Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters











Publication year range
1.
Cancer Lett ; 445: 1-10, 2019 03 31.
Article in English | MEDLINE | ID: mdl-30583077

ABSTRACT

We previously demonstrated that some N-biphenylanilides caused cell-cycle arrest at G2/M transition in breast cancer cells. Among them we choose three derivatives, namely PTA34, PTA73 and RS35 for experimentation in solid tumor cell lines, classical Hodgkin Lymphoma (cHL) cell lines and bona fide normal cell lines. Almost all tumor cells were sensitive to compounds in the nanomolar range whereas, they were not cytotoxic to normal ones. Interestingly the compounds caused a strong G2/M phase arrest in cHL cell lines, thus, here we investigated whether they affected the integrity of microtubules in such cells. We found that they induced a long prometaphase arrest, followed by induction of apoptosis which involved mitochondria. PTA73 and RS35 induced the mitotic arrest through the fragmentation of microtubules which prevented the kinethocore-mitotic spindle interaction and the exit from mitosis. PTA34 is instead a tubulin-targeting agent because it inhibited the tubulin polymerization as vinblastine. As such, PTA34 maintained the Cyclin B1-CDK1 regulatory complex activated during the G2/M arrest while inducing the inactivation of Bcl-2 through phosphorylation in Ser70, the degradation of Mcl-1 and a strong activation of BIML and BIMS proapoptotic isoforms. In addition PTA34 exerted an antiangiogenic effect by suppressing microvascular formation.


Subject(s)
Antimitotic Agents/chemical synthesis , Biphenyl Compounds/chemical synthesis , Hodgkin Disease/metabolism , Nicotine/chemistry , Antimitotic Agents/chemistry , Antimitotic Agents/pharmacology , Biphenyl Compounds/chemistry , Biphenyl Compounds/pharmacology , CDC2 Protein Kinase/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclin B1/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Hodgkin Disease/drug therapy , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Molecular Structure , Prometaphase/drug effects
2.
Arch Dermatol Res ; 307(6): 495-503, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25736238

ABSTRACT

Environmental and genetic risk factors are involved in the development of melanoma. The role of the melanocortin 1 receptor (MC1R) gene has been investigated and differences according to geographic areas have been described. To evaluate the role of some clinical and genetic risk factors in melanoma development, we performed a case-control study involving 101 melanoma patients and 103 controls coming from South-Eastern Italy (Puglia), after achieving informed consent. We confirmed the role of known clinical risk factors for melanoma. Furthermore, 42 MC1R polymorphisms were observed. Three of these variants (L26V, H232L, D294Y) were not previously reported in the literature. Their predicted impact on receptor function was evaluated using bioinformatic tools. We report an overall frequency of MC1R variants in our population higher than in Northern or Central Italy. The most common polymorphism found was V60L, that has been recently reported to spread among South Mediterranean population. This variant influenced phenotypic characteristics of our population while it did not impinge on melanoma risk. An increased risk of melanoma was associated with two or more MC1R variants, when at least one was RHC, compared to people carrying the MC1R consensus sequence or a single MC1R polymorphism. Interestingly, we observed an increased risk of melanoma in subjects with darker skin and lower nevus count, usually considered at low risk, when carrying MC1R polymorphisms.


Subject(s)
Melanoma/genetics , Polymorphism, Genetic/physiology , Receptor, Melanocortin, Type 1/genetics , Skin Neoplasms/genetics , Adult , Aged , Case-Control Studies , Female , Genotyping Techniques , Humans , Italy/epidemiology , Logistic Models , Male , Melanoma/epidemiology , Middle Aged , Risk Factors , Skin Neoplasms/epidemiology
3.
Biochim Biophys Acta ; 1837(8): 1338-49, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24709060

