Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 15 de 15
1.
Biol Direct ; 18(1): 43, 2023 08 01.
Article En | MEDLINE | ID: mdl-37528429

Antipsychotic drugs are the mainstay of treatment for schizophrenia and provide adjunct therapies for other prevalent psychiatric conditions, including bipolar disorder and major depressive disorder. However, they also induce debilitating extrapyramidal syndromes (EPS), such as Parkinsonism, in a significant minority of patients. The majority of antipsychotic drugs function as dopamine receptor antagonists in the brain while the most recent 'third'-generation, such as aripiprazole, act as partial agonists. Despite showing good clinical efficacy, these newer agents are still associated with EPS in ~ 5 to 15% of patients. However, it is not fully understood how these movement disorders develop. Here, we combine clinically-relevant drug concentrations with mutliscale model systems to show that aripiprazole and its primary active metabolite induce mitochondrial toxicity inducing robust declines in cellular ATP and viability. Aripiprazole, brexpiprazole and cariprazine were shown to directly inhibit respiratory complex I through its ubiquinone-binding channel. Importantly, all three drugs induced mitochondrial toxicity in primary embryonic mouse neurons, with greater bioenergetic inhibition in ventral midbrain neurons than forebrain neurons. Finally, chronic feeding with aripiprazole resulted in structural damage to mitochondria in the brain and thoracic muscle of adult Drosophila melanogaster consistent with locomotor dysfunction. Taken together, we show that antipsychotic drugs acting as partial dopamine receptor agonists exhibit off-target mitochondrial liabilities targeting complex I.


Antipsychotic Agents , Depressive Disorder, Major , Animals , Mice , Aripiprazole/pharmacology , Aripiprazole/therapeutic use , Antipsychotic Agents/pharmacology , Antipsychotic Agents/therapeutic use , Depressive Disorder, Major/drug therapy , Drosophila melanogaster , Electron Transport
2.
Cell Metab ; 35(10): 1799-1813.e7, 2023 10 03.
Article En | MEDLINE | ID: mdl-37633273

The mammalian respiratory chain complexes I, III2, and IV (CI, CIII2, and CIV) are critical for cellular bioenergetics and form a stable assembly, the respirasome (CI-CIII2-CIV), that is biochemically and structurally well documented. The role of the respirasome in bioenergetics and the regulation of metabolism is subject to intense debate and is difficult to study because the individual respiratory chain complexes coexist together with high levels of respirasomes. To critically investigate the in vivo role of the respirasome, we generated homozygous knockin mice that have normal levels of respiratory chain complexes but profoundly decreased levels of respirasomes. Surprisingly, the mutant mice are healthy, with preserved respiratory chain capacity and normal exercise performance. Our findings show that high levels of respirasomes are dispensable for maintaining bioenergetics and physiology in mice but raise questions about their alternate functions, such as those relating to the regulation of protein stability and prevention of age-associated protein aggregation.


Mitochondria , Mitochondrial Membranes , Animals , Mice , Electron Transport , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Electron Transport Complex IV/metabolism , Energy Metabolism , Mammals/metabolism
3.
Sci Adv ; 9(31): eadi1359, 2023 08 02.
Article En | MEDLINE | ID: mdl-37531432

Respiratory complex I, a key enzyme in mammalian metabolism, captures the energy released by reduction of ubiquinone by NADH to drive protons across the inner mitochondrial membrane, generating the proton-motive force for ATP synthesis. Despite remarkable advances in structural knowledge of this complicated membrane-bound enzyme, its mechanism of catalysis remains controversial. In particular, how ubiquinone reduction is coupled to proton pumping and the pathways and mechanisms of proton translocation are contested. We present a 2.4-Å resolution cryo-EM structure of complex I from mouse heart mitochondria in the closed, active (ready-to-go) resting state, with 2945 water molecules modeled. By analyzing the networks of charged and polar residues and water molecules present, we evaluate candidate pathways for proton transfer through the enzyme, for the chemical protons for ubiquinone reduction, and for the protons transported across the membrane. Last, we compare our data to the predictions of extant mechanistic models, and identify key questions to answer in future work to test them.


