Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Hematol., Transfus. Cell Ther. (Impr.) ; 46(1): 49-57, Jan.-Mar. 2024. graf
Article in English | LILACS-Express | LILACS | ID: biblio-1557876

ABSTRACT

Abstract The evolutionary conserved link between coagulation and innate immunity is a biological process characterized by the thrombosis formation stimulus of immune cells and specific thrombosis-related molecules. In physiological settings, the relationship between the immune system and thrombosis facilitates the recognition of pathogens and damaged cells and inhibits pathogen proliferation. However, when deregulated, the interplay between hemostasis and innate immunity becomes a pathological process named immunothrombosis, which is at the basis of several infectious and inflammation-related thrombotic disorders, including coronavirus disease 2019 (COVID-19). In advanced stages, alterations in both coagulation and immune cell function due to extreme inflammation lead to an increase in blood coagulability, with high rates of thrombosis and mortality. Therefore, understanding underlying mechanisms in immunothrombosis has become decisive for the development of more efficient therapies to treat and prevent thrombosis in COVID-19 and in other thrombotic disorders. In this review, we outline the existing knowledge on the molecular and cellular processes involved in immunothrombosis, focusing on the role of neutrophil extracellular traps (NETs), platelets and the coagulation pathway. We also describe how the deregulation of hemostasis is associated with pathological conditions and can significantly aggravate a patient's condition, using COVID-19 as a clinical model.

2.
Hematol Transfus Cell Ther ; 46(1): 49-57, 2024.
Article in English | MEDLINE | ID: mdl-37451977

ABSTRACT

The evolutionary conserved link between coagulation and innate immunity is a biological process characterized by the thrombosis formation stimulus of immune cells and specific thrombosis-related molecules. In physiological settings, the relationship between the immune system and thrombosis facilitates the recognition of pathogens and damaged cells and inhibits pathogen proliferation. However, when deregulated, the interplay between hemostasis and innate immunity becomes a pathological process named immunothrombosis, which is at the basis of several infectious and inflammation-related thrombotic disorders, including coronavirus disease 2019 (COVID-19). In advanced stages, alterations in both coagulation and immune cell function due to extreme inflammation lead to an increase in blood coagulability, with high rates of thrombosis and mortality. Therefore, understanding underlying mechanisms in immunothrombosis has become decisive for the development of more efficient therapies to treat and prevent thrombosis in COVID-19 and in other thrombotic disorders. In this review, we outline the existing knowledge on the molecular and cellular processes involved in immunothrombosis, focusing on the role of neutrophil extracellular traps (NETs), platelets and the coagulation pathway. We also describe how the deregulation of hemostasis is associated with pathological conditions and can significantly aggravate a patient's condition, using COVID-19 as a clinical model.

4.
J Biol Chem ; 284(52): 36213-36222, 2009 Dec 25.
Article in English | MEDLINE | ID: mdl-19858212

ABSTRACT

In diet-induced obesity, hypothalamic and systemic inflammatory factors trigger intracellular mechanisms that lead to resistance to the main adipostatic hormones, leptin and insulin. Tumor necrosis factor-alpha (TNF-alpha) is one of the main inflammatory factors produced during this process and its mechanistic role as an inducer of leptin and insulin resistance has been widely investigated. Most of TNF-alpha inflammatory signals are delivered by TNF receptor 1 (R1); however, the role played by this receptor in the context of obesity-associated inflammation is not completely known. Here, we show that TNFR1 knock-out (TNFR1 KO) mice are protected from diet-induced obesity due to increased thermogenesis. Under standard rodent chow or a high-fat diet, TNFR1 KO gain significantly less body mass despite increased caloric intake. Visceral adiposity and mean adipocyte diameter are reduced and blood concentrations of insulin and leptin are lower. Protection from hypothalamic leptin resistance is evidenced by increased leptin-induced suppression of food intake and preserved activation of leptin signal transduction through JAK2, STAT3, and FOXO1. Under the high-fat diet, TNFR1 KO mice present a significantly increased expression of the thermogenesis-related neurotransmitter, TRH. Further evidence of increased thermogenesis includes increased O(2) consumption in respirometry measurements, increased expressions of UCP1 and UCP3 in brown adipose tissue and skeletal muscle, respectively, and increased O(2) consumption by isolated skeletal muscle fiber mitochondria. This demonstrates that TNF-alpha signaling through TNFR1 is an important mechanism involved in obesity-associated defective thermogenesis.


