Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Dis Model Mech ; 5(1): 63-72, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21903676

ABSTRACT

Because Ras signaling is frequently activated by major hepatocellular carcinoma etiological factors, a transgenic zebrafish constitutively expressing the kras(V12) oncogene in the liver was previously generated by our laboratory. Although this model depicted and uncovered the conservation between zebrafish and human liver tumorigenesis, the low tumor incidence and early mortality limit its use for further studies of tumor progression and inhibition. Here, we employed a mifepristone-inducible transgenic system to achieve inducible kras(V12) expression in the liver. The system consisted of two transgenic lines: the liver-driver line had a liver-specific fabp10 promoter to produce the LexPR chimeric transactivator, and the Ras-effector line contained a LexA-binding site to control EGFP-kras(V12) expression. In double-transgenic zebrafish (driver-effector) embryos and adults, we demonstrated mifepristone-inducible EGFP-kras(V12) expression in the liver. Robust and homogeneous liver tumors developed in 100% of double-transgenic fish after 1 month of induction and the tumors progressed from hyperplasia by 1 week post-treatment (wpt) to carcinoma by 4 wpt. Strikingly, liver tumorigenesis was found to be 'addicted' to Ras signaling for tumor maintenance, because mifepristone withdrawal led to tumor regression via cell death in transgenic fish. We further demonstrated the potential use of the transparent EGFP-kras(V12) larvae in inhibitor treatments to suppress Ras-driven liver tumorigenesis by targeting its downstream effectors, including the Raf-MEK-ERK and PI3K-AKT-mTOR pathways. Collectively, this mifepristone-inducible and reversible kras(V12) transgenic system offers a novel model for understanding hepatocarcinogenesis and a high-throughput screening platform for anti-cancer drugs.


Subject(s)
Cell Transformation, Neoplastic/pathology , Drug Screening Assays, Antitumor/methods , Genes, ras/genetics , Liver Neoplasms/pathology , Zebrafish/genetics , ras Proteins/genetics , Animals , Animals, Genetically Modified , Disease Models, Animal , Disease Progression , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Liver Neoplasms/enzymology , Mifepristone/pharmacology , Models, Biological , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects
2.
Dis Model Mech ; 4(6): 801-13, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21729876

ABSTRACT

Human liver cancer is one of the deadliest cancers worldwide, with hepatocellular carcinoma (HCC) being the most common type. Aberrant Ras signaling has been implicated in the development and progression of human HCC, but a complete understanding of the molecular mechanisms of this protein in hepatocarcinogenesis remains elusive. In this study, a stable in vivo liver cancer model using transgenic zebrafish was generated to elucidate Ras-driven tumorigenesis in HCC. Using the liver-specific fabp10 (fatty acid binding protein 10) promoter, we overexpressed oncogenic kras(V12) specifically in the transgenic zebrafish liver. Only a high level of kras(V12) expression initiated liver tumorigenesis, which progressed from hyperplasia to benign and malignant tumors with activation of the Ras-Raf-MEK-ERK and Wnt-ß-catenin pathways. Histological diagnosis of zebrafish tumors identified HCC as the main lesion. The tumors were invasive and transplantable, indicating malignancy of these HCC cells. Oncogenic kras(V12) was also found to trigger p53-dependent senescence as a tumor suppressive barrier in the pre-neoplastic stage. Microarray analysis of zebrafish liver hyperplasia and HCC uncovered the deregulation of several stage-specific and common biological processes and signaling pathways responsible for kras(V12)-driven liver tumorigenesis that recapitulated the molecular hallmarks of human liver cancer. Cross-species comparisons of cancer transcriptomes further defined a HCC-specific gene signature as well as a liver cancer progression gene signature that are evolutionarily conserved between human and zebrafish. Collectively, our study presents a comprehensive portrait of molecular mechanisms during progressive Ras-induced HCC. These observations indicate the validity of our transgenic zebrafish to model human liver cancer, and this model might act as a useful platform for drug screening and identifying new therapeutic targets.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Liver/metabolism , Oncogenes , Zebrafish/genetics , ras Proteins/genetics , Aging/pathology , Animals , Animals, Genetically Modified , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Disease Models, Animal , Disease Progression , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Green Fluorescent Proteins/metabolism , Humans , Liver Neoplasms/enzymology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , MAP Kinase Signaling System , Mitogen-Activated Protein Kinases/metabolism , Neoplasm Transplantation , Organ Specificity , Transcriptome/genetics , Tumor Suppressor Protein p53/metabolism , Wnt Signaling Pathway
3.
Neuropharmacology ; 53(5): 687-98, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17889908