Electron Transport Complex I , Protons , Mice , Animals , Oxidation-Reduction , Electron Transport Complex I/metabolism , Ubiquinone/chemistry , Ubiquinone/metabolism , Cryoelectron Microscopy , Mammals/metabolism , Water/metabolism
4.
Elife ; 122023 01 09.
Article En | MEDLINE | ID: mdl-36622099

Respiratory complex I powers ATP synthesis by oxidative phosphorylation, exploiting the energy from NADH oxidation by ubiquinone to drive protons across an energy-transducing membrane. Drosophila melanogaster is a candidate model organism for complex I due to its high evolutionary conservation with the mammalian enzyme, well-developed genetic toolkit, and complex physiology for studies in specific cell types and tissues. Here, we isolate complex I from Drosophila and determine its structure, revealing a 43-subunit assembly with high structural homology to its 45-subunit mammalian counterpart, including a hitherto unknown homologue to subunit NDUFA3. The major conformational state of the Drosophila enzyme is the mammalian-type 'ready-to-go' active resting state, with a fully ordered and enclosed ubiquinone-binding site, but a subtly altered global conformation related to changes in subunit ND6. The mammalian-type 'deactive' pronounced resting state is not observed: in two minor states, the ubiquinone-binding site is unchanged, but a deactive-type π-bulge is present in ND6-TMH3. Our detailed structural knowledge of Drosophila complex I provides a foundation for new approaches to disentangle mechanisms of complex I catalysis and regulation in bioenergetics and physiology.


Drosophila melanogaster , Electron Transport Complex I , Animals , Cryoelectron Microscopy , Drosophila melanogaster/metabolism , Electron Transport Complex I/metabolism , Electron Transport Complex I/ultrastructure , Mitochondria/metabolism , Ubiquinone/metabolism
5.
Science ; 379(6630): 351-357, 2023 01 27.
Article En | MEDLINE | ID: mdl-36701435

The molecular mode of action of biguanides, including the drug metformin, which is widely used in the treatment of diabetes, is incompletely characterized. Here, we define the inhibitory drug-target interaction(s) of a model biguanide with mammalian respiratory complex I by combining cryo-electron microscopy and enzyme kinetics. We interpret these data to explain the selectivity of biguanide binding to different enzyme states. The primary inhibitory site is in an amphipathic region of the quinone-binding channel, and an additional binding site is in a pocket on the intermembrane-space side of the enzyme. An independent local chaotropic interaction, not previously described for any drug, displaces a portion of a key helix in the membrane domain. Our data provide a structural basis for biguanide action and enable the rational design of medicinal biguanides.


Biguanides , Electron Transport Complex I , Animals , Cryoelectron Microscopy , Electron Transport Complex I/antagonists & inhibitors , Metformin/pharmacology , Mitochondria/metabolism , Biguanides/pharmacology
6.
Curr Opin Struct Biol ; 77: 102447, 2022 Dec.
Article En | MEDLINE | ID: mdl-36087446

Respiratory complex I (NADH:ubiquinone oxidoreductase) is a multi-subunit, energy-transducing mitochondrial enzyme that is essential for oxidative phosphorylation and regulating NAD+/NADH pools. Despite recent advances in structural knowledge and a long history of biochemical analyses, the mechanism of redox-coupled proton translocation by complex I remains unknown. Due to its ability to separate molecules in a mixed population into distinct classes, single-particle electron cryomicroscopy has enabled identification and characterisation of different complex I conformations. However, deciding on their catalytic and/or regulatory properties to underpin mechanistic hypotheses, especially without detailed biochemical characterisation of the structural samples, has proven challenging. In this review we explore different mechanistic interpretations of the closed and open states identified in cryoEM analyses of mammalian complex I.


Electron Transport Complex I , NAD , Animals , Electron Transport Complex I/chemistry , Cryoelectron Microscopy , NAD/chemistry , NAD/metabolism , Ubiquinone/chemistry , Ubiquinone/metabolism , Oxidation-Reduction , Mammals/metabolism
7.
Nat Commun ; 13(1): 2758, 2022 05 19.
Article En | MEDLINE | ID: mdl-35589726

Mitochondrial complex I is a central metabolic enzyme that uses the reducing potential of NADH to reduce ubiquinone-10 (Q10) and drive four protons across the inner mitochondrial membrane, powering oxidative phosphorylation. Although many complex I structures are now available, the mechanisms of Q10 reduction and energy transduction remain controversial. Here, we reconstitute mammalian complex I into phospholipid nanodiscs with exogenous Q10. Using cryo-EM, we reveal a Q10 molecule occupying the full length of the Q-binding site in the 'active' (ready-to-go) resting state together with a matching substrate-free structure, and apply molecular dynamics simulations to propose how the charge states of key residues influence the Q10 binding pose. By comparing ligand-bound and ligand-free forms of the 'deactive' resting state (that require reactivating to catalyse), we begin to define how substrate binding restructures the deactive Q-binding site, providing insights into its physiological and mechanistic relevance.