Subject(s)
Obesity/metabolism , Oxygen Consumption , Receptors, Tumor Necrosis Factor, Type I/metabolism , Thermogenesis , Tumor Necrosis Factor-alpha/metabolism , Abdominal Fat/metabolism , Adipose Tissue, Brown/metabolism , Animals , Diet/adverse effects , Dietary Fats/adverse effects , Forkhead Box Protein O1 , Forkhead Transcription Factors/metabolism , Inflammation/genetics , Inflammation/metabolism , Insulin/metabolism , Ion Channels/metabolism , Janus Kinase 2/metabolism , Leptin/metabolism , Mice , Mice, Knockout , Mitochondrial Proteins/metabolism , Muscle, Skeletal/metabolism , Nerve Tissue Proteins/metabolism , Obesity/genetics , Rats , Receptors, Tumor Necrosis Factor, Type I/genetics , STAT3 Transcription Factor/metabolism , Uncoupling Protein 1 , Uncoupling Protein 3
5.
Methods Enzymol ; 457: 395-404, 2009.
Article in English | MEDLINE | ID: mdl-19426880

ABSTRACT

Uncoupling protein 2 (UCP2) is a member of the uncoupling protein family. It is expressed in the inner mitochondrial membrane and plays a role in the control of free radical production, oxidative damage, insulin secretion, and fatty-acid peroxide exportation. Although UCP2 expression occurs in several tissues, some of its most remarkable functions are exerted in organs of difficult experimental access, such as the central nervous system, particularly the hypothalamus and the pancreatic islets. In addition, due to its low levels of expression in the mitochondrial membrane, studying UCP2 expression and function depends on specific- and well-established methods. This chapter describes methods for directly assessing UCP2 expression and function in different tissues. Purified mitochondria preparations are used for enhancing the capacity of detection of UCP2 protein or for evaluating the role of UCP2 in mitochondria respiration. Exposure of experimental animals to cold environment leads to increased UCP2 expression, while reduction of its expression can be achieved directly by targeting its mRNA with antisense oligonucleotides, or indirectly by targeting PGC-1alpha expression with antisense oligonucleotides.


Subject(s)
Gene Expression Regulation , Ion Channels/genetics , Ion Channels/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Animals , Antisense Elements (Genetics) , Cold Temperature , Hypothalamus/metabolism , Immunoblotting , Ion Channels/isolation & purification , Islets of Langerhans/metabolism , Male , Mice , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Proteins/isolation & purification , Oxygen/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Polymerase Chain Reaction , RNA-Binding Proteins/genetics , Rats , Rats, Wistar , Transcription Factors/genetics , Uncoupling Protein 2
6.
Clin Biochem ; 42(12): 1222-7, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19467225

ABSTRACT

OBJECTIVES: Experimental and in vitro evidences have established that reactive oxygen species (ROS) generated by vascular wall cells play a key role in atherogenesis. Here, we evaluated the rate of ROS generation by resting peripheral monocytes in naive hyperlipidemic subjects. DESIGN AND METHODS: Primary hypercholesterolemic, combined hyperlipidemic, and normolipidemic individuals were studied. ROS generation and the mitochondrial electrical transmembrane potential were estimated by flow cytometry. Plasma oxidized (ox) LDL levels and lipid profile were measured by ELISA and enzymatic colorimetric methods. RESULTS: Both hyperlipidemic groups presented significantly higher rates of monocyte ROS generation and elevated plasma levels of ox-LDL. Combined hyperlipidemic subjects presented increased levels of small dense LDL and insulin. Significant positive correlations between monocyte ROS generation and ox-LDL concentrations were found in pooled data. CONCLUSIONS: These data provide evidence that ROS production by circulating monocytes from hyperlipidemic subjects may contribute to the systemic oxidative stress and possibly to atherogenesis.