ABSTRACT

Apoptosis in neuronal tissue is an efficient mechanism which contributes to both normal cell development and pathological cell death. The present study explores the effects of extracellular ADP on low [K(+)]-induced apoptosis in rat cerebellar granule cells. ADP, released into the extracellular space in brain by multiple mechanisms, can interact with its receptor or be converted, through the actions of ectoenzymes, to adenosine. The findings reported in this paper demonstrate that ADP inhibits the proapoptotic stimulus supposedly via: i) inhibition of ROS production during early stages of apoptosis, an effect mediated by its interaction with cell receptor/s. This conclusion is validated by the increase in SOD and catalase activities as well as by the GSSG/GSH ratio value decrease, in conjunction with the drop of ROS level and the prevention of the ADP protective effect by pyridoxalphosphate-6-azophenyl-2',4'-disulfonic acid (PPADS), a novel functionally selective antagonist of purine receptor; ii) safeguard of the functionality of the mitochondrial adenine nucleotide-1 translocator (ANT-1), which is early impaired during apoptosis. This effect is mediated by its plausible internalization into cell occurring as such or after its hydrolysis, by means of plasma membrane nucleotide metabolizing enzymes, and resynthesis into the cell. Moreover, the findings that ADP also protects ANT-1 from the toxic action of the two Alzheimer's disease peptides, i.e. Aß1-42 and NH2htau, which are known to be produced in apoptotic cerebellar neurons, further corroborate the molecular mechanism of neuroprotection by ADP, herein proposed.


Subject(s)
Adenine Nucleotide Translocator 1/metabolism , Adenosine Diphosphate/metabolism , Antioxidants/metabolism , Mitochondria/drug effects , Adenine Nucleotide Translocator 1/genetics , Amyloid beta-Peptides/toxicity , Animals , Apoptosis/genetics , Cerebellum/cytology , Cerebellum/drug effects , Cerebellum/metabolism , Humans , Mitochondria/metabolism , Neurons/metabolism , Neuroprotective Agents/metabolism , Primary Cell Culture , Rats , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , tau Proteins/toxicity
4.
Curr Med Chem ; 21(8): 1039-49, 2014.
Article in English | MEDLINE | ID: mdl-23992324

ABSTRACT

Antivascular approaches aim to cause rapid and catastrophic shutdown in the vascular function of the tumour, leading to extensive tumour cell death. Tumour vascular disrupting agents (VDAs) are a new class of cancer therapies that target the existing vasculature of tumours, taking advantage of the relative instability of tumour vasculature and its supporting structures. Treatment with VDAs induces a rapid collapse and regression of tumour vessels, with a consequent deprivation of blood and oxygen which leads to ischemic or hemorrhagic necrosis of the tumour. In this review, an overview of the most recently developed vascular disrupting agents is reported, focusing on the biological effects exerted by these compounds on endothelial cells and tumour vasculature, potentially effective in the treatment of several malignancies including upper gastrointestinal tumours. In particular, we have focused on the antimitotic agent combretastatin and its numerous synthetic analogues such as combretastatin A-4-phosphate, OXI4503, and AVE8062, and on the colchicine analogue ZD6126.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Bibenzyls/therapeutic use , Gastrointestinal Neoplasms/blood supply , Gastrointestinal Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Upper Gastrointestinal Tract/blood supply , Upper Gastrointestinal Tract/pathology , Animals , Diphosphates/therapeutic use , Gastrointestinal Neoplasms/pathology , Humans , Neovascularization, Pathologic/pathology , Organophosphorus Compounds/therapeutic use , Serine/analogs & derivatives , Serine/therapeutic use , Stilbenes/therapeutic use , Upper Gastrointestinal Tract/drug effects
5.
Curr Pharm Des ; 19(5): 918-26, 2013.
Article in English | MEDLINE | ID: mdl-22973960

ABSTRACT

Single-agent therapy with molecularly targeted agents has shown limited success in tumor growth control, mainly because escape or resistance mechanisms are activated once a signalling molecule is inhibited. Rational combinations of target-specific agents could counteract this response providing a useful strategy in cancer treatment. In this regard, the EGFR and mTOR inhibitors have been used together to generate a synergistic effect and maximize the efficacy of each individual agent. Overall, the in vivo and in vitro evidences support the utilization of combinations targeting EGFR and mTOR, for malignancies characterized by deregulated EGFR/PI3K/Akt/ mTOR signalling cascade; whereas the clinical experience points out that the assessment of the therapeutic value of such combination awaits further investigations.