Electron Transport Complex I , Ubiquinone , Animals , Binding Sites , Cryoelectron Microscopy , Electron Transport Complex I/metabolism , Mammals/metabolism , Mitochondrial Membranes/metabolism , Oxidation-Reduction , Ubiquinone/metabolism
8.
J Am Chem Soc ; 144(15): 6791-6801, 2022 04 20.
Article En | MEDLINE | ID: mdl-35380814

Respiratory complex I is an essential metabolic enzyme that uses the energy from NADH oxidation and ubiquinone reduction to translocate protons across an energy transducing membrane and generate the proton motive force for ATP synthesis. Under specific conditions, complex I can also catalyze the reverse reaction, Δp-linked oxidation of ubiquinol to reduce NAD+ (or O2), known as reverse electron transfer (RET). Oxidative damage by reactive oxygen species generated during RET underpins ischemia reperfusion injury, but as RET relies on several converging metabolic pathways, little is known about its mechanism or regulation. Here, we demonstrate Δp-linked RET through complex I in a synthetic proteoliposome system for the first time, enabling complete kinetic characterization of RET catalysis. We further establish the capability of our system by showing how RET in the mammalian enzyme is regulated by the active-deactive transition and by evaluating RET by complex I from several species in which direct assessment has not been otherwise possible. We thus provide new insights into the reversibility of complex I catalysis, an important but little understood mechanistic and physiological feature.


Electron Transport Complex I , Electrons , Animals , Catalysis , Electron Transport , Electron Transport Complex I/metabolism , Mammals/metabolism , NAD/metabolism , Oxidation-Reduction
9.
Sci Adv ; 7(20)2021 05.
Article En | MEDLINE | ID: mdl-33990335

Mitochondrial complex I (NADH:ubiquinone oxidoreductase), a major contributor of free energy for oxidative phosphorylation, is increasingly recognized as a promising drug target for ischemia-reperfusion injury, metabolic disorders, and various cancers. Several pharmacologically relevant but structurally unrelated small molecules have been identified as specific complex I inhibitors, but their modes of action remain unclear. Here, we present a 3.0-Å resolution cryo-electron microscopy structure of mammalian complex I inhibited by a derivative of IACS-010759, which is currently in clinical development against cancers reliant on oxidative phosphorylation, revealing its unique cork-in-bottle mechanism of inhibition. We combine structural and kinetic analyses to deconvolute cross-species differences in inhibition and identify the structural motif of a "chain" of aromatic rings as a characteristic that promotes inhibition. Our findings provide insights into the importance of π-stacking residues for inhibitor binding in the long substrate-binding channel in complex I and a guide for future biorational drug design.

10.
Elife ; 92020 05 20.
Article En | MEDLINE | ID: mdl-32432547

Disruption of mitochondrial function selectively targets tumour cells that are dependent on oxidative phosphorylation. However, due to their high energy demands, cardiac cells are disproportionately targeted by mitochondrial toxins resulting in a loss of cardiac function. An analysis of the effects of mubritinib on cardiac cells showed that this drug did not inhibit HER2 as reported, but directly inhibits mitochondrial respiratory complex I, reducing cardiac-cell beat rate, with prolonged exposure resulting in cell death. We used a library of chemical variants of mubritinib and showed that modifying the 1H-1,2,3-triazole altered complex I inhibition, identifying the heterocyclic 1,3-nitrogen motif as the toxicophore. The same toxicophore is present in a second anti-cancer therapeutic carboxyamidotriazole (CAI) and we demonstrate that CAI also functions through complex I inhibition, mediated by the toxicophore. Complex I inhibition is directly linked to anti-cancer cell activity, with toxicophore modification ablating the desired effects of these compounds on cancer cell proliferation and apoptosis.