Subject(s)
Hyperlipidemias/blood , Lipoproteins, LDL/blood , Monocytes/metabolism , Reactive Oxygen Species/blood , Adult , Female , Humans , Lipoproteins/blood , Male , Middle Aged
7.
Metabolism ; 58(8): 1087-95, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19439330

ABSTRACT

Infiltrating macrophages play an important role in the production of inflammatory mediators by the adipose tissue of obese subjects. To reach the adipose tissue, peripheral monocytes are recruited by locally produced chemoattractants. However, little is known about the activation of monocytes in the peripheral blood of obese subjects. The objective of this study was to determine reactive oxygen species and endoplasmic reticulum stress as early markers of monocytic commitment with an inflammatory phenotype in the peripheral blood of nondiabetic obese patients. Patients were recruited from an academic general hospital; controls were voluntary students. Seven lean controls and 6 nondiabetic obese patients were included in the study. Monocytes were prepared from peripheral blood. Immunoblot, flow cytometry, and polymerase chain reaction were used to determine reactive oxygen species and endoplasmic reticulum stress. Increased reactive oxygen species and activation of endoplasmic reticulum stress were detected in the monocytes from obese patients. Reducing endoplasmic reticulum stress with a chemical chaperone reversed monocytic activation, as determined by the reduction of reactive oxygen species production. Thus, monocytes from nondiabetic obese patients are already committed with an inflammatory phenotype in peripheral blood; and reducing endoplasmic reticulum stress negatively modulates their activation.


Subject(s)
Endoplasmic Reticulum/metabolism , Inflammation/metabolism , Monocytes/metabolism , Obesity/blood , Oxidative Stress , Reactive Oxygen Species/blood , Adult , Calcium/metabolism , Catalase/metabolism , Cytosol/metabolism , DNA-Binding Proteins/metabolism , Electrophoresis, Polyacrylamide Gel , Female , Flow Cytometry , Humans , Immunoblotting , Immunoprecipitation , Inflammation/blood , Male , Monocytes/enzymology , Phenotype , Polymerase Chain Reaction , RNA Splicing , RNA, Messenger/metabolism , Regulatory Factor X Transcription Factors , Superoxide Dismutase/metabolism , Transcription Factors/metabolism
8.
FEBS Lett ; 582(20): 3103-10, 2008 Sep 03.
Article in English | MEDLINE | ID: mdl-18703058

ABSTRACT

Uncoupling protein 2 (UCP2) is highly expressed in the hypothalamus; however, little is known about the functions it exerts in this part of the brain. Here, we hypothesized that UCP2 protects hypothalamic cells from oxidative and pro-apoptotic damage generated by inflammatory stimuli. Intracerebroventricular injection of tumor necrosis factor alpha (TNF-alpha)-induced an increase of UCP2 expression in the hypothalamus, which was accompanied by increased expression of markers of oxidative stress and pro-apoptotic proteins. The inhibition of UCP2 expression by an antisense oligonucleotide enhanced the damaging effects of TNF-alpha. Conversely, increasing the hypothalamic expression of UCP2 by cold exposure reversed most of the effects of the cytokine. Thus, UCP2 acts as a protective factor against cellular damage induced by an inflammatory stimulus in the hypothalamus.


Subject(s)
Apoptosis , Hypothalamus/cytology , Hypothalamus/metabolism , Ion Channels/physiology , Mitochondrial Proteins/physiology , Animals , Cells, Cultured , Cold Temperature , Ion Channels/antagonists & inhibitors , Ion Channels/biosynthesis , Male , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/biosynthesis , Rats , Rats, Wistar , Tumor Necrosis Factor-alpha/pharmacology , Tumor Necrosis Factor-alpha/physiology , Uncoupling Protein 2
9.
An Acad Bras Cienc ; 80(1): 137-48, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18345382

ABSTRACT

The present study provides evidence that activated spleen lymphocytes from Walker 256 tumor bearing rats are more susceptible than controls to tert-butyl hydroperoxide (t-BOOH)-induced necrotic cell death in vitro. The iron chelator and antioxidant deferoxamine, the intracellular Ca2+ chelator BAPTA, the L-type Ca2+ channel antagonist nifedipine or the mitochondrial permeability transition inhibitor cyclosporin A, but not the calcineurin inhibitor FK-506, render control and activated lymphocytes equally resistant to the toxic effects of t-BOOH. Incubation of activated lymphocytes in the presence of t-BOOH resulted in a cyclosporin A-sensitive decrease in mitochondrial membrane potential. These results indicate that the higher cytosolic Ca2+ level in activated lymphocytes increases their susceptibility to oxidative stress-induced cell death in a mechanism involving the participation of mitochondrial permeability transition.


