Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Proc Natl Acad Sci U S A ; 108(32): 13258-63, 2011 Aug 09.
Article in English | MEDLINE | ID: mdl-21788490

ABSTRACT

Myocardial ischemic disease is the major cause of death worldwide. After myocardial infarction, reperfusion of infracted heart has been an important objective of strategies to improve outcomes. However, cardiac ischemia/reperfusion (I/R) is characterized by inflammation, arrhythmias, cardiomyocyte damage, and, at the cellular level, disturbance in Ca(2+) and redox homeostasis. In this study, we sought to determine how acute inflammatory response contributes to reperfusion injury and Ca(2+) homeostasis disturbance after acute ischemia. Using a rat model of I/R, we show that circulating levels of TNF-α and cardiac caspase-8 activity were increased within 6 h of reperfusion, leading to myocardial nitric oxide and mitochondrial ROS production. At 1 and 15 d after reperfusion, caspase-8 activation resulted in S-nitrosylation of the RyR2 and depletion of calstabin2 from the RyR2 complex, resulting in diastolic sarcoplasmic reticulum (SR) Ca(2+) leak. Pharmacological inhibition of caspase-8 before reperfusion with Q-LETD-OPh or prevention of calstabin2 depletion from the RyR2 complex with the Ca(2+) channel stabilizer S107 ("rycal") inhibited the SR Ca(2+) leak, reduced ventricular arrhythmias, infarct size, and left ventricular remodeling after 15 d of reperfusion. TNF-α-induced caspase-8 activation leads to leaky RyR2 channels that contribute to myocardial remodeling after I/R. Thus, early prevention of SR Ca(2+) leak trough normalization of RyR2 function is cardioprotective.


Subject(s)
Caspase 8/metabolism , Heart Ventricles/pathology , Myocardial Reperfusion Injury/enzymology , Myocardial Reperfusion Injury/pathology , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Enzyme Activation , Fluorescence , Myocardial Reperfusion Injury/blood , Myocardial Reperfusion Injury/physiopathology , Myocardium/metabolism , Myocardium/pathology , Phenanthridines/metabolism , Rats , Rats, Inbred WKY , Tumor Necrosis Factor-alpha/blood , Ventricular Remodeling
2.
Behav Brain Res ; 212(1): 56-63, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20347879

ABSTRACT

Among experimental models of perinatal ischemic stroke, Renolleau's model mimics selected types of stroke at birth, including ischemia and reperfusion. However, its behavioural consequences on development have been poorly described. Here, ischemia-reperfusion was performed in 7-day-old Wistar rats. Between the ages of 9 and 40 days, sensorimotor and memory functions were assessed. The infarcted area was analysed by immunohistochemistry at 40 days of age. The remaining lesion was in the parietal cortex, in the form of a cone-shaped area. This area contained glial cells but neither neurons nor macrophages. Transient focal neonatal ischemia led to sensorimotor alterations in early adulthood, such as postural asymmetry, motor coordination and somatosensory deficits, and hyperactivity, as well as cognitive impairments, such as spatial reference memory deficits. Based on these results, we propose here a selection of behavioural tests that should constitute meaningful tools for assessing sensory and cognitive functions after experimental neonatal ischemic stroke.


Subject(s)
Gait Disorders, Neurologic/diagnosis , Gait Disorders, Neurologic/etiology , Ischemic Attack, Transient/complications , Memory Disorders/diagnosis , Memory Disorders/etiology , Reperfusion , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Antigens/metabolism , Avoidance Learning/physiology , Brain/metabolism , Brain/pathology , Disease Models, Animal , Double-Blind Method , Ectodysplasins/metabolism , Exploratory Behavior/physiology , Female , Glial Fibrillary Acidic Protein/metabolism , Male , Maze Learning/physiology , Motor Activity/physiology , Phosphopyruvate Hydratase/metabolism , Predictive Value of Tests , Pregnancy , Proteoglycans/metabolism , Rats , Rats, Wistar , Reflex/physiology , Statistics as Topic
3.
Apoptosis ; 14(10): 1190-203, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19693674