The pharmaceutical industry needs to make safe and effective drugs. At the same time this industry is under pressure to keep the costs of developing these drugs at an acceptable level. Drugs work by interacting with and typically blocking a specific target, such as a protein in a particular type of cell. Sometimes, however, drugs also bind other unexpected targets. These "off-target" effects can be the reason for a drug's toxicity, and it is important ­ both for the benefit of patients and the money that can be saved when developing drugs ­ to identify how drugs cause toxic side effects. The earlier researchers detect off-target effects, the better. Recent data has suggested that an anti-cancer drug called mubritinib has off-target effects on the compartments within cells that provide the cell with most of their energy, the mitochondria. This drug's intended target is a protein called HER2, which is found in large amounts on the surfaces of some breast cancer cells. Yet if mubritinib has this off-target effect on mitochondria, it may be harmful to other cells including heart cells because the heart is an organ that needs a large amount of energy from its mitochondria. Stephenson et al. have now performed experiments to show that mubritinib does not actually interact with HER2 at all, but only targets mitochondria. The effect of mubritinib as an anti-cancer drug is therefore only due to its activity against mitochondria. Digging deeper into the chemistry revealed the small parts of its chemical structure that was responsible for mubritinib's toxicity against heart cells, the so-called toxic substructure. Another anti-cancer drug called carboxyamidotriazole also has the same toxic substructure. Carboxyamidotriazole is supposed to stop cells from taking up calcium ions, but a final set of experiments demonstrated that this drug also only acts by inhibiting mitochondria. Often there is not enough information about many drugs' substructures, meaning off-target effects and toxicities cannot be predicted. The pharmaceutical industry will now be able to benefit from this new knowledge about the toxic substructures within some drugs. This research may also help patients who take mubritinib or carboxyamidotriazole, because their doctors will have to check for side effects on the heart more carefully.


Electron Transport Complex I/metabolism , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Oxazoles/pharmacology , Triazoles/pharmacology , Adenosine Triphosphate/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Death , Cell Line , Cell Proliferation/drug effects , Gene Expression Regulation/drug effects , Humans , Mitochondria/metabolism , Myocytes, Cardiac , Oxazoles/chemistry , Oxazoles/toxicity , Oxidative Phosphorylation , Protein Binding , Receptor, ErbB-2 , Triazoles/chemistry , Triazoles/toxicity
11.
BMC Cancer ; 13: 604, 2013 Dec 21.
Article En | MEDLINE | ID: mdl-24359526

BACKGROUND: Chk1 inhibitors have emerged as promising anticancer therapeutic agents particularly when combined with antimetabolites such as gemcitabine, cytarabine or hydroxyurea. Here, we address the importance of appropriate drug scheduling when gemcitabine is combined with the Chk1 inhibitor MK-8776, and the mechanisms involved in the schedule dependence. METHODS: Growth inhibition induced by gemcitabine plus MK-8776 was assessed across multiple cancer cell lines. Experiments used clinically relevant "bolus" administration of both drugs rather than continuous drug exposures. We assessed the effect of different treatment schedules on cell cycle perturbation and tumor cell growth in vitro and in xenograft tumor models. RESULTS: MK-8776 induced an average 7-fold sensitization to gemcitabine in 16 cancer cell lines. The time of MK-8776 administration significantly affected the response of tumor cells to gemcitabine. Although gemcitabine induced rapid cell cycle arrest, the stalled replication forks were not initially dependent on Chk1 for stability. By 18 h, RAD51 was loaded onto DNA indicative of homologous recombination. Inhibition of Chk1 at 18 h rapidly dissociated RAD51 leading to the collapse of replication forks and cell death. Addition of MK-8776 from 18-24 h after a 6-h incubation with gemcitabine induced much greater sensitization than if the two drugs were incubated concurrently for 6 h. The ability of this short incubation with MK-8776 to sensitize cells is critical because of the short half-life of MK-8776 in patients' plasma. Cell cycle perturbation was also assessed in human pancreas tumor xenografts in mice. There was a dramatic accumulation of cells in S/G2 phase 18 h after gemcitabine administration, but cells had started to recover by 42 h. Administration of MK-8776 18 h after gemcitabine caused significantly delayed tumor growth compared to either drug alone, or when the two drugs were administered with only a 30 min interval. CONCLUSIONS: There are two reasons why delayed addition of MK-8776 enhances sensitivity to gemcitabine: first, there is an increased number of cells arrested in S phase; and second, the arrested cells have adequate time to initiate recombination and thereby become Chk1 dependent. These results have important implications for the design of clinical trials using this drug combination.