Subject(s)
Antineoplastic Agents/pharmacology , ErbB Receptors/antagonists & inhibitors , TOR Serine-Threonine Kinases/antagonists & inhibitors , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Drug Resistance, Neoplasm , Drug Synergism , Humans , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects
6.
Curr Med Chem ; 19(23): 3858-73, 2012.
Article in English | MEDLINE | ID: mdl-22788762

ABSTRACT

A large body of evidence point out that the onset of synthetic lethality may provide a useful tool for amplifying the efficacy of drugs in anticancer regimens, to uncover interdependence between genes and to identify predictive factors that would be extremely useful to guide in the selection of more effective targeted drugs and drug combinations for each patient. Here, we provide an overview on the exploitation of synthetic lethality to overcome drug resistance to conventional chemotherapy in several types of solid tumors. We report recent findings on cellular markers and gene mutations which are specifically essential for the viability of cancer cells and for resistance to chemotherapeutics. In addition, new molecularly targeted strategies to overcome drug resistance are suggested.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , DNA Repair , Humans , Mutation , Neoplasms/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Receptors, Death Domain/genetics , Receptors, Death Domain/metabolism
7.
J Med Chem ; 55(1): 37-54, 2012 Jan 12.
Article in English | MEDLINE | ID: mdl-22081932

ABSTRACT

A series of ureidofibrate-like derivatives was prepared and assayed for their PPAR functional activity. A calorimetric approach was used to characterize PPARγ-ligand interactions, and docking experiments and X-ray studies were performed to explain the observed potency and efficacy. R-1 and S-1 were selected to evaluate several aspects of their biological activity. In an adipogenic assay, both enantiomers increased the expression of PPARγ target genes and promoted the differentiation of 3T3-L1 fibroblasts to adipocytes. In vivo administration of these compounds to insulin resistant C57Bl/6J mice fed a high fat diet reduced visceral fat content and body weight. Examination of different metabolic parameters showed that R-1 and S-1 are insulin sensitizers. Notably, they also enhanced the expression of hepatic PPARα target genes indicating that their in vivo effects stemmed from an activation of both PPARα and γ. Finally, the capability of R-1 and S-1 to inhibit cellular proliferation in colon cancer cell lines was also evaluated.


Subject(s)
Benzoxazoles/chemistry , Fibric Acids/chemistry , PPAR alpha/metabolism , PPAR gamma/metabolism , Propionates/chemistry , Urea/chemistry , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Benzoxazoles/chemical synthesis , Benzoxazoles/pharmacology , Body Weight/drug effects , Calorimetry , Cell Differentiation/drug effects , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Crystallography, X-Ray , Drug Partial Agonism , Drug Screening Assays, Antitumor , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Profiling , Humans , Insulin Resistance , Intra-Abdominal Fat/drug effects , Liver/drug effects , Liver/metabolism , Mice , Mice, Inbred C57BL , Models, Molecular , PPAR alpha/agonists , PPAR alpha/genetics , PPAR gamma/agonists , PPAR gamma/genetics , Propionates/chemical synthesis , Propionates/pharmacology , Stereoisomerism , Structure-Activity Relationship
8.
Br J Cancer ; 104(5): 769-80, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21304529