ABSTRACT

Dengue viruses belong to the Flavivirus family and are responsible for hemorrhagic fever in Human. Dengue virus infection triggers apoptosis especially through the expression of the small membrane (M) protein. Using isolated mitochondria, we found that synthetic peptides containing the C-terminus part of the M ectodomain caused apoptosis-related mitochondrial membrane permeabilization (MMP) events. These events include matrix swelling and the dissipation of the mitochondrial transmembrane potential (DeltaPsi(m)). Protein M Flavivirus sequence alignments and helical wheel projections reveal a conserved distribution of charged residues. Moreover, when combined to the cell penetrating HIV-1 Tat peptide transduction domain (Tat-PTD), this sequence triggers a caspase-dependent cell death associated with DeltaPsi(m) loss and cytochrome c release. Mutational approaches coupled to functional screening on isolated mitochondria resulted in the selection of a protein M derived sequence containing nine residues with potent MMP-inducing properties on isolated mitochondria. A chimeric peptide composed of a Tat-PTD linked to the 9-mer entity triggers MMP and cell death. Finally, local administration of this chimeric peptide induces growth inhibition of xenograft prostate PC3 tumors in immuno-compromised mice, and significantly enhances animal survival. Together, these findings support the notion of using viral genomes as valuable sources to discover mitochondria-targeted sequences that may lead to the development of new anticancer compounds.


Subject(s)
Flavivirus/chemistry , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/metabolism , Peptides/pharmacology , Viral Proteins/chemistry , Xenograft Model Antitumor Assays , Amino Acid Motifs , Amino Acid Sequence , Animals , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Nude , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Swelling/drug effects , Molecular Sequence Data , Peptides/chemistry , Permeability/drug effects , Protein Structure, Tertiary , Survival Analysis , tat Gene Products, Human Immunodeficiency Virus/pharmacology
4.
Ann Neurol ; 64(6): 664-73, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19107989

ABSTRACT

OBJECTIVE: Prematurely born infants are at risk for development of neurocognitive impairment in later life. Oxygen treatment has been recently identified as a trigger of neuronal and oligodendrocyte apoptosis in the developing rodent brain. We investigated the role of the Fas death receptor pathway in oxygen-triggered developmental brain injury. METHODS: Six-day-old Wistar rats were exposed to 80% oxygen for various periods (2, 6, 12, 24, 48, and 72 hours), and mice deficient in either Fas (B6.MRL-Tnfrsf6(lpr)) or Fas ligand (B6Smn.C3-Fasl(gld)) and control mice (C57BL/6J) were exposed to 80% oxygen for 24 hours. Polymerase chain reaction, Western blotting, and caspase activity assays of thalamus and cortex tissue were performed. RESULTS: Fas and Fas ligand messenger RNA and protein were upregulated. Furthermore, hyperoxia resulted in induction of downstream signaling events of Fas, such as Fas-associated death domain (FADD), the long and short form of FADD-like interleukin-1beta-converting enzyme (FLICE) inhibitory protein (FLIP-L, FLIP-S), and cleavage of caspase-8 and caspase-3. Injection of a selective caspase-8 inhibitor (TRP801, 1mg/kg) at the beginning of hyperoxia blocked subsequent caspase-3 cleavage in this model. B6.MRL-Tnfrsf6(lpr) mice were protected against oxygen-mediated injury, confirming Fas involvement in hyperoxia-induced cell death. Mice deficient in Fas ligand did not differ from control animals in the amount of cell death. INTERPRETATION: We conclude that neonatal hyperoxia triggers Fas receptor and its downstream signaling events in a Fas ligand-independent fashion. Lack of functional Fas receptors and selective pharmacological inhibition of caspase-8 prevents activation of caspase-3 and provides significant neuroprotection.


Subject(s)
Brain Injuries/etiology , Brain Injuries/pathology , Fas Ligand Protein/physiology , Hyperoxia/etiology , Hyperoxia/pathology , Signal Transduction/physiology , Animals , Animals, Newborn , Mice , Mice, Inbred C57BL , Mice, Transgenic , Rats , Rats, Wistar
5.
J Neurochem ; 100(4): 1062-71, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17166174