Antineoplastic Combined Chemotherapy Protocols/pharmacology , Pancreatic Neoplasms/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , DNA Damage , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Drug Administration Schedule , Drug Resistance, Neoplasm , Drug Synergism , Humans , Mice , Mice, Nude , Pancreatic Neoplasms/pathology , Pyrazoles/administration & dosage , Pyrimidines/administration & dosage , Recombinational DNA Repair , S Phase Cell Cycle Checkpoints/drug effects , Tumor Burden/drug effects , Xenograft Model Antitumor Assays , Gemcitabine
12.
Mol Cancer Ther ; 11(2): 427-38, 2012 Feb.
Article En | MEDLINE | ID: mdl-22203733

Many anticancer agents damage DNA and arrest cell-cycle progression primarily in S or G(2) phase of the cell cycle. Previous studies with the topoisomerase I inhibitor SN38 have shown the efficacy of the Chk1 inhibitor UCN-01 to overcome this arrest and induce mitotic catastrophe. UCN-01 was limited in clinical trials by unfavorable pharmacokinetics. SCH900776 is a novel and more selective Chk1 inhibitor that potently inhibits Chk1 and abrogates cell-cycle arrest induced by SN38. Like UCN-01, abrogation of SN38-induced arrest enhances the rate of cell death but does not increase overall cell death. In contrast, SCH900776 reduced the growth-inhibitory concentration of hydroxyurea by 20- to 70-fold. A similar magnitude of sensitization was observed with cytarabine. A 5- to 10-fold sensitization occurred with gemcitabine, but no sensitization occurred with cisplatin, 5-fluorouracil, or 6-thioguanine. Sensitization occurred at hydroxyurea concentrations that marginally slowed DNA replication without apparent activation of Chk1, but this led to dependence on Chk1 that increased with time. For example, when added 18 hours after hydroxyurea, SCH900776 induced DNA double-strand breaks consistent with rapid collapse of replication forks. In addition, some cell lines were highly sensitive to SCH900776 alone, and these cells required lower concentrations of SCH900776 to sensitize them to hydroxyurea. We conclude that some tumors may be very sensitive to the combination of SCH900776 and hydroxyurea. Delayed administration of SCH900776 may be more effective than concurrent treatment. SCH900776 is currently in phase I clinical trials, and these results provide the rationale and schedule for future clinical trials.


Antimetabolites/pharmacology , Antineoplastic Agents/pharmacology , Cell Cycle Checkpoints/drug effects , Protein Kinases/metabolism , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Blotting, Western , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Checkpoint Kinase 1 , Cytarabine/pharmacology , DNA Breaks, Double-Stranded/drug effects , DNA Damage , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Drug Synergism , Flow Cytometry , Humans , Hydroxyurea/pharmacology , Irinotecan , Molecular Structure , Phosphorylation/drug effects , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Pyrazoles/chemistry , Pyrimidines/chemistry , Staurosporine/analogs & derivatives , Staurosporine/pharmacology , Gemcitabine
13.
Arch Pharm Res ; 30(6): 770-7, 2007 Jun.
Article En | MEDLINE | ID: mdl-17679557

Great expectations are set on gene therapy for the treatment of malignant hepatocellular carcinomas (HCC) in East Asia. Recombinant adenoviral vectors (AV) have been developed in which the L-plastin promoter (LP) regulates the expression of transgenes, in a tumor cell specific manner, resulting in an increase in the therapeutic index. The development of the AdLPCD vector, a replication-incompetent AV, containing a transcription unit of LP and E. coli cytosine deaminase (CD), was reported in our previous work. In the present study, the AdLPCD vector combined with 5-fluorocytosine (5-FC) administration was tested to see if it might have significant utility in the chemosensitization of L-plastin positive HCC. Four HCC cell lines (HepG2, Chang Liver, Huh-7 and SK-Hep-1 cells) were investigated for the expression of LacZ after infecting the cells with the AdLPLacZ vector containing a 2.4 kb fragment of LP and the LacZ gene. Relatively high levels of LP activity were detected in HepG2, followed by Chang Liver cells; whereas, no promoter activity was found in Huh-7 and SK-Hep-1 cells, as determined by AdLPLacZ infection followed by the beta-galactosidase assay. In addition, the results of RT-PCR assays for the detection of endogenous L-plastin mRNA in these cells lines correlated well with those of the beta-galactosidase activity after infection with AdLPLacZ. Based on these data, the cytotoxic effect of AdLPCD/5-FC was evaluated in HepG2 cells. These results indicate that the CD gene delivered by AV could sensitize HepG2 cells to the prodrug, 5-FC. However, the observed effects were insufficient to cause the death of most of cells. This suggests that the screening of patients for an AdLP/5-FC strategy based on AdLPLacZ data might not always guarantee a good therapeutic outcome.