Subject(s)
Apoptosis , Lymphocyte Activation/drug effects , Lymphocytes/drug effects , Oxidative Stress , Spleen/pathology , tert-Butylhydroperoxide/pharmacology , Animals , Calcium/antagonists & inhibitors , Calcium/metabolism , Carcinoma 256, Walker , Chelating Agents/pharmacology , Deferoxamine/pharmacology , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Flow Cytometry , Male , Membrane Potentials/drug effects , Mitochondria/drug effects , Nifedipine/pharmacology , Oxidation-Reduction/drug effects , Rats , Rats, Wistar , Siderophores/pharmacology , Spleen/drug effects , Time Factors
10.
An. acad. bras. ciênc ; 80(1): 137-148, Mar. 2008. graf
Article in English | LILACS | ID: lil-477421

ABSTRACT

The present study provides evidence that activated spleen lymphocytes from Walker 256 tumor bearing rats are more susceptible than controls to tert-butyl hydroperoxide (t-BOOH)-induced necrotic cell death in vitro. The iron chelator and antioxidant deferoxamine, the intracellular Ca2+ chelator BAPTA, the L-type Ca2+ channel antagonist nifedipine or the mitochondrial permeability transition inhibitor cyclosporin A, but not the calcineurin inhibitor FK-506, render control and activated lymphocytes equally resistant to the toxic effects of t-BOOH. Incubation of activated lymphocytes in the presence of t-BOOH resulted in a cyclosporin A-sensitive decrease in mitochondrial membrane potential. These results indicate that the higher cytosolic Ca2+ level in activated lymphocytes increases their susceptibility to oxidative stress-induced cell death in a mechanism involving the participation of mitochondrial permeability transition.


O presente estudo demonstra que linfócitos ativados de baço de ratos portadores do tumor de Walker 256 são mais susceptíveis à morte celular necrótica induzida por tert-butil hidroperóxido (t-BOOH) in vitro quando comparados aos controles. O quelante de ferro e antioxidante deferoxamina, o quelante intracelular de Ca2+ BAPTA, o antagonista de canal de Ca2+ nifedipina ou o inibidor da transição de permeabilidade mitocondrial ciclosporina-A, mas não o inibidor de calcineurina FK-506, inibiram de maneira similar a morte celular induzida por t-BOOH em linfócitos ativados e controles. Os linfócitos ativados apresentaram redução do potencial de membrana mitocondrial induzida por t-BOOH num mecanismo sensível a ciclosporina-A. Nossos resultados indicam que o aumento da concentração de Ca2+ citosólico em linfócitos ativados aumenta a susceptibilidade dos mesmos à morte celular induzida por estresse oxidativo, num mecanismo envolvendo a participação do poro de transição de permeabilidade mitocondrial.


Subject(s)
Animals , Male , Rats , Apoptosis , Lymphocyte Activation/drug effects , Lymphocytes/drug effects , Oxidative Stress , Spleen/pathology , tert-Butylhydroperoxide/pharmacology , Calcium/antagonists & inhibitors , Calcium/metabolism , Chelating Agents/pharmacology , Deferoxamine/pharmacology , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Flow Cytometry , Membrane Potentials/drug effects , Mitochondria/drug effects , Nifedipine/pharmacology , Oxidation-Reduction/drug effects , Rats, Wistar , Siderophores/pharmacology , Spleen/drug effects , Time Factors
11.
J Bioenerg Biomembr ; 39(2): 186-94, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17431754

ABSTRACT

It has been previously shown that Walker 256 tumor cells express a high content of the anti-apoptotic protein Bcl-2 which protects mitochondria against the damaging effects of Ca(2+). In the present study, we analyze H(2)O(2)-induced apoptotic death in two different types of tumor cells: Walker 256 and SCC-25. Treatment with H(2)O(2) (4mM) increased reactive oxygen species generation and the concentration of cytosolic free Ca(2+). These alterations preceded apoptosis in both cell lines. In Walker cells, which show a high Bcl-2/Bax ratio, apoptosis was dependent on calcineurin activation and independent of changes in mitochondrial membrane potential (DeltaPsi(m)), as well as cytochrome c release. In contrast, in SCC-25 cells, which show a lower Bcl-2/Bax ratio, apoptosis was preceded by a decrease in DeltaPsi(m), mitochondrial permeability transition, and cytochrome c release. Caspase-3 activation occurred in both cell lines. The data suggest that although the high Bcl-2/Bax ratio protected the mitochondria of Walker cells from oxidative stress, it was not sufficient to prevent apoptosis through calcineurin pathways.