ABSTRACT

Hypoxia-ischaemia in the developing brain results in brain injury with prominent features of apoptosis. In the present study, a third generation dipeptidyl broad-spectrum caspase inhibitor, quinoline-Val-Asp(Ome)-CH2-O-phenoxy (Q-VD-OPh), was tested in a model of unilateral focal ischaemia with reperfusion in 7-day-old rats. Q-VD-OPh (1 mg/kg, i.p.) reduced cell death, resulting in significant neuroprotection at 48 h of recovery (infarct volume of 12.6 +/- 2.8 vs. 24.3 +/- 2.2%, p = 0.006). The neuroprotective effects observed at 48 h post-ischaemia hold up at 21 days of survival time and attenuate neurological dysfunction. Analysis by gender revealed that females were strongly protected (6.7 +/- 3.3%, p = 0.006), in contrast to males in which there was no significant effect, when Q-VD-OPh was given after clip removal on the left common carotid artery. Immunoblot analysis demonstrated that Q-VD-OPh inhibits caspase 3 cleavage into its p17 active form and caspase 1 up-regulation and cleavage in vivo. Following ischaemia in P7 rats, males and females displayed different time course and pattern of cytochrome c release and active p17 caspase 3 during the first 24 h of recovery. In contrast, no significant difference was observed for caspase 1 expression between genders. These results indicate that ischaemia activates caspases shortly after reperfusion and that the sex of the animal may strongly influences apoptotic pathways in the pathogenesis of neonatal brain injury. The specificity, effectiveness, and reduced toxicity of Q-VD-OPh may determine the potential use of peptide-derived irreversible caspase inhibitors as promising therapeutics.


Subject(s)
Amino Acid Chloromethyl Ketones/therapeutic use , Enzyme Inhibitors/therapeutic use , Gender Identity , Quinolines/therapeutic use , Stroke/prevention & control , Animals , Animals, Newborn , Caspase Inhibitors , Cell Death/drug effects , Gene Expression Regulation/drug effects , Humans , In Situ Nick-End Labeling , Male , Neurologic Examination/methods , Rats , Statistics, Nonparametric , Stroke/pathology
6.
J Pharm Sci ; 91(8): 1765-75, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12115804

ABSTRACT

The multidrug-resistance (MR) status of camptothecin (CPT) was investigated in colon adenocarcinoma HT29 cells, leukemia K562, and breast carcinoma MCF7 cells expressing P-glycoprotein (Pgp) and/or MR-associated protein (MRP1). The concentration that induced 50% growth inhibition (IC(50)) against CPT was 0.14 and 0.20 microM in parental K562/WT and MCF7/WT cells, respectively. The drug resistant subline KH30 and MCF7/VP cells, which both overexpress MRP1, presented IC(50) values of 0.63 and 3.10 microM, respectively. The resulting resistance indexes were 3.80 and 12.50, respectively. However, in KH300 cells, a cell line that preferentially overexpresses Pgp, the IC(50) of CPT was 0.08 microM and thus did not exhibit resistance against CPT. In MCF7/DoX cells, preferentially overexpressing Pgp, but also a significant level of MRP1, the IC(50) of CPT was 0.64 microM and thus presented a resistance index of 3.26 against CPT. The cytotoxic effect of CPT was modulated in cells expressing MRP1 (MCF7/VP, HT29 cells) by the specific MRP1 modulators, probenecid and MK571. These results led us to consider CPT as a substrate for MRP1 and a potential modulator of MRP1 activity. To test this hypothesis, we examined the ability of nontoxic concentrations of CPT to sensitize MRP1-overexpressing cells to daunorubicin (DNR). In MCF7/VP and KH30 cells, nontoxic concentrations of CPT were able to enhance cytotoxicity of DNR and its nuclear accumulation. Sequential and simultaneous associations of CPT (100 nM) and DNR provided complete reversal of resistance, thus showing a synergistic effect in KH30 cells. However, simultaneous association (with 10 or 20 nM CPT) had an additive effect in MCF7/VP. These data suggest that CPT could be proposed as a candidate for the reversal of the MRP1 phenotype at clinically achievable concentrations.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Camptothecin/pharmacology , Daunorubicin/pharmacology , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Antibiotics, Antineoplastic/metabolism , Breast Neoplasms/pathology , Cell Line , Cell Nucleus/metabolism , Chemokines, CC/biosynthesis , Chemokines, CC/genetics , Daunorubicin/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic/drug effects , Humans , Leukemia/pathology , Microscopy, Confocal , Reverse Transcriptase Polymerase Chain Reaction , Tetrazolium Salts , Thiazoles , Tumor Cells, Cultured
7.
Breast Cancer Res Treat ; 73(2): 113-25, 2002 May.
Article in English | MEDLINE | ID: mdl-12088114

