Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Acta Radiol ; 60(3): 315-326, 2019 Mar.
Article in English | MEDLINE | ID: mdl-29890843

ABSTRACT

BACKGROUND: Mouse models of human-malignant-melanoma (MM) are important tools to study tumor dynamics. The enhanced green fluorescent protein (EGFP) is widely used in molecular imaging approaches, together with optical scanners, and fluorescence imaging. PURPOSE: Currently, there are no data available as to whether other fluorescent proteins are more suitable. The goal of this preclinical study was to analyze two fluorescent proteins of the GFP superfamily under real-time in vivo conditions using fluorescence reflectance imaging (FRI). MATERIAL AND METHODS: The human melanoma cell line MeWo was stable transfected with one plasmid: pEGFP-C1 or pDsRed1-N1. We investigated two severe combined immunodeficiency (SCID)-mice groups: A (solid xenografts) and B (xenografts as metastases). After three weeks, the animals were weekly imaged by FRI. Afterwards the mice were euthanized and metastases were imaged in situ: to quantify the cutis-dependent reduction of emitted light, we compared signal intensities obtained by metastases in vivo with signal intensities obtained by in situ liver parenchyma preparations. RESULTS: More than 90% of cells were stable transfected. EGFP-/DsRed-xenograft tumors had identical growth kinetics. In vivo the emitted light by DsRed tumors/metastases was much brighter than by EGFP. DsRed metastases were earlier (3 vs. 5 weeks) and much more sensitive detectable than EGFP metastases. Cutis-dependent reduction of emitted light was greater in EGFP than in DsRed mice (tenfold). Autofluorescence of DsRed was lower than of EGFP. CONCLUSION: We established an in vivo xenograft mouse model (DsRed-MeWo) that is reliable, reproducible, and superior to the EGFP model as a preclinical tool to study innovative therapies by FRI under real-time in vivo conditions.


Subject(s)
Green Fluorescent Proteins/pharmacokinetics , Melanoma/diagnostic imaging , Animals , Cell Line, Tumor , Disease Models, Animal , Heterografts , Humans , Luminescent Proteins/pharmacokinetics , Male , Mice , Mice, SCID , Microscopy, Fluorescence , Random Allocation , Transfection , Tumor Burden
2.
PLoS Genet ; 13(5): e1006785, 2017 May.
Article in English | MEDLINE | ID: mdl-28520736

ABSTRACT

Notch (N) signaling is central to the self-renewal of neural stem cells (NSCs) and other tissue stem cells. Its deregulation compromises tissue homeostasis and contributes to tumorigenesis and other diseases. How N regulates stem cell behavior in health and disease is not well understood. Here we show that N regulates bantam (ban) microRNA to impact cell growth, a process key to NSC maintenance and particularly relied upon by tumor-forming cancer stem cells. Notch signaling directly regulates ban expression at the transcriptional level, and ban in turn feedback regulates N activity through negative regulation of the Notch inhibitor Numb. This feedback regulatory mechanism helps maintain the robustness of N signaling activity and NSC fate. Moreover, we show that a Numb-Myc axis mediates the effects of ban on nucleolar and cellular growth independently or downstream of N. Our results highlight intricate transcriptional as well as translational control mechanisms and feedback regulation in the N signaling network, with important implications for NSC biology and cancer biology.


Subject(s)
Brain/metabolism , Drosophila Proteins/genetics , Drosophila/genetics , Juvenile Hormones/genetics , MicroRNAs/genetics , Neoplastic Stem Cells/metabolism , Receptors, Notch/metabolism , Animals , Brain/cytology , Cell Differentiation , Cell Growth Processes , Drosophila/cytology , Drosophila/metabolism , Drosophila Proteins/metabolism , Feedback, Physiological , Juvenile Hormones/metabolism , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/physiology , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neural Stem Cells/physiology , Receptors, Notch/genetics , Signal Transduction
3.
Cell Metab ; 21(1): 95-108, 2015 Jan 06.
Article in English | MEDLINE | ID: mdl-25565208