ABSTRACT

Studies have shown that the lipid peroxidation by-product, 4-hydroxynonenal (HNE), is involved in many pathological events in several neurodegenerative diseases. A number of signaling pathways mediating HNE-induced cell death in the brain have been proposed. However, the exact mechanism remains unknown. In the present study, we have examined the effects of HNE on cultured primary cortical neurons and found that HNE treatment leads to cell death via apoptosis. Both the caspase and calpain proteolytic systems were activated. There were also increased levels of phospho-p53 and cell cycle-related proteins. Gene transcription was further studied using microarray analysis. Results showed that majority of the genes associated with cell cycle regulation, response to stress, and signal transduction were differentially expressed. The various categories of differentially-expressed genes suggested that there are other parallel pathways regulating HNE-induced neuronal apoptosis. Collectively, these might help to elucidate similar molecular mechanisms involved during cell death in neurodegenerative diseases.


Subject(s)
Aldehydes/pharmacology , Apoptosis/drug effects , Cerebral Cortex/cytology , Neurons/drug effects , Signal Transduction/physiology , Transcription, Genetic/physiology , Acetylcysteine/pharmacology , Animals , Calpain/metabolism , Caspases/metabolism , Cell Cycle Proteins/physiology , Cell Death/drug effects , Cell Death/physiology , Cell Survival/drug effects , Cells, Cultured , Cerebral Cortex/drug effects , Cytoskeleton/physiology , Free Radical Scavengers/pharmacology , Gene Expression Regulation/physiology , Genes, p53 , Mice , Microarray Analysis , Oxidative Stress/physiology , Signal Transduction/drug effects , Ubiquitin/physiology
4.
J Cell Physiol ; 211(1): 63-87, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17245753

ABSTRACT

Studies suggest that cholesterol imbalance in the brain might be related to the development of neurological disorders. U18666A is a well-known amphiphile which inhibits intracellular cholesterol transport in treated cells. We have previously shown that U18666A leads to apoptosis and cholesterol accumulation in primary cortical neurons, which is associated with activation of caspases and calpains, hyperphosphorylation of tau, and increased oxidative stress markers. However, the mechanisms involved in U18666A-mediated apoptosis remain unknown. In this report, we sought to gain an insight into the molecular processes contributing to the neuronal apoptosis induced by U18666A. The microarray approach was used in conjunction with proteomics techniques to identify specific proteins which may serve as signature biomarkers during U18666A treatment. Eleven differentially expressed proteins were correlated at the gene expression level in a time-dependent manner. These proteins have been shown to play a role in lipid metabolism and transport, responses to cell death, protein folding and trafficking, and regulation of transcription. The identification of these differentially expressed proteins might provide a clue to decipher the intracellular biochemical changes during U18666A-mediated neuronal apoptosis. Our results provide, for the first time, a combined microarray and proteomics analysis of neuronal apoptosis mediated by inhibition of intracellular cholesterol transport. This new insight may greatly facilitate the study of neurodegenerative diseases.


Subject(s)
Apoptosis , Cerebral Cortex/cytology , Cholesterol/metabolism , Neurons/cytology , Oligonucleotide Array Sequence Analysis , Proteomics , Androstenes/pharmacology , Animals , Apoptosis/drug effects , Biological Transport/drug effects , Cells, Cultured , Cerebral Cortex/drug effects , Cluster Analysis , Electrophoresis, Gel, Two-Dimensional , Gene Expression Profiling , Genomics , Mass Spectrometry , Mice , Neurons/drug effects , Proteins/chemistry , Proteins/genetics , Proteins/isolation & purification
5.
Cell Signal ; 18(11): 1844-53, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16797161