ABSTRACT

BACKGROUND: AZD1152, the prodrug for AZD1152-hydroxyquinazoline pyrazol anilide (HQPA), is a selective inhibitor of Aurora B kinase activity. Preclinical evaluation of AZD1152 has been reported in several human cancer models. The potentiality of this compound in combination therapy warrants further investigation in solid tumours. EXPERIMENTAL DESIGN: This study explored the effects of AZD1152-HQPA in colon and pancreatic tumour cells. The antitumour properties of AZD1152, either as single agent or in combination with chemotherapeutics, were evaluated in each study model. The efficacy and the toxicity of AZD1152 alone and in combination with gemcitabine were validated in pancreatic tumour xenograft model. RESULTS: AZD1152-HQPA treatment resulted in a dramatic increase of chromosome number, modification of cell cycle and induction of apoptosis. The most effective combination was that with chemotherapeutics given soon after AZD1152 in both tumour cell types. The effectiveness of the sequential schedule of AZD1152 with gemcitabine was confirmed in nude mice bearing MiaPaCa-2 tumours, showing inhibition of tumour volumes and delaying of tumour growth after the interruption of the treatments. Here we show that AZD1152-HQPA enhances oxaliplatin and gemcitabine effectiveness in colon and pancreatic cancer, respectively. First, we provide advances into administration schedules and dosing regimens for the combination treatment in in vivo pancreatic tumour.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Deoxycytidine/analogs & derivatives , Organophosphates/therapeutic use , Pancreatic Neoplasms/drug therapy , Protein Serine-Threonine Kinases/antagonists & inhibitors , Quinazolines/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Aurora Kinase B , Aurora Kinases , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Colonic Neoplasms , Deoxycytidine/administration & dosage , Drug Screening Assays, Antitumor , Humans , Male , Mice , Mice, Nude , Organophosphates/adverse effects , Organoplatinum Compounds/administration & dosage , Oxaliplatin , Quinazolines/adverse effects , Xenograft Model Antitumor Assays , Gemcitabine
9.
Ann Oncol ; 22 Suppl 1: i53-9, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21285153

ABSTRACT

A number of inhibitors of DNA repair have been evaluated or are undergoing development as potential cancer treatments. Inhibitors of poly(ADP-ribose) polymerase (PARP) are of particular interest in treating hereditary breast cancers occurring in patients who are carriers of BRCA1 or BRCA2 mutations. In vitro PARP inhibitors are highly cytotoxic to cell lines carrying BRCA mutations while only minimally toxic to cell lines without these mutations. This is thought to be due to a phenomenon known as synthetic lethality where the accumulation of single-strand breaks consequent on PARP inhibition are converted to double-strand breaks on cell division. Cancer cells in BRCA carriers are uniquely unable to repair the consequent double-strand breaks that result during cell division. PARP inhibitors were initially developed as possible chemo-potentiating agents but have now been evaluated clinically in BRCA-related tumors, showing remarkable single-agent activity. The potential future development and use is reviewed.


Subject(s)
Enzyme Inhibitors/pharmacology , Neoplasms/drug therapy , Neoplasms/enzymology , Poly(ADP-ribose) Polymerase Inhibitors , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Female , Genes, BRCA1 , Genes, BRCA2 , Humans , Neoplasms/genetics , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/genetics
10.
Cancer Treat Rev ; 36 Suppl 3: S46-55, 2010 Nov.
Article in English | MEDLINE | ID: mdl-21129610