ABSTRACT

Mitochondria play essential roles in many aspects of biology, and their dysfunction has been linked to diverse diseases. Central to mitochondrial function is oxidative phosphorylation (OXPHOS), accomplished by respiratory chain complexes (RCCs) encoded by nuclear and mitochondrial genomes. How RCC biogenesis is regulated in metazoans is poorly understood. Here we show that Parkinson's disease (PD)-associated genes PINK1 and Parkin direct localized translation of certain nuclear-encoded RCC (nRCC) mRNAs. Translationally repressed nRCC mRNAs are localized in a PINK1/Tom20-dependent manner to mitochondrial outer membrane, where they are derepressed and activated by PINK1/Parkin through displacement of translation repressors, including Pumilio and Glorund/hnRNP-F, a Parkin substrate, and enhanced binding of activators such as eIF4G. Inhibiting the translation repressors rescued nRCC mRNA translation and neuromuscular-degeneration phenotypes of PINK1 mutant, whereas inhibiting eIF4G had opposite effects. Our results reveal previously unknown functions of PINK1/Parkin in RNA metabolism and suggest new approaches to mitochondrial restoration and disease intervention.


Subject(s)
Drosophila Proteins/metabolism , Electron Transport Chain Complex Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Protein Serine-Threonine Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/genetics , Drosophila melanogaster/metabolism , Electron Transport Chain Complex Proteins/genetics , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , HEK293 Cells , Heterogeneous-Nuclear Ribonucleoprotein Group F-H/metabolism , Humans , Lipid Peroxidation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Biosynthesis , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering/metabolism , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Rats , Rats, Sprague-Dawley , Ubiquitin-Protein Ligases/genetics , Ubiquitination
4.
Brain Res ; 1584: 105-15, 2014 Oct 10.
Article in English | MEDLINE | ID: mdl-24631951

ABSTRACT

Neurodegenerative diseases such as Parkinson׳s disease are progressive disorders of the nervous system that affect the function and maintenance of specific neuronal populations. While most disease cases are sporadic with no known cause, a small percentage of disease cases are caused by inherited genetic mutations. The identification of genes associated with the familial forms of the diseases and subsequent studies of proteins encoded by the disease genes in cellular or animal models have offered much-needed insights into the molecular and cellular mechanisms underlying disease pathogenesis. Recent studies of the familial Parkinson׳s disease genes have emphasized the importance of RNA metabolism, particularly mRNA translation, in the disease process. It is anticipated that continued studies on the role of RNA metabolism in Parkinson׳s disease will offer unifying mechanisms for understanding the cause of neuronal dysfunction and degeneration and facilitate the development of novel and rational strategies for treating this debilitating disease.


Subject(s)
Parkinson Disease/genetics , Parkinson Disease/metabolism , RNA/genetics , RNA/metabolism , Animals , Eukaryotic Initiation Factor-4G/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Mutation , Oncogene Proteins/genetics , Protein Biosynthesis , Protein Deglycase DJ-1 , Protein Kinases/genetics , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Ubiquitin-Protein Ligases/genetics , alpha-Synuclein/genetics
5.
Biochem Soc Trans ; 40(5): 1047-51, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22988863