ABSTRACT

Neuronal cell death can occur by means of either necrosis or apoptosis. Both necrosis and apoptosis are generally believed to be distinct mechanisms of cell death with different characteristic features distinguished on the basis of their morphological and biochemical properties. The brain is the most cholesterol-rich organ in the body but not much is known about the mechanisms that regulate cholesterol homeostasis in the brain. Recently, several clinical and biochemical studies suggest that cholesterol imbalance in the brain may be a risk factor related to the development of neurological disorders such as Niemann-Pick disease type C (NPC) and Alzheimer's disease (AD). NPC is a fatal juvenile neurodegenerative disorder characterized by premature neuronal death and somatically altered cholesterol metabolism. The main biochemical manifestation in NPC is elevated intracellular accumulation of free cholesterol caused by a genetic deficit in cholesterol trafficking. The pharmacological agent, U18666A (3-beta-[2-(diethylamino)ethoxy]androst-5-en-17-one), is a well-known class-2 amphiphile which inhibits cholesterol transport. Cells treated with this agent accumulate intracellular cholesterol to massive levels, similar to that observed in cells from NPC patients. NPC and AD have some pathological similarities which may share a common underlying cause. AD is one of the most common types of dementia affecting the elderly. However, the molecular mechanisms of neurodegeneration in NPC and AD are largely unknown. This review provides a consolidation of work done using U18666A in the past half century and focuses on the implications of our research findings on the mechanism of U18666A-mediated neuronal apoptosis in primary cortical neurons, which may provide an insight to elucidate the mechanisms of neurodegenerative diseases, particularly NPC and AD, where apoptosis might occur through a similar mechanism.


Subject(s)
Alzheimer Disease/metabolism , Androstenes/pharmacology , Apoptosis , Neurons/drug effects , Niemann-Pick Disease, Type C/metabolism , Animals , Anticholesteremic Agents/pharmacology , Apoptosis/drug effects , Cerebral Cortex/pathology , Cholesterol/metabolism , Humans , Neurons/metabolism
6.
J Neurochem ; 98(4): 1278-89, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16771828

ABSTRACT

Findings that antioxidant treatment may be beneficial in Alzheimer's disease indicate that oxidative stress is an important factor in its pathogenesis. Studies have also suggested that cholesterol imbalance in the brain might be related to the development of neurological disorders. Previously, we have reported that U18666A, a cholesterol transport-inhibiting agent, leads to apoptosis and intracellular cholesterol accumulation in primary cortical neurons. In this study, we found that neuronal apoptosis mediated by U18666A is associated with oxidative stress in the treated cortical neurons. Cortical neurons treated with U18666A also showed decreased secretion and increased intraneuronal accumulation of beta-amyloid. The association of neuronal apoptosis with oxidative stress and Abeta accumulation may provide clues to the pathogenesis of Alzheimer's disease, as well as the role oxidative stress plays in other neurodegenerative diseases.


Subject(s)
Androstenes/pharmacology , Cerebral Cortex/cytology , Neurons/drug effects , Oxidative Stress/drug effects , Adenosine Triphosphate/metabolism , Amyloid beta-Peptides/physiology , Animals , Apoptosis/drug effects , Blotting, Western , Caspase 3 , Caspases/metabolism , Cells, Cultured , Cerebral Cortex/drug effects , Cholesterol/metabolism , Gas Chromatography-Mass Spectrometry , Glutathione/metabolism , Lipid Peroxidation/drug effects , Membrane Potentials/drug effects , Mice , Neurodegenerative Diseases/pathology , Proteasome Endopeptidase Complex , Tetrazolium Salts , Thiazoles
7.
Cell Signal ; 18(10): 1572-83, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16446076

ABSTRACT

Studies have suggested that cholesterol imbalance in the brain might be related to the development of neurological disorders such as Alzheimer's disease and Niemann-Pick disease type C. Previously, we have reported that U18666A, a cholesterol transport-inhibiting agent, leads to apoptosis and intracellular cholesterol accumulation in primary cortical neurons. In this study, we examined the effects of U18666A-mediated neuronal apoptosis, and found that chronic exposure to U18666A led to the activation of caspases and calpains and hyperphosphorylation of tau. Tau hyperphosphorylation is regulated by several kinases that phosphorylate specific sites of tau in vitro. Surprisingly, the kinase activity of cyclin-dependent kinase 5 decreased in U18666A-treated cortical neurons whereas its protein level remained unchanged. The amount of glycogen synthase kinase 3 and mitogen-activated protein kinases were found to decrease in their phosphorylated states by Western blot analysis. Gene transcription was further studied using microarray analysis. Genes encoding for kinases and phosphatases were differentially expressed with most up-regulated and some down-regulated in expression upon U18666A treatment. The activation of cysteine proteases and cholesterol accumulation with tauopathies may provide clues to the cellular mechanism of the inhibition of cholesterol transport-mediated cell death in neurodegenerative diseases.