ABSTRACT

Capecitabine is an orally administered fluoropyrimidine carbamate which has been developed as a prodrug of 5-FU with the goal to improve its tolerability and intratumoral drug concentration. The review aims to provide an evidence-based update of clinical trials investigating the clinical efficacy, adverse-event profile, dosage and administration of this drug, alone or in combination with conventional chemotherapeutics and/or new target-oriented drugs, in the management of colorectal cancer patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/drug therapy , Deoxycytidine/analogs & derivatives , Fluorouracil/analogs & derivatives , Administration, Oral , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Humanized , Antimetabolites, Antineoplastic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Bevacizumab , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Capecitabine , Chemotherapy, Adjuvant , Deoxycytidine/administration & dosage , Deoxycytidine/adverse effects , Deoxycytidine/therapeutic use , Diarrhea/chemically induced , Drug Administration Schedule , Fluorouracil/administration & dosage , Fluorouracil/adverse effects , Fluorouracil/therapeutic use , Humans , Irinotecan , Leucovorin/administration & dosage , Neoadjuvant Therapy , Organoplatinum Compounds/administration & dosage , Oxaloacetates , Prodrugs/therapeutic use , Radiotherapy, Adjuvant
11.
Int J Mol Med ; 21(6): 809-17, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18506376

ABSTRACT

Recently improved understanding of the pathogenesis of human head and neck squamous cell carcinoma (HNSCC) has led to the development of new, molecular-based therapeutic strategies, one of the more promising is the utilisation of tyrosine kinase (TK) inhibitors, targeting epidermal growth factor receptor (EGFR). In this study, we tested for gefitinib effectiveness in a broad panel of 12 newly established HNSCC cell lines, investigating its ability to reduce cell growth, to induce apoptosis and to modulate cell cycle and various EGFR pathway-related targets. Gefitinib IC50 values ranged between 0.064 and 33 microM, its capability to induce apoptosis and cell accumulation in G0/G1 phase was cell line-specific, and the main EGFR-related pathway involved in gefitinib activity was PI3K/Akt/mTor. We characterised our in vitro panel extensively, with the aim to identify predictive factors for gefitinib effectiveness; all cell lines were free of human papillomavirus infection, two were positive for Fhit expression, four expressed wild-type p53, and all of them variously expressed the other two p53 family members, p63 and p73. The comparison between the targets analysed and gefitinib effectiveness evidenced the absence of a clear relationship, excluding them as predictive factors for gefitinib efficacy. Our results confirmed the in vitro efficacy of an anti-EGFR approach, but other targets than those analysed here should be characterised in order to identify valid predictive factors for gefitinib utilisation.


Subject(s)
ErbB Receptors/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Acid Anhydride Hydrolases/genetics , Acid Anhydride Hydrolases/metabolism , Apoptosis/drug effects , Blotting, Western , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Cycle/drug effects , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , ErbB Receptors/genetics , ErbB Receptors/metabolism , G1 Phase/drug effects , Gefitinib , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Immunoprecipitation , Inhibitory Concentration 50 , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mutation , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Resting Phase, Cell Cycle/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Tumor Protein p73 , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
12.
Br J Cancer ; 91(11): 1964-9, 2004 Nov 29.
Article in English | MEDLINE | ID: mdl-15545972

ABSTRACT

Prognosis and survival of patients with hepatocellular carcinoma (HCC) is still very poor, and no therapies are currently available to inhibit tumour growth and metastases. Recently, we reported that the expression of an extracellular matrix component (ECM), namely Laminin-5 (Ln-5), is directly related to poor prognosis in HCC patients. The aim of our study is to investigate the preclinical effect of gefitinib in an in vitro HCC model. We found that the IC(50) of gefitinib in HCC cells ranged from 0.7 to 10.0 muM, whereas Ln-5 inhibited the activity of gefitinib in a dose-dependent manner. Complete inhibition of phosphorylated (p)-EGFR (epidermal growth factor receptor) was obtained within 6 h exposure to gefitinib and complete restoration of the receptor status was obtained after 24 h. A downstream effect yields a decrease in p-Akt and p-Erk 1/2. The addition of exogenous Ln-5 has no effect on p-EGFR, whereas it restores p-Erk 1/2 and p-Akt. Consistently, Ln-5 induces recovery of HCC cells from Gefitinib-induced apoptosis. In conclusion, gefitinib inhibits HCC cell growth and we report for the first time that Ln-5, but not other ECM molecules, reduces the ability of gefitinib to inhibit cell growth via Akt. As patients with HCC have different Ln-5 expression levels, these results may help to better understand which patients might benefit from gefitinib treatment.