ABSTRACT

Mutations in LRRK2 (leucine-rich repeat kinase 2) are the most frequent genetic lesions so far found in familial as well as sporadic forms of PD (Parkinson's disease), a neurodegenerative disease characterized by the dysfunction and degeneration of dopaminergic and other neuronal types. The molecular and cellular mechanisms underlying LRRK2 action remain poorly defined. Synaptic dysfunction has been increasingly recognized as an early event in the pathogenesis of major neurological disorders. Using Drosophila as a model system, we have shown that LRRK2 controls synaptic morphogenesis. Loss of dLRRK (Drosophila LRRK2) results in synaptic overgrowth at the Drosophila neuromuscular junction synapse, whereas overexpression of wild-type dLRRK, hLRRK2 (human LRRK2) or the pathogenic hLRRK2-G2019S mutant has the opposite effect. Alteration of LRRK2 activity also affects synaptic transmission in a complex manner. LRRK2 exerts its effects on synaptic morphology by interacting with distinct downstream effectors at the pre- and post-synaptic compartments. At the postsynapse, LRRK2 functionally interacts with 4E-BP (eukaryotic initiation factor 4E-binding protein) and the microRNA machinery, both of which negatively regulate protein synthesis. At the presynapse, LRRK2 phosphorylates and negatively regulates the microtubule-binding protein Futsch and functionally interacts with the mitochondrial transport machinery. These results implicate compartment-specific synaptic dysfunction caused by altered protein synthesis, cytoskeletal dynamics and mitochondrial transport in LRRK2 pathogenesis and offer a new paradigm for understanding and ultimately treating LRRK2-related PD.


Subject(s)
Drosophila Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Drosophila Proteins/deficiency , Drosophila Proteins/genetics , Drosophila melanogaster/metabolism , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Mutation , Parkinson Disease/genetics , Parkinson Disease/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics
6.
Nature ; 466(7306): 637-41, 2010 Jul 29.
Article in English | MEDLINE | ID: mdl-20671708

ABSTRACT

Gain-of-function mutations in leucine-rich repeat kinase 2 (LRRK2) cause familial as well as sporadic Parkinson's disease characterized by age-dependent degeneration of dopaminergic neurons. The molecular mechanism of LRRK2 action is not known. Here we show that LRRK2 interacts with the microRNA (miRNA) pathway to regulate protein synthesis. Drosophila e2f1 and dp messenger RNAs are translationally repressed by let-7 and miR-184*, respectively. Pathogenic LRRK2 antagonizes these miRNAs, leading to the overproduction of E2F1/DP, previously implicated in cell cycle and survival control and shown here to be critical for LRRK2 pathogenesis. Genetic deletion of let-7, antagomir-mediated blockage of let-7 and miR-184* action, transgenic expression of dp target protector, or replacement of endogenous dp with a dp transgene non-responsive to let-7 each had toxic effects similar to those of pathogenic LRRK2. Conversely, increasing the level of let-7 or miR-184* attenuated pathogenic LRRK2 effects. LRRK2 associated with Drosophila Argonaute-1 (dAgo1) or human Argonaute-2 (hAgo2) of the RNA-induced silencing complex (RISC). In aged fly brain, dAgo1 protein level was negatively regulated by LRRK2. Further, pathogenic LRRK2 promoted the association of phospho-4E-BP1 with hAgo2. Our results implicate deregulated synthesis of E2F1/DP caused by the miRNA pathway impairment as a key event in LRRK2 pathogenesis and suggest novel miRNA-based therapeutic strategies.


Subject(s)
Down-Regulation , MicroRNAs/genetics , MicroRNAs/metabolism , Protein Biosynthesis , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Animals , Argonaute Proteins , Cell Line , Dopamine/metabolism , Drosophila Proteins/biosynthesis , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster , E2F1 Transcription Factor/biosynthesis , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , Eukaryotic Initiation Factor-2/metabolism , Eukaryotic Initiation Factors/biosynthesis , Eukaryotic Initiation Factors/metabolism , Female , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Male , MicroRNAs/antagonists & inhibitors , Neurons/cytology , Neurons/metabolism , Parkinson Disease/etiology , Parkinson Disease/genetics , Parkinson Disease/metabolism , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Induced Silencing Complex/antagonists & inhibitors , RNA-Induced Silencing Complex/chemistry , RNA-Induced Silencing Complex/metabolism , Trans-Activators/biosynthesis , Trans-Activators/genetics , Trans-Activators/metabolism , Up-Regulation
7.
BMC Neurosci ; 10: 109, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19723328