Subject(s)
Apoptosis/physiology , Calcineurin/metabolism , Hydrogen Peroxide/pharmacology , Mitochondria/physiology , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Apoptosis/drug effects , Carcinoma 256, Walker , Caspase 3/metabolism , Cell Line, Tumor , Cytochromes c/metabolism , Humans , Male , Membrane Potential, Mitochondrial , Mitochondria/drug effects , Oxidative Stress/physiology , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Signal Transduction , bcl-2-Associated X Protein/metabolism
12.
Eur J Pharmacol ; 549(1-3): 179-84, 2006 Nov 07.
Article in English | MEDLINE | ID: mdl-16996495

ABSTRACT

Recently, we demonstrated that verapamil, an L-type Ca2+ channel blocker, inhibits the activation of splenic lymphocytes during Walker 256 ascitic tumor development in adult rats. In the present study we have analyzed the changes in spleen size, splenic lymphocyte proliferation, white pulp organization and relative size as well as food intake, and levels of blood haemoglobin in Walker 256 tumor bearing rats. These rats displayed a spleen enlargement associated with a significant increase in white pulp area and TCD8+ lymphocyte proliferation. Levels of interferon-gamma, but not of interleukin-10, were elevated in tumor bearing rats, indicating a Th1-type immune response. These manifestations were accompanied by reduced food intake and anaemia. Treatment of tumor bearing rats with verapamil avoided spleen enlargement and increased expression of cytokines, as well as the splenic TCD8+ lymphocyte proliferation. In addition, verapamil treatment promoted an exacerbation of the anorexia and anaemia caused by Walker tumor development. No such effect was observed in control rats treated with verapamil. Taken together, these findings suggest that verapamil inhibits the immune response to cancer, resulting in an increase of the systemic effects induced by Walker 256 tumor.


Subject(s)
Calcium Channels/physiology , Cell Proliferation/drug effects , Neoplasms, Experimental/prevention & control , T-Lymphocytes/drug effects , Verapamil/pharmacology , Animals , Blotting, Western , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Calcium Channel Blockers/pharmacology , Cell Line, Tumor , Eating/drug effects , Hemoglobins/metabolism , Interferon-gamma/metabolism , Interleukin-10/metabolism , Male , Neoplasm Transplantation , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Organ Size/drug effects , Rats , Rats, Wistar , Spleen/drug effects , Spleen/metabolism , Spleen/pathology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Th1 Cells/drug effects , Th1 Cells/metabolism , Th1 Cells/pathology , Time Factors
13.
J Bioenerg Biomembr ; 38(1): 1-10, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16779655

ABSTRACT

This study demonstrates that Ca2+ stimulates mitochondrial energy metabolism during spleen lymphocyte activation in response to the ascitic Walker 256 tumor in rats. Intracellular Ca2+ concentrations, phosphorylated protein kinase C (pPKC) levels, Bcl-2 protein contents, interleukin-2 (IL-2) levels, mitochondrial uncoupling protein-2 (UCP-2) contents and reactive oxygen species (ROS) were significantly elevated in these activated lymphocytes. Mitochondria of activated lymphocytes exhibited high free Ca2+ concentrations in the matrix and enhanced oligomycin-sensitive oxygen consumption, indicating an increased rate of oxidative phosphorylation. The production of ROS was largely decreased by diphenylene iodinium in the activated lymphocytes, suggesting that NADPH oxidase is the prevalent source of these species. Accumulation of UCP-2 and the anti-apoptotic protein Bcl-2 is probably important to prevent mitochondrial dysfunction and cell death elicited by the sustained high levels of intracellular Ca2+ and ROS and may explain the observed higher resistance from activated lymphocytes against the opening of the mitochondrial membrane permeability pore (MPT). All these changes were blocked by pretreatment of the rats with verapamil, an L-type Ca2+ channel antagonist. These data demonstrate a central role of Ca2+ in the control of mitochondrial bioenergetics in spleen lymphocytes during the immune response to cancer.