Adenoviridae/genetics , Carcinoma, Hepatocellular/therapy , Cytosine Deaminase/genetics , Genetic Vectors/therapeutic use , Liver Neoplasms/therapy , Phosphoproteins/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cytomegalovirus/genetics , Escherichia coli/genetics , Genetic Vectors/genetics , Humans , Lac Operon , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Membrane Glycoproteins , Microfilament Proteins , Promoter Regions, Genetic/genetics , Reverse Transcriptase Polymerase Chain Reaction , Virus Replication/genetics , beta-Galactosidase/metabolism
14.
Arch Pharm Res ; 27(6): 633-9, 2004 Jun.
Article En | MEDLINE | ID: mdl-15283466

The expression of therapeutic transgenes in recombinant adenoviral vectors is a major cause of toxicity in dividing cancer cells as well as non dividing normal cells. To solve the problem of toxicity to normal cells, we have reported on a recombinant adenoviral vector system (AdLP-) in which the expression of the transgene is directed by the tumor-specific L-plastin promoter (LP) (Chung et al., 1999). The object of this study was to generate a recombinant adenoviral vector system which would generate tumor cell specific expression of cytosine deaminase (CD) gene. We report the construction of a replication-incompetent adenoviral vector in which CD is driven by the L-plastin promoter (AdLPCD). Infection of 293 cells by AdLPCD generated the functional CD protein as measured by HPLC analysis for the conversion of 5-Fluorocytosine (5-FC) to 5-Fluorouracil (5-FU). HPLC analysis in conjunction with counting radioactivity for [6-3H]-5FC and [6-3H]-5FU demonstrated vector dose-dependent conversion of 5-FC to 5-FU in AdLPCD infected ovarian cancer cells. The results from present and previous studies (Peng et al., 2001; Akbulut et al., 2003) suggest that the use of the AdLPCD/5-FC system may be of value in the treatment of cancer including microscopic ovarian cancer in the peritoneal cavity.


Adenoviridae/genetics , Cytosine Deaminase/genetics , Gene Transfer Techniques , Phosphoproteins/genetics , Artificial Gene Fusion , Cell Line, Tumor , Chromatography, High Pressure Liquid , Cytosine Deaminase/biosynthesis , Female , Flucytosine/pharmacokinetics , Fluorouracil/metabolism , Genetic Vectors , Humans , Membrane Glycoproteins , Microfilament Proteins , Ovarian Neoplasms , Polymerase Chain Reaction , Promoter Regions, Genetic
15.
Arch Pharm Res ; 25(3): 375-80, 2002 Jun.
Article En | MEDLINE | ID: mdl-12135113

Cytochrome P4501A2 (CYP1A2) is a member of the cytochrome P450 family of isozymes involved in the phase I drug metabolism of vertebrates. CYP1A2 is responsible for the activation of a number of aromatic amines to mutagenic and carcinogenic forms. Thus, the level of CYP1A2, which varies among different populations, may determine an individual's susceptibility to these chemicals. We have previously reported on the importance of a cis element named PRB (protected region B) in the regulation of human Cytochrome P4501A2 (CYP1A2) gene, which appeared to act as a positive regulatory element. Closer examination of the PRB sequence (-2218 to -2187 bp) revealed a putative AP-1 binding site, TGACTAA, at -2212 bp (Chung and Bresnick, 1997). To elucidate the role of AP-1 in CYP1A2 regulation, we transiently overexpressed c-Jun and c-Fos transcription factors in human hepatoma HepG2 cells, and examined their influence on the CYP1A2 promoter activity by reporter gene assays. Cotransfection of the c-Jun and the c-Fos expression vectors increased the induced transactivation by five to six fold from the CYP1A2 promoter constructs. However, deletion of the PRB element did not affect the degree of activation by the c-Jun and the c-Fos. Therefore, it is unlikely that the c-Jun and the c-Fos activate the CYP1A2 promoter through this AP-1 consensus-like sequence in the PRB region.


Cytochrome P-450 CYP1A2/genetics , Response Elements/genetics , Transcription Factor AP-1/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cells, Cultured , Genes, fos/genetics , Genes, jun/genetics , Humans , Luciferases/genetics , Plasmids/genetics , Promoter Regions, Genetic/genetics , Protein Biosynthesis , Transfection
...