ABSTRACT

BACKGROUND: Parkinson's disease (PD) is the most common movement disorder. Extrapyramidal motor symptoms stem from the degeneration of the dopaminergic pathways in patient brain. Current treatments for PD are symptomatic, alleviating disease symptoms without reversing or retarding disease progression. Although the cause of PD remains unknown, several pathogenic factors have been identified, which cause dopaminergic neuron (DN) death in the substantia nigra (SN). These include oxidative stress, mitochondrial dysfunction, inflammation and excitotoxicity. Manipulation of these factors may allow the development of disease-modifying treatment strategies to slow neuronal death. Inhibition of DJ-1A, the Drosophila homologue of the familial PD gene DJ-1, leads to oxidative stress, mitochondrial dysfunction, and DN loss, making fly DJ-1A model an excellent in vivo system to test for compounds with therapeutic potential. RESULTS: In the present study, a Drosophila DJ-1A model of PD was used to test potential neuroprotective drugs. The drugs applied are the Chinese herb celastrol, the antibiotic minocycline, the bioenergetic amine coenzyme Q10 (coQ10), and the glutamate antagonist 2,3-dihydroxy-6-nitro-7-sulphamoylbenzo[f]-quinoxaline (NBQX). All of these drugs target pathogenic processes implicated in PD, thus constitute mechanism-based treatment strategies. We show that celastrol and minocycline, both having antioxidant and anti-inflammatory properties, confer potent dopaminergic neuroprotection in Drosophila DJ-1A model, while coQ10 shows no protective effect. NBQX exerts differential effects on cell survival and brain dopamine content: it protects against DN loss but fails to restore brain dopamine level. CONCLUSION: The present study further validates Drosophila as a valuable model for preclinical testing of drugs with therapeutic potential for neurodegenerative diseases. The lower cost and amenability to high throughput testing make Drosophila PD models effective in vivo tools for screening novel therapeutic compounds. If our findings can be further validated in mammalian PD models, they would implicate drugs combining antioxidant and anti-inflammatory properties as strong therapeutic candidates for mechanism-based PD treatment.


Subject(s)
Dopamine/metabolism , Neurons/metabolism , Neuroprotective Agents/therapeutic use , Parkinsonian Disorders/drug therapy , Age Factors , Animals , Animals, Genetically Modified , Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Brain/metabolism , Cell Count , Cell Survival , Chromatography, High Pressure Liquid , Drosophila , Drug Evaluation, Preclinical , Excitatory Amino Acid Antagonists/therapeutic use , Immunohistochemistry , Oxidative Stress , Parkinsonian Disorders/genetics , Parkinsonian Disorders/metabolism , Pentacyclic Triterpenes , Quinoxalines/therapeutic use , Triterpenes/therapeutic use , Ubiquinone/analogs & derivatives , Ubiquinone/therapeutic use
8.
Hum Mol Genet ; 18(22): 4390-404, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19692353

ABSTRACT

Mutations in the LRRK2 gene are the most common genetic cause of familial Parkinson's disease (PD). However, its physiological and pathological functions are unknown. Therefore, we generated several independent Drosophila lines carrying WT or mutant human LRRK2 (mutations in kinase, COR or LRR domains, resp.). Ectopic expression of WT or mutant LRRK2 in dopaminergic neurons caused their significant loss accompanied by complex age-dependent changes in locomotor activity. Overall, the ubiquitous expression of LRRK2 increased lifespan and fertility of the flies. However, these flies were more sensitive to rotenone. LRRK2 expression in the eye exacerbated retinal degeneration. Importantly, in double transgenic flies, various indices of the eye and dopaminergic survival were modified in a complex fashion by a concomitant expression of PINK1, DJ-1 or Parkin. This evidence suggests a genetic interaction between these PD-relevant genes.