Subject(s)
Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Carcinoma, Hepatocellular/drug therapy , Cell Adhesion Molecules/metabolism , Drug Resistance, Neoplasm , Quinazolines/therapeutic use , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/drug effects , ErbB Receptors/metabolism , Gefitinib , Humans , Phosphorylation/drug effects , Predictive Value of Tests , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Kalinin
13.
J Biol Chem ; 275(47): 37159-66, 2000 Nov 24.
Article in English | MEDLINE | ID: mdl-10980192

ABSTRACT

In rat cerebellar granule cells both reactive oxygen species production and release of cytochrome c take place during glutamate toxicity. This investigation was aimed (i) to ascertain whether and how these two processes are related and (ii) to gain insight into the role played by the released cytochrome c in the onset of neurotoxicity. Cytochrome c release takes place owing to the generation of reactive oxygen species both in glutamate-treated cerebellar granule cells and in sister control cultures incubated in the presence of the reactive oxygen species-generating system consisting of xanthine plus xanthine oxidase. In the early phase of neurotoxicity (30-min glutamate exposure) about 40% of the maximum (as measured at 3 h of glutamate exposure) cytochrome c release was found to occur in cerebellar granule cells from mitochondria that were essentially coupled and intact and that had a negligible production of oxygen free radicals. Contrarily, mitochondria from cells treated with glutamate for 3 h were mostly uncoupled and produced reactive oxygen species at a high rate. The cytosolic fraction containing the released cytochrome c was able to transfer electrons from superoxide anion to molecular oxygen via the respiratory chain and was found to partially prevent glutamate toxicity when added externally to cerebellar neurons undergoing necrosis. In the light of these findings, we propose that in the early phase of neurotoxicity, cytochrome c release can be part of a cellular and mitochondrial defense mechanism against oxidative stress.


Subject(s)
Cerebellum/cytology , Cytochrome c Group/metabolism , Mitochondria/metabolism , Neurons/metabolism , Reactive Oxygen Species/metabolism , Animals , Cells, Cultured , Culture Techniques , Glutamic Acid/pharmacology , Oxidative Stress , Oxygen Consumption , Rats
14.
Biochim Biophys Acta ; 1386(1): 29-38, 1998 Jul 28.
Article in English | MEDLINE | ID: mdl-9675237

ABSTRACT

Kinetic properties and thermal stabilities of the precursor form of mitochondrial aspartate aminotransferase, the mature form lacking 9 amino acids from the N-terminus, and forms of the mature protein in which cysteine-166 had been mutated to serine or alanine were compared with those of the mature enzyme. The precursor and the cysteine mutants showed moderately impaired catalytic properties consistent with decreased ability to undergo transition from the open to the closed conformation which is an integral part of the mechanism of action of the enzyme. The deletion mutant had a kcat only 2% of that of the mature enzyme but also much reduced Km values for both substrates. In addition it showed enhanced reactivity of cysteine-166 with 5,5'-dithiobis(2-nitrobenzoate), which is characteristic of the closed form of the enzyme, with no enhancement of reactivity in the presence of substrates. This is taken to show that the deletion mutant adopts a conformation that is significantly different from that of the mature enzyme particularly in respect of the small domain. The deletion mutant was found to be more resistant to thermal inactivation over a range of temperatures than were the other forms of the enzyme consistent with its having a more tightly packed small domain.