ABSTRACT

The multidrug-resistance (MDR) status of a novel camptothecin analogue, homocamptothecin (hCPT), was investigated in human colon adenocarcinoma HT29 cells, myelogenous leukemia K562 cells and breast carcinoma MCF7 cells. The cytotoxicity of hCPT was not sensitive to the MDR status in K562 cell lines. However, its cytotoxicity was altered by MRP1, but not Pgp, in naturally MRP1-expressing HT29 cells, and etoposide- and doxorubicin-resistant MCF7/VP and MCF7/DOX cells, respectively. These cells were sensitized to hCPT in presence of MK571, probenecid but not verapamil. These results led to consider hCPT as a substrate for MRP1 and a potential modulator of MRP1 activity. The relationship between the cytotoxic effect of anthracyclines and their nuclear localization had been previously demonstrated. We show that MRPI mediated the daunorubicin (DNR) efflux in MCF7/VP and MCF7/DOX cells. The combination of sub-toxic doses of hCPT with DNR resulted in the potentiation of DNR activity, well-correlated with an increase in its nuclear accumulation in MCF7/VP cells. Simultaneous pattern was shown to provide higher cytotoxic response than sequential one. In agreement, hCPT increased also the DNR nuclear accumulation in low MRP1-expressing MCF7/DOX cells. However, the enhancement of cytotoxicity in the DNR-hCPT combination was poorly correlated with the nuclear concentration of DNR in MCF7/DOX cells. In addition to the increase in DNR accumulation, the potentiation of DNR activity by hCPT in MCF7/DOX cells implied a synergistic mechanism between both drugs. These data suggest that the present topoisomerase I/II inhibitors combination may be of clinical interest to overcome MDR phenotype in DNR-treated breast cancer patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Camptothecin/analogs & derivatives , Carcinoma/drug therapy , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Multidrug Resistance-Associated Proteins/biosynthesis , Adenocarcinoma/drug therapy , Antibiotics, Antineoplastic/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/metabolism , Camptothecin/administration & dosage , Carcinoma/metabolism , Colonic Neoplasms/drug therapy , Daunorubicin/administration & dosage , Drug Synergism , Female , Flow Cytometry , Gene Expression Regulation, Neoplastic/drug effects , Humans , In Vitro Techniques , Leukemia, Myeloid/drug therapy , Microbial Sensitivity Tests , Multidrug Resistance-Associated Proteins/drug effects , Multidrug Resistance-Associated Proteins/genetics , Polymerase Chain Reaction , RNA, Neoplasm/analysis , Treatment Outcome , Tumor Cells, Cultured
8.
Int J Oncol ; 20(4): 855-63, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11894136

ABSTRACT

Topoisomerase I inhibitors of the camptothecin (CPT) family have emerged as potent clinical chemotherapeutic agents in first-line treatment of solid colorectal cancer and in second-line for 5-fluorouracil resistant patients. CPT and homocamptothecin (hCPT), derivative with enhanced lactone stability, induced growth inhibition in HT29 cells via p53-independent apoptosis. hCPT- and CPT-induced apoptosis was dependent on caspase-3 but not caspase-1. We report here substantial evidence that ceramide, resulting from de novo pathway or catabolism modulation, acted as a second messenger of these antitumor drugs in HT29 cells and leads to the activation of caspase-3. In addition, hCPT and CPT may favor ceramide signaling by disturbing sites of synthesis (Golgi) and trafficking of glucosylceramide from Golgi to lipid droplets. This work contributes to the understanding of the mechanism of action of CPTs, and suggests that inhibitors of glycosylation or activators of de novo metabolism could be of clinical interest in enhancing the effects of CPTs.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Ceramides/metabolism , Colonic Neoplasms/pathology , HT29 Cells/drug effects , trans-Golgi Network/metabolism , Camptothecin/adverse effects , Caspases/metabolism , Cell Survival/drug effects , Enzyme Inhibitors/pharmacology , Genes, p53 , HT29 Cells/cytology , HT29 Cells/metabolism , Humans , Lipids/chemistry , Mutation , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , Sphingomyelins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...