Subject(s)
Androstenes/pharmacology , Apoptosis/drug effects , Calpain/metabolism , Caspases/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neurons/drug effects , Animals , Cells, Cultured , Cyclin-Dependent Kinase 5/metabolism , Cytoskeleton/drug effects , Enzyme Activation/drug effects , Gene Expression Regulation/drug effects , Glycogen Synthase Kinase 3/metabolism , Mice , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/enzymology , Phosphoprotein Phosphatases/metabolism , Phosphorylation/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , tau Proteins/metabolism
8.
Neurosci Lett ; 397(1-2): 69-73, 2006.
Article in English | MEDLINE | ID: mdl-16412576

ABSTRACT

Rotenone is an inhibitor of mitochondrial complex I that produces a model of Parkinson's disease (PD), where neurons undergo apoptosis by caspase-dependent and/or caspase-independent pathways. Inhibition of calpains has recently been shown to attenuate neuronal apoptosis. This study aims to establish for the first time, the time-point of calpain activation with respect to the caspase activation and the possibility of cell cycle re-entry in rotenone-mediated cell death. Immunoblot results revealed calpain activation occurred at 5, 10h prior to caspase-3 activation (at 15 h), suggesting calpain activation was an earlier cellular event compared to caspase activation in the rotenone-mediated apoptosis. In addition, an upregulation of phospho-p53 was observed at 21 h. However, no expression or upregulation of cell cycle regulatory proteins including cdc25a, cyclin-D1 and cyclin-D3 were observed, strongly suggesting that cell cycle re-entry did not occur. These findings provide new insights into the differential patterns of calpain and caspase activation that result from rotenone poisoning and which may be relevant to the therapeutic management of PD.


Subject(s)
Apoptosis/drug effects , Calpain/biosynthesis , Enzyme Induction/drug effects , Insecticides/pharmacology , Neurons/drug effects , Rotenone/pharmacology , Analysis of Variance , Animals , Blotting, Western/methods , Carrier Proteins/metabolism , Caspase 3 , Caspases/metabolism , Cell Survival/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Dose-Response Relationship, Drug , Embryo, Mammalian , Gene Expression Regulation/drug effects , Mice , Microfilament Proteins/metabolism , Neurons/cytology , Rats , Time Factors , Tumor Suppressor Protein p53/metabolism
9.
Biochem Biophys Res Commun ; 315(2): 408-17, 2004 Mar 05.
Article in English | MEDLINE | ID: mdl-14766223

ABSTRACT

Niemann-Pick disease type C (NPC) is a juvenile neurodegenerative disorder characterized by premature neuronal loss and altered cholesterol metabolism. Previous reports applying an 8-h exposure of U18666A, a cholesterol transport-inhibiting agent, demonstrated a dose-dependent reduction in beta-amyloid (Abeta) deposition and secretion in cortical neurons, with no significant cell injury. In the current study, we examined the chronic effect of 24-72h of U18666A treatment on primary cortical neurons and several cell lines. Our results showed caspase-3 activation and cellular injury in U18666A-treated cortical neurons but not in the cell lines, suggesting cell death by apoptosis only occurred in cortical neurons after chronic exposure to U18666A. We also demonstrated through filipin staining the accumulation of intracellular cholesterol in cortical neurons treated with U18666A, indicating the phenotypic mimic of NPC by U18666A. However, additions of 10 and 25microM pravastatin with 0.5microg/ml U18666A significantly attenuated toxicity. Taken together, these data showed for the first time that U18666A induces cell death by apoptosis and suggested an important in vitro model system to study NPC.


Subject(s)
Androstenes/pharmacology , Apoptosis , Cerebral Cortex/drug effects , Enzyme Inhibitors/pharmacology , Neurons/drug effects , Animals , Anticholesteremic Agents/pharmacology , Blotting, Western , CHO Cells , Caspase 3 , Caspases/metabolism , Cell Line , Cell Survival , Cells, Cultured , Cholesterol/metabolism , Cricetinae , Dose-Response Relationship, Drug , Enzyme Activation , Filipin/metabolism , Humans , Mice , Microscopy, Electron , NIH 3T3 Cells , Neurons/metabolism , Pravastatin/pharmacology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...