Subject(s)
Drosophila melanogaster/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Oncogene Proteins/metabolism , Parkinson Disease/metabolism , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Animals, Genetically Modified , Disease Models, Animal , Drosophila melanogaster/metabolism , Female , Humans , Intracellular Signaling Peptides and Proteins/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Life Expectancy , Male , Oncogene Proteins/genetics , Parkinson Disease/genetics , Parkinson Disease/physiopathology , Protein Binding , Protein Deglycase DJ-1 , Protein Kinases/genetics , Protein Serine-Threonine Kinases/genetics , Ubiquitin-Protein Ligases/genetics
9.
EMBO J ; 27(18): 2432-43, 2008 Sep 17.
Article in English | MEDLINE | ID: mdl-18701920

ABSTRACT

Dominant mutations in leucine-rich repeat kinase 2 (LRRK2) are the most frequent molecular lesions so far found in Parkinson's disease (PD), an age-dependent neurodegenerative disorder affecting dopaminergic (DA) neuron. The molecular mechanisms by which mutations in LRRK2 cause DA degeneration in PD are not understood. Here, we show that both human LRRK2 and the Drosophila orthologue of LRRK2 phosphorylate eukaryotic initiation factor 4E (eIF4E)-binding protein (4E-BP), a negative regulator of eIF4E-mediated protein translation and a key mediator of various stress responses. Although modulation of the eIF4E/4E-BP pathway by LRRK2 stimulates eIF4E-mediated protein translation both in vivo and in vitro, it attenuates resistance to oxidative stress and survival of DA neuron in Drosophila. Our results suggest that chronic inactivation of 4E-BP by LRRK2 with pathogenic mutations deregulates protein translation, eventually resulting in age-dependent loss of DA neurons.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Dopamine/metabolism , Drosophila Proteins/physiology , Intracellular Signaling Peptides and Proteins/physiology , Neurons/metabolism , Peptide Initiation Factors/physiology , Phosphoproteins/physiology , Protein Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins , Drosophila Proteins/metabolism , Drosophila melanogaster , Eukaryotic Initiation Factor-4E/metabolism , Genes, Dominant , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Models, Biological , Mutation , Oxidative Stress , Peptide Initiation Factors/metabolism , Phosphoproteins/metabolism , Phosphorylation , Protein Biosynthesis
10.
Proc Natl Acad Sci U S A ; 103(28): 10793-8, 2006 Jul 11.
Article in English | MEDLINE | ID: mdl-16818890

ABSTRACT

Mutations in Pink1, a gene encoding a Ser/Thr kinase with a mitochondrial-targeting signal, are associated with Parkinson's disease (PD), the most common movement disorder characterized by selective loss of dopaminergic neurons. The mechanism by which loss of Pink1 leads to neurodegeneration is not understood. Here we show that inhibition of Drosophila Pink1 (dPink1) function results in energy depletion, shortened lifespan, and degeneration of select indirect flight muscles and dopaminergic neurons. The muscle pathology was preceded by mitochondrial enlargement and disintegration. These phenotypes could be rescued by the wild type but not the pathogenic C-terminal deleted form of human Pink1 (hPink1). The muscle and dopaminergic phenotypes associated with dPink1 inactivation show similarity to that seen in parkin mutant flies and could be suppressed by the overexpression of Parkin but not DJ-1. Consistent with the genetic rescue results, we find that, in dPink1 RNA interference (RNAi) animals, the level of Parkin protein is significantly reduced. Together, these results implicate Pink1 and Parkin in a common pathway that regulates mitochondrial physiology and cell survival in Drosophila.


Subject(s)
Drosophila Proteins/physiology , Gene Silencing , Mitochondria/pathology , Muscles/pathology , Neurons/pathology , Protein Kinases/genetics , Animals , Animals, Genetically Modified , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Humans , Muscles/physiopathology , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Parkinson Disease , Protein Kinases/deficiency , RNA Interference , Signal Transduction/genetics , Ubiquitin-Protein Ligases
11.
Proc Natl Acad Sci U S A ; 102(38): 13670-5, 2005 Sep 20.
Article in English | MEDLINE | ID: mdl-16155123

ABSTRACT

Parkinson's disease (PD) is the most common movement disorder characterized by dopaminergic dysfunction and degeneration. The cause of most PD cases is unknown, although postmortem studies have implicated the involvement of oxidative stress. The identification of familial PD-associated genes offers the opportunity to study mechanisms of PD pathogenesis in model organisms. Here, we show that DJ-1A, a Drosophila homologue of the familial PD-associated gene DJ-1, plays an essential role in oxidative stress response and neuronal maintenance. Inhibition of DJ-1A function through RNA interference (RNAi) results in cellular accumulation of reactive oxygen species, organismal hypersensitivity to oxidative stress, and dysfunction and degeneration of dopaminergic and photoreceptor neurons. To identify other genes that may interact with DJ-1A in regulating cell survival, we performed genetic interaction studies and identified components of the phosphatidylinositol 3-kinase (PI3K)/Akt-signaling pathway as specific modulators of DJ-1A RNAi-induced neurodegeneration. PI3K signaling suppresses DJ-1A RNAi phenotypes at least in part by reducing cellular reactive oxygen species levels. Consistent with the genetic interaction results, we also found reduced phosphorylation of Akt in DJ-1A RNAi animals, indicating an impairment of PI3K/Akt signaling by DJ-1A down-regulation. Together with recent findings in mammalian systems, these results implicate impairments of PI3K/Akt signaling and oxidative stress response in DJ-1-associated disease pathogenesis. We also observed impairment of PI3K/Akt signaling in the fly parkin model of PD, hinting at a common molecular event in the pathogenesis of PD. Manipulation of PI3K/Akt signaling may therefore offer therapeutic benefits for the treatment of PD.


Subject(s)
Drosophila Proteins/metabolism , Oxidative Stress/physiology , Phosphatidylinositol 3-Kinases/metabolism , Photoreceptor Cells, Invertebrate/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction/physiology , Animals , Disease Models, Animal , Dopamine/metabolism , Drosophila , Drosophila Proteins/genetics , Enzyme Activation/genetics , Humans , Oxidative Stress/genetics , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology , Proto-Oncogene Proteins c-akt , RNA Interference/physiology , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Signal Transduction/genetics
12.
Gene ; 322: 137-43, 2003 Dec 11.
Article in English | MEDLINE | ID: mdl-14644505

ABSTRACT

Hepatocyte-directed delivery of therapeutic genes is a major field of gene therapy. An important issue in this context is the availability of promoters units providing for maximum transcriptional activity and specificity. Although a number of liver-specific promoters and transcriptional control elements have been identified and used for gene delivery, no systematic study has been performed to identify the best suitable combination of known liver-specific promoter and enhancer elements. We now report the results of a comparative investigation addressing this issue. We tested a total of 25 synthetic transcriptional control units consisting of either of the four core promoters from liver-specific genes linked in various combinations and configurations to hepatocyte-specific enhancer elements. These constructs were analyzed for transcriptional activity in different cell types in cell culture and in mouse liver in vivo. The data lead to the clear conclusion that a combination of the alcohol dehydrogenase 6 (ADH6) basal promoter linked to two tandem copies of an apoplipoprotein E enhancer element is the transcriptional control unit of choice for the liver-specific expression of transgenes.


Subject(s)
Gene Expression Regulation , Liver/metabolism , Recombinant Fusion Proteins/metabolism , Transgenes/genetics , Alcohol Dehydrogenase/genetics , Animals , Cell Line, Tumor , Cholesterol 7-alpha-Hydroxylase/genetics , Enhancer Elements, Genetic/genetics , Factor IX/genetics , Humans , Injections, Intravenous , Luciferases/genetics , Luciferases/metabolism , Male , Mice , Mice, Inbred Strains , Mice, Nude , Plasmids/administration & dosage , Plasmids/genetics , Promoter Regions, Genetic/genetics , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Transcription, Genetic/genetics , Transfection , alpha 1-Antitrypsin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...