as ciclopirox or deferiprone, which might be repositioned to control cancer cell growth.


Cell Proliferation , Gene Expression Regulation, Neoplastic , Peptide Initiation Factors/antagonists & inhibitors , Protein Processing, Post-Translational/drug effects , RNA-Binding Proteins/antagonists & inhibitors , Uterine Cervical Neoplasms/metabolism , Antifungal Agents/pharmacology , Ciclopirox , Deferiprone , Female , Gene Expression Regulation, Enzymologic , Gene Silencing , HeLa Cells , Humans , Iron Chelating Agents/pharmacology , Mixed Function Oxygenases/metabolism , NF-kappa B/metabolism , Proteomics/methods , Pyridones/pharmacology , RNA Interference , Eukaryotic Translation Initiation Factor 5A
20.
Exp Cell Res ; 319(17): 2708-17, 2013 Oct 15.
Article En | MEDLINE | ID: mdl-23958463

Hepatocellular carcinoma (HCC) cells undergo the epithelial-mesenchymal transition (EMT) during chemotherapy, which reduces the efficacy of doxorubicin-based chemotherapy. We investigated N1-guanyl-1,7-diaminoheptane (GC7) which inhibits eukaryotic translation initiation factor 5A2 (eIF5A2) activation; eIF5A2 is associated with chemoresistance. GC7 enhanced doxorubicin cytotoxicity in epithelial HCC cells (Huh7, Hep3B and HepG2) but had little effect in mesenchymal HCC cells (SNU387, SNU449). GC7 suppressed the doxorubicin-induced EMT in epithelial HCC cells; knockdown of eIF5A2 inhibited the doxorubicin-induced EMT and enhanced doxorubicin cytotoxicity. GC7 combination therapy may enhance the therapeutic efficacy of doxorubicin in HCC by inhibiting eIF5A2 activation and preventing the EMT.


Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Hepatocellular/drug therapy , Doxorubicin/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Guanine/analogs & derivatives , Liver Neoplasms/drug therapy , Peptide Initiation Factors/antagonists & inhibitors , RNA-Binding Proteins/antagonists & inhibitors , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Doxorubicin/therapeutic use , Guanine/pharmacology , Guanine/therapeutic use , Hep G2 Cells , Humans , Peptide Initiation Factors/genetics , Peptide Initiation Factors/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Eukaryotic Translation Initiation Factor 5A
...