Subject(s)
Calcium/physiology , Carcinoma 256, Walker/immunology , Lymphocytes/immunology , Mitochondria/physiology , Animals , Cell Death , Energy Metabolism , In Vitro Techniques , Interleukin-2/metabolism , Ion Channels , Lymphocyte Activation , Male , Membrane Transport Proteins/metabolism , Mitochondria/immunology , Mitochondrial Membranes/physiology , Mitochondrial Proteins/metabolism , NADPH Oxidases/antagonists & inhibitors , Phosphorylation , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Spleen/cytology , Uncoupling Protein 2
14.
Toxicology ; 219(1-3): 124-32, 2006 Feb 15.
Article in English | MEDLINE | ID: mdl-16343726

ABSTRACT

Statins (3-hydroxy-3-methylglutaryl-CoA reductase inhibitors) are used in the treatment of hypercholesterolemic patients to reduce risk of cardiovascular diseases because of their cholesterol lowering action. Other lipid independent protective actions of statins have been reported. However, some adverse side effects have, also, been described. We report, here, that liver mitochondria isolated from hypercholesterolemic LDL receptor knockout mice treated during 15 days with therapeutic doses (100 mg/kg, p.o.) of lovastatin presented a higher susceptibility to develop membrane permeability transition (MPT). In experiments in vitro, lovastatin-induced MPT in a dose-dependent manner (10-80 microM) by a mechanism sensitive to cyclosporin A (cyclophilin sequestrant), dithiothreitol (reducing agent), adenine nucleotide carrier inhibitor (ADP), catalase (H2O2 reductant) and EGTA (calcium chelator). In agreement with the inhibition of the mitochondrial swelling by dithiothreitol, lovastatin, also, decreased the content of total mitochondrial membrane protein thiol groups. Simvastatin had similar effects on mitochondria; however, pravastatin, a hydrophilic statin, had a weaker effect in inducing MPT. In conclusion, statins can act directly on mitochondria either in vivo or in vitro inducing permeability transition, which is a process involved in cell death.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Mitochondria, Liver/drug effects , Mitochondria, Muscle/drug effects , Adenosine Diphosphate/pharmacology , Animals , Catalase/pharmacology , Chelating Agents/pharmacology , Cholesterol/blood , Dithiothreitol/pharmacology , Egtazic Acid/pharmacology , Electrophysiology , Hindlimb/metabolism , In Vitro Techniques , Lovastatin/pharmacology , Mice , Mice, Knockout , Mitochondrial Swelling/drug effects , Permeability/drug effects , Phenazines , Proteins/metabolism , Receptors, LDL/genetics , Sulfhydryl Compounds/metabolism , Sulfhydryl Reagents/pharmacology
15.
Biochem Pharmacol ; 68(11): 2197-206, 2004 Dec 01.
Article in English | MEDLINE | ID: mdl-15498510

ABSTRACT

The participation of mitochondria in the mechanism of tumor cell death induced by non-steroid anti-inflammatory drugs is uncertain. Here we show that ibuprofen induces death of Walker 256 tumor cells independently on mitochondrial depolarization as estimated by flow cytometry using DioC(6)(3). Oligomycin increased mitochondrial transmembrane potential in both ibuprofen-treated and non-treated cells, indicating that ATP synthesis was sustained during cell death. Cyclosporin A, but not bongkrekic acid, both mitochondrial permeability transition inhibitors, increased the percentage of cell death in the presence of ibuprofen. FK506, a calcineurin inhibitor like cyclosporin A, also increased ibuprofen-induced cell death. Moreover, we showed that cytochrome c was released during ibuprofen-induced cell death. In conclusion, death of Walker 256 tumor cells induced by ibuprofen does not impair mitochondrial function, involves cytochrome c release and is accompanied by a rescue pathway via calcineurin activation.


Subject(s)
Calcineurin Inhibitors , Cell Death/drug effects , Cytochromes c/metabolism , Ibuprofen/pharmacology , Mitochondria/drug effects , Adenosine Triphosphate/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cyclosporine/pharmacology , Hydrolysis , Ion Channels/metabolism , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mitochondria/enzymology , Mitochondria/physiology , Mitochondrial Membrane Transport Proteins , Mitochondrial Permeability Transition Pore , Rats , Respiration , Tacrolimus/pharmacology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...