Subject(s)
Aspartate Aminotransferases/chemistry , Aspartate Aminotransferases/metabolism , Mitochondria/enzymology , Animals , Aspartate Aminotransferases/genetics , Chickens , Enzyme Stability , Hot Temperature , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Kinetics , Mutation , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Engineering , Protein Precursors/chemistry , Protein Precursors/genetics , Protein Precursors/metabolism , Sequence Deletion , Structure-Activity Relationship
15.
Biochem Biophys Res Commun ; 215(3): 800-7, 1995 Oct 24.
Article in English | MEDLINE | ID: mdl-7488044

ABSTRACT

Sensitivity to digestion with pronase has been used to show that the precursor form of mitochondrial aspartate aminotransferase, the form lacking the N-terminal presequence, that with a deletion of the first 9 residues and mutants of the mature enzyme in which residue Cys-166 is mutated to alanine or serine, all retain unfolded conformations after synthesis in a reticulocyte lysate. In the presence of lysed mitochondria the various forms of mitochondrial aspartate aminotransferase retained their susceptibilities to pronase in a way that mirrored the efficiencies with which they are imported into intact mitochondria. The results are interpreted as showing that the presequence of mitochondrial aspartate aminotransferase is not uniquely required for interaction with cytosolic factors required to maintain the newly synthesised protein in a form competent for interacting with, and being imported into, mitochondria.


Subject(s)
Aspartate Aminotransferases/chemistry , Cysteine , Mitochondria, Liver/enzymology , Point Mutation , Pronase/metabolism , Protein Conformation , Alanine , Animals , Aspartate Aminotransferases/biosynthesis , Aspartate Aminotransferases/metabolism , Cell-Free System , Kinetics , Mutagenesis, Site-Directed , Plasmids , Rats , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Serine , Tetrahydrofolate Dehydrogenase/chemistry , Tetrahydrofolate Dehydrogenase/metabolism
16.
Biochem Biophys Res Commun ; 214(2): 511-7, 1995 Sep 14.
Article in English | MEDLINE | ID: mdl-7677759

ABSTRACT

Mutant genes were constructed which coded for the precursor form of mitochondrial aspartate aminotransferase in which residue cysteine 166 was mutated to either serine or alanine and for forms of the protein lacking both the presequence and residues 1-9 of the mature protein but carrying the same cysteine mutations. The protein products of all of these mutant genes were imported into mitochondria that had been added to the expression system but with varying degrees of efficiency. The results showed that the effects of mutation of cysteine 166 and of deletion of residues 1-9 of the mature protein on sequestration into mitochondria were essentially cumulative, suggesting that these parts of the protein are involved in distinct steps on the recognition/uptake pathway.


Subject(s)
Aspartate Aminotransferases/metabolism , Mitochondria/metabolism , Point Mutation , Alanine , Amino Acid Sequence , Animals , Aspartate Aminotransferases/biosynthesis , Aspartate Aminotransferases/genetics , Cysteine , Kinetics , Mutagenesis, Site-Directed , Protein Processing, Post-Translational , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Serine
17.
Biochem Biophys Res Commun ; 201(3): 1059-65, 1994 Jun 30.
Article in English | MEDLINE | ID: mdl-8024546

ABSTRACT

Two fused genes were constructed which encode for two chimeric proteins in which either 10 or 191 N-terminal amino acids of mature mitochondrial aspartate aminotransferase had been attached to the entire polypeptide chain of cytosolic dihydrofolate reductase. The precursor and mature form of mitochondrial aspartate aminotransferase, dihydrofolate reductase and both chimeric proteins were synthesized in vitro and their import into isolated mitochondria was studied. Both chimeric proteins were taken up by isolated organelles, where they became protease resistant, thus indicating the ability of the N-terminal portion of the mature moiety of the precursor of mitochondrial aspartate aminotransferase to direct cytosolic dihydrofolate reductase into mitochondria.


Subject(s)
Aspartate Aminotransferases/metabolism , Mitochondria/enzymology , Tetrahydrofolate Dehydrogenase/metabolism , Aspartate Aminotransferases/chemistry , Cell Compartmentation , Cytosol/enzymology , In Vitro Techniques , Recombinant Fusion Proteins , Structure-Activity Relationship , Tetrahydrofolate Dehydrogenase/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL