Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
Add more filters

Publication year range
1.
Exp Eye Res ; 116: 359-65, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24416768

ABSTRACT

The objective of this study was to elucidate possible reasons for the remarkable resistance of human retinal pigment epithelial (RPE) cells to oxidative stress. Much oxidative damage is due to hydrogen peroxide meeting redox-active iron in the acidic and reducing lysosomal environment, resulting in the production of toxic hydroxyl radicals that may oxidize intralysosomal content, leading to lipofuscin (LF) formation or, if more extensive, to permeabilization of lysosomal membranes. Formation of LF is a risk factor for age-related macular degeneration (AMD) and known to jeopardize normal autophagic rejuvenation of vital cellular biomolecules. Lysosomal membrane permeabilization causes release of lysosomal content (redox-active iron, lytic enzymes), which may then cause cell death. Total cellular and lysosomal low-mass iron of cultured, immortalized human RPE (ARPE-19) cells was compared to that of another professional scavenger cell line, J774, using atomic absorption spectroscopy and the cytochemical sulfide-silver method (SSM). It was found that both cell lines contained comparable levels of total as well as intralysosomal iron, suggesting that the latter is mainly kept in a non-redox-active state in ARPE-19 cells. Basal levels and capacity for upregulation of the iron-binding proteins ferritin, metallothionein and heat shock protein 70 were tested in both cell lines using immunoblotting. Compared to J774 cells, ARPE-19 cells were found to contain very high basal levels of all these proteins, which could be even further upregulated following appropriate stimulation. These findings suggest that a high basal expression of iron-binding stress proteins, which during their normal autophagic turnover in lysosomes may temporarily bind iron prior to their degradation, could contribute to the unusual oxidative stress-resistance of ARPE-19 cells. A high steady state influx of such proteins into lysosomes would keep the level of lysosomal redox-active iron permanently low. This, in turn, should delay intralysosomal accumulation of LF in RPE cells, which is known to reduce autophagic turnover as well as uptake and degradation of worn out photoreceptor tips. This may explain why severe LF accumulation and AMD normally do not develop until fairly late in life, in spite of RPE cells being continuously exposed to high levels of oxygen and light, as well as large amounts of lipid-rich material.


Subject(s)
Autophagy/physiology , Iron-Binding Proteins/metabolism , Macular Degeneration/metabolism , Oxidative Stress/physiology , Retinal Pigment Epithelium/metabolism , Blotting, Western , Cell Line , Humans , Macular Degeneration/pathology , Retinal Pigment Epithelium/pathology
2.
J Lipid Res ; 53(6): 1134-43, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22454477

ABSTRACT

Normally, cell proliferation and death are carefully balanced in higher eukaryotes, but one of the most important regulatory mechanisms, apoptosis, is upset in many malignancies, including hepatocellular-derived ones. Therefore, reinforcing cell death often is mandatory in anticancer therapy. We previously reported that a combination of tumor necrosis factor-α (TNF) and cycloheximide (CHX) efficiently kill HTC cells, a rat hepatoma line, in an apoptosis-like mode. Death is actively mediated by the lysosomal compartment, although lysosomal ceramide was previously shown not to be directly implicated in this process. In the present study, we show that TNF/CHX increase lysosomal ceramide that is subsequently converted into sphingosine. Although ceramide accumulation does not significantly alter the acidic compartment, the sphingosine therein generated causes lysosomal membrane permeabilization (LMP) followed by relocation of lysosomal cathepsins to the cytoplasm. TNF/CHX-induced LMP is effectively abrogated by siRNAs targeting acid sphingomyelinase or acid ceramidase, which prevent both LMP and death induced by TNF/CHX. Taken together, our results demonstrate that lysosomal accumulation of ceramide is not detrimental per se, whereas its degradation product sphingosine, which has the capacity to induce LMP, appears responsible for the observed apoptotic-like death.


Subject(s)
Carcinoma, Hepatocellular/pathology , Intracellular Membranes/drug effects , Intracellular Membranes/metabolism , Lysosomes/metabolism , Sphingosine/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cell Death/drug effects , Cell Line, Tumor , Ceramidases/deficiency , Ceramidases/genetics , Ceramides/biosynthesis , Ceramides/metabolism , Cycloheximide/pharmacology , Gene Silencing , Permeability/drug effects , RNA, Small Interfering/genetics , Rats , Receptors, Tumor Necrosis Factor/metabolism , Sphingomyelin Phosphodiesterase/deficiency , Sphingomyelin Phosphodiesterase/genetics , Sphingosine/biosynthesis , Sphingosine/metabolism
3.
Am J Physiol Regul Integr Comp Physiol ; 302(7): R805-14, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22277937

ABSTRACT

The main objective of this study was to investigate the activity of polydatin on mitochondrial dysfunction and lysosomal stability of arteriolar smooth muscle cells (ASMCs) in severe shock. The experimental animals (rats) were divided into five groups: control, hemorrhagic shock, shock + CsA, shock + Res, and shock + PD (exposed to cyclosporin A, resveratrol, or polydatin following induction of hemorrhagic shock, respectively). The calcein-Co(2+) technique revealed opening of ASMC mitochondrial permeability transition pores (mPTP) after shock with resulting mitochondrial swelling, decreased mitochondrial membrane potential (ΔΨm), and reduced intracellular ATP levels. These alterations were all inhibited by exposure to PD, which was significantly more effective than CsA and Res. PD also preserved lysosomal stability, suppressed activation of K(ATP) channels, ASMC hyperpolarization, and reduced vasoresponsiveness to norepinephrine that normally follows severe shock. The results demonstrate that exposure to PD after initiation of severe shock effectively preserves ASMC mitochondrial integrity and has a significant therapeutic effect in severe shock. The effects may partially result from lysosomal stabilization against shock-induced oxidative stress and depressed relocation of hydrolytic enzymes and redox-active lysosomal iron that, in turn, may induce mPTP opening.


Subject(s)
Cytoprotection , Glucosides/administration & dosage , Lysosomes/drug effects , Mitochondria, Muscle/drug effects , Mitochondrial Diseases/prevention & control , Myocytes, Smooth Muscle/drug effects , Shock, Hemorrhagic/complications , Stilbenes/administration & dosage , Adenosine Triphosphate/analysis , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cells, Cultured , Cyclosporine/pharmacology , KATP Channels/drug effects , Membrane Potential, Mitochondrial/drug effects , Mitochondrial Diseases/etiology , Mitochondrial Membrane Transport Proteins/drug effects , Mitochondrial Permeability Transition Pore , Norepinephrine/pharmacology , Rats , Resveratrol , Stilbenes/pharmacology
4.
Nutr Cancer ; 64(3): 464-72, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22420317

ABSTRACT

In a Wistar rat model, prolonged supplementation of mustard seed (MS) to the diet significantly ameliorates the induction of colorectal carcinomas by 1,2-dimethylhydrazine (DMH). The expression of the splenocyte major histocompatibility complex class I (MHCI) was found significantly enhanced, whereas that of the major histocompatibility complex class II (MHCII) was significantly decreased. Compared to that of control animals, the proportion of spleenic B- and dendritic cells (DC) was amplified in the MS group. The expressions of MHCI, as well as that of MHCII, were increased in DC cells; whereas in B cells, MHCI expression was augmented but that of MHCII moderately decreased. The percentages of CD8+CD28+ and CD4+CD28+ cells were increased in the MS group, while the CD4+CD25+Foxp3+ subset was depressed. Plasma analysis showed that DMH-exposure induced amplified amounts of interleukin (IL)-4, IL-5, IL-10, and transforming growth factor-beta, whereas MS feeding counteracted this effect but enhanced IL-2, IL12p70, IL21, TNF-alpha, and interferon-gamma. In the SW480 colon adenocarcinoma cell-line, the cytotoxicity of spleenic T-cells from MS-fed animals was significantly increased. In the DMH-exposed rats, the expression of perforin in the spleenic T-cells was dramatically decreased, whereas MS abolished this depression. In summary, dietary MS suppresses DMH-induced immuno-imbalance as well as colon carcinogenesis in rats.


Subject(s)
1,2-Dimethylhydrazine/toxicity , Colonic Neoplasms/pathology , Plant Extracts/pharmacology , Seeds/chemistry , Sinapis/chemistry , Animals , Apoptosis/drug effects , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Colon/drug effects , Colon/metabolism , Colon/pathology , Colonic Neoplasms/chemically induced , Dendritic Cells/metabolism , Diet , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Interferon-gamma/blood , Interleukin-10/blood , Interleukin-2/blood , Interleukin-5/blood , Male , Rats , Rats, Wistar , Spices/analysis , Tumor Necrosis Factor-alpha/blood , Up-Regulation
5.
Cell Mol Life Sci ; 68(24): 3963-9, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22015613

ABSTRACT

Vitamin-B(12) is a generic term for corrinoid compounds that exhibit the biological activity of cyanocobalamin and are collectively referred to as cobalamins. Methylcobalamin and 5-deoxyadenosylcobalamin are the active cobalamins in human metabolism. Cobalamin plays a crucial role in the maintenance of homocysteine and methylmalonyl-CoA homeostasis and is required for erythrocyte formation and DNA synthesis. Data from human and animal studies indicate that cobalamin deficiency impairs neuronal function; a process that is thought to contribute to age-related cognitive decline and dementia. Cobalamin deficiency also results in dysfunction of the peripheral nervous system; among other disorders. Although there is a detailed understanding of the biochemical pathways that are perturbed in cobalamin deficiency, the mechanisms underlying age-related dyshomeostasis in such pathways remain to be addressed. Because cobalamin utilization is dependent on its efficient transit through lysosomes, and mounting evidence indicates that lysosomal function deteriorates in aging long-lived post-mitotic cells such as neurons, in the present article we review published data that supports the proposition that impaired lysosomal processing of cobalamin may play a significant role in age-related (neuro) degenerative diseases.


Subject(s)
Lysosomes/metabolism , Neurodegenerative Diseases/metabolism , Vitamin B 12/metabolism , Age Factors , Biological Transport , Homeostasis , Humans , Models, Biological , Neurodegenerative Diseases/pathology , Vitamin B 12/chemistry
6.
Biochem J ; 435(1): 207-16, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21210766

ABSTRACT

Cellular deposits of oxidized and aggregated proteins are hallmarks of a variety of age-related disorders, but whether such proteins contribute to pathology is not well understood. We previously reported that oxidized proteins form lipofuscin/ceroid-like bodies with a lysosomal-type distribution and up-regulate the transcription and translation of proteolytic lysosomal enzymes in cultured J774 mouse macrophages. Given the recently identified role of lysosomes in the induction of apoptosis, we have extended our studies to explore a role for oxidized proteins in apoptosis. Oxidized proteins were biosynthetically generated in situ by substituting oxidized analogues for parent amino acids. Apoptosis was measured with Annexin-V/PI (propidium iodide), TUNEL (terminal deoxynucleotidyltransferase-mediated dUTP nick-end labelling), MMP (mitochondrial membrane permeabilization), caspase activation and cytochrome c release, and related to lysosomal membrane permeabilization. Synthesized proteins containing the tyrosine oxidation product L-DOPA (L-3,4-dihydroxyphenylalanine) were more potent inducers of apoptosis than proteins containing the phenylalanine oxidation product o-tyrosine. Apoptosis was dependent upon incorporation of oxidized residues, as indicated by complete abrogation in cultures incubated with the non-incorporation control D-DOPA (D-3,4-dihydroxyphenylalanine) or when incorporation was competed out by parent amino acids. The findings of the present study suggest that certain oxidized proteins could play an active role in the progression of age-related disorders by contributing to LMP (lysosomal membrane permeabilization)-initiated apoptosis and may have important implications for the long-term use of L-DOPA as a therapeutic agent in Parkinson's disease.


Subject(s)
Apoptosis , Levodopa/adverse effects , Levodopa/metabolism , Monocytes/metabolism , Protein Biosynthesis , Tyrosine/adverse effects , Tyrosine/metabolism , Aging , Annexin A5/metabolism , Caspase 3/metabolism , Cell Line , Ceroid/adverse effects , DNA Fragmentation , Enzyme Activation , Humans , Intracellular Membranes , Lipofuscin/adverse effects , Lysosomes , Membrane Potential, Mitochondrial , Mitochondrial Membranes , Oxidation-Reduction , Permeability
7.
Biochem J ; 432(2): 295-301, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-20846118

ABSTRACT

Ionizing radiation causes DNA damage and consequent apoptosis, mainly due to the production of hydroxyl radicals (HO•) that follows radiolytic splitting of water. However, superoxide (O2•-) and H2O2 also form and induce oxidative stress with resulting LMP (lysosomal membrane permeabilization) arising from iron-catalysed oxidative events. The latter will contribute significantly to radiation-induced cell death and its degree largely depends on the quantities of lysosomal redox-active iron present as a consequence of autophagy and endocytosis of iron-rich compounds. Therefore radiation sensitivity might be depressed by lysosome-targeted iron chelators. In the present study, we have shown that cells in culture are significantly protected from ionizing radiation damage if initially exposed to the lipophilic iron chelator SIH (salicylaldehyde isonicotinoyl hydrazone), and that this effect is based on SIH-dependent lysosomal stabilization against oxidative stress. According to its dose-response-modifying effect, SIH is a most powerful radioprotector and a promising candidate for clinical application, mainly to reduce the radiation sensitivity of normal tissue. We propose, as an example, that inhalation of SIH before each irradiation session by patients undergoing treatment for lung malignancies would protect normally aerated lung tissue against life-threatening pulmonary fibrosis, whereas the sensitivity of malignant lung tumours, which usually are non-aerated, will not be affected by inhaled SIH.


Subject(s)
Iron Chelating Agents/metabolism , Iron/metabolism , Lysosomes/metabolism , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Gamma Rays , HeLa Cells/cytology , HeLa Cells/drug effects , HeLa Cells/metabolism , HeLa Cells/radiation effects , Humans , Hydrogen Peroxide/pharmacology , Iron Chelating Agents/pharmacology , Lysosomes/drug effects , Lysosomes/physiology , Lysosomes/radiation effects , Oxidative Stress
8.
Biochem J ; 428(2): 183-90, 2010 May 13.
Article in English | MEDLINE | ID: mdl-20331437

ABSTRACT

H(2)DCF-DA (dihydrodichlorofluorescein diacetate) is widely used to evaluate 'cellular oxidative stress'. After passing through the plasma membrane, this lipophilic and non-fluorescent compound is de-esterified to a hydrophilic alcohol [H(2)DCF (dihydrodichlorofluorescein)] that may be oxidized to fluorescent DCF (2',7'-dichlorofluorescein) by a process usually considered to involve ROS (reactive oxygen species). It is, however, not always recognized that, being a hydrophilic molecule, H(2)DCF does not cross membranes, except for the outer fenestrated mitochondrial ones. It is also not generally realized that oxidation of H(2)DCF is dependent either on Fenton-type reactions or on unspecific enzymatic oxidation by cytochrome c, for neither superoxide, nor H(2)O(2), directly oxidizes H(2)DCF. Consequently, oxidation of H(2)DCF requires the presence of either cytochrome c or of both redox-active transition metals and H(2)O(2). Redox-active metals exist mainly within lysosomes, whereas cytochrome c resides bound to the outer side of the inner mitochondrial membrane. Following exposure to H(2)DCF-DA, weak mitochondrial fluorescence was found in both the oxidation-resistant ARPE-19 cells and the much more sensitive J774 cells. This fluorescence was only marginally enhanced following short exposure to H(2)O(2), showing that by itself it is unable to oxidize H(2)DCF. Cells that were either exposed to the lysosomotropic detergent MSDH (O-methylserine dodecylamide hydrochloride), exposed to prolonged oxidative stress, or spontaneously apoptotic showed lysosomal permeabilization and strong DCF-induced fluorescence. The results suggest that DCF-dependent fluorescence largely reflects relocation to the cytosol of lysosomal iron and/or mitochondrial cytochrome c.


Subject(s)
Fluoresceins/metabolism , Oxidative Stress/physiology , Animals , Apoptosis/physiology , Cell Line , Cytochromes c/metabolism , Cytosol/metabolism , Fluorescence , Humans , Hydrogen Peroxide/metabolism , Lysosomes/metabolism , Mice , Mitochondria/metabolism , Organometallic Compounds/metabolism , Oxidation-Reduction , Reactive Oxygen Species/metabolism
9.
Biochim Biophys Acta ; 1793(7): 1182-90, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19328214

ABSTRACT

We previously showed that, in the rat hepatoma cell line HTC, TNF brings about a non-caspase-dependent, apoptosis-like process requiring NADPH oxidase activity, an iron-mediated pro-oxidant status, and a functional acidic vacuolar compartment. This process may thus involve mechanisms such as autophagy or relocation of lysosomal enzymes, perhaps secondary to the formation of ceramide by acidic sphingomyelinase. Here we investigated whether ceramide formation contributes to the apoptogenic process. HTC cells were found to be sensitive to exogenous ceramide and significantly protected against TNF by desipramine, an inhibitor of lysosomal acid sphingomyelinase. However, Bcl-2 transfection and Bcl-x(L) upregulation by dexamethasone significantly diminished the apoptogenic effect of ceramide but not that of TNF, suggesting that ceramide is not directly involved in TNF toxicity. Moreover, Bcl-x(L) silencing precluded dexamethasone-induced protection against ceramide and, by itself, induced massive death, demonstrating the strict dependence of HTC cells on Bcl-x(L) for survival also under standard culture conditions.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Ceramides/toxicity , Liver Neoplasms, Experimental/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/toxicity , Animals , Apoptosis/drug effects , Carcinoma, Hepatocellular/pathology , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Liver Neoplasms, Experimental/pathology , Lysosomes , Membrane Potential, Mitochondrial/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Interference , Rats , Tumor Cells, Cultured , bcl-X Protein/antagonists & inhibitors , bcl-X Protein/genetics , bcl-X Protein/metabolism
10.
Clin Cancer Res ; 15(5): 1593-600, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19223492

ABSTRACT

PURPOSE: Vitamin E analogues are potent novel anticancer drugs. The purpose of this study was to elucidate the cellular target by which these agents, represented by alpha-tocopoheryl succinate (alpha-TOS), suppress tumors in vivo, with the focus on the mitochondrial complex II (CII). EXPERIMENTAL DESIGN: Chinese hamster lung fibroblasts with functional, dysfunctional, and reconstituted CII were transformed using H-Ras. The cells were then used to form xenografts in immunocompromized mice, and response of the cells and the tumors to alpha-TOS was studied. RESULTS: The CII-functional and CII-reconstituted cells, unlike their CII-dysfunctional counterparts, responded to alpha-TOS by reactive oxygen species generation and apoptosis execution. Tumors derived from these cell lines reciprocated their responses to alpha-TOS. Thus, growth of CII-functional and CII-reconstituted tumors was strongly suppressed by the agent, and this was accompanied by high level of apoptosis induction in the tumor cells. On the other hand, alpha-TOS did not inhibit the CII-dysfunctional tumors. CONCLUSIONS: We document in this report a novel paradigm, according to which the mitochondrial CII, which rarely mutates in human neoplasias, is a plausible target for anticancer drugs from the group of vitamin E analogues, providing support for their testing in clinical trials.


Subject(s)
Antioxidants/therapeutic use , Electron Transport Complex II/metabolism , Lung Neoplasms/prevention & control , Mitochondria/metabolism , alpha-Tocopherol/therapeutic use , Animals , Apoptosis/drug effects , Blotting, Western , Cell Transformation, Neoplastic , Cells, Cultured , Colony-Forming Units Assay , Cricetinae , Cricetulus , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Green Fluorescent Proteins/genetics , Lung/cytology , Lung/drug effects , Lung/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Membrane Proteins/physiology , Mice , Mice, Inbred BALB C , Mice, Nude , Oxygen Consumption , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction
11.
Biochim Biophys Acta ; 1780(11): 1291-303, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18255041

ABSTRACT

The lysosomal compartment consists of numerous acidic vesicles (pH approximately 4-5) that constantly fuse and divide. It receives a large number of hydrolases from the trans-Golgi network, while their substrates arrive from both the cell's outside (heterophagy) and inside (autophagy). Many macromolecules under degradation inside lysosomes contain iron that, when released in labile form, makes lysosomes sensitive to oxidative stress. The magnitude of generated lysosomal destabilization determines if reparative autophagy, apoptosis, or necrosis will follow. Apart from being an essential turnover process, autophagy is also a mechanism for cells to repair inflicted damage, and to survive temporary starvation. The inevitable diffusion of hydrogen peroxide into iron-rich lysosomes causes the slow oxidative formation of lipofuscin in long-lived postmitotic cells, where it finally occupies a substantial part of the volume of the lysosomal compartment. This seems to result in a misdirection of lysosomal enzymes away from autophagosomes, resulting in depressed autophagy and the accumulation of malfunctioning mitochondria and proteins with consequent cellular dysfunction. This scenario might put aging into the category of autophagy disorders.


Subject(s)
Aging/metabolism , Apoptosis , Lysosomes/metabolism , Oxidative Stress , Animals , Humans , Iron/metabolism , Mitosis
12.
IUBMB Life ; 61(5): 522-7, 2009 May.
Article in English | MEDLINE | ID: mdl-19391165

ABSTRACT

Elevated levels of oxidized proteins are reported in diseased tissue from age-related pathologies such as atherosclerosis, neurodegenerative disorders, and cataract. Unlike the precise mechanisms that exist for the repair of nucleic acids, lipids, and carbohydrates, the primary pathway for the repair of oxidized proteins is complete catabolism to their constitutive amino acids. This process can be inefficient as is evidenced by their accumulation. It is generally considered that damaged proteins are degraded by the proteasome; however, this is only true for mildly oxidized proteins, because substrates must be unfolded to enter the narrow catalytic core. Rather, evidence suggests that moderately or heavily oxidized proteins are endocytosed and enter the endosomal/lysosomal system, indicating co-operation between the proteasomes and the lysosomes. Heavily modified substrates are incompletely degraded and accumulate within the lysosomal compartments resulting in the formation of lipofuscin-like, autofluorescent aggregates. Accumulation eventually results in impaired turnover of large organelles such as proteasomes and mitochondria, lysosomal destablization, leakage of proteases into the cytosol and apoptosis. In this review, we summarize reports published since our last assessments of the field of oxidized protein degradation including a role for modified proteins in the induction of apoptosis.


Subject(s)
Aging/metabolism , Apoptosis/physiology , Lysosomes/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteins/metabolism , Endosomes/metabolism , Oxidation-Reduction
13.
Biochem J ; 403(2): 261-6, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17233627

ABSTRACT

The calcein-AM (calcein-acetoxymethyl ester) method is a widely used technique that is supposed to assay the intracellular 'labile iron pool' (LIP). When cells in culture are exposed to this ester, it passes the plasma membrane and reacts with cytosolic unspecific esterases. One of the reaction products, calcein, is a fluorochrome and a hydrophilic alcohol to which membranes are non-permeable and which, consequently, is retained within the cytosol of cells. Calcein fluorescence is quenched following chelation of low-mass labile iron, and the degree of quenching gives an estimate of the amounts of chelatable iron. However, a requirement for the assay to be able to demonstrate cellular LIP in total is that such iron be localized in the cytosol and not in a membrane-limited compartment. For some time it has been known that a major part of cellular, redox-active, labile, low-mass iron is temporarily localized in the lysosomal compartment as a result of the autophagic degradation of ferruginous materials, such as mitochondrial complexes and ferritin. Even if some calcein-AM may escape cytosolic esterases and enter lysosomes to be cleaved by lysosomal acidic esterases, the resulting calcein does not significantly chelate iron at

Subject(s)
Esters/analysis , Fluoresceins/analysis , Iron Chelating Agents/analysis , Iron/analysis , Iron/metabolism , Cell Membrane Permeability , Cytosol/enzymology , Esterases/metabolism , Esters/metabolism , Fluoresceins/metabolism , HeLa Cells , Humans , Hydrogen-Ion Concentration , Iron Chelating Agents/metabolism , Kinetics , Lysosomes/metabolism , Oxidative Stress , Phagocytosis , Substrate Specificity
14.
FASEB J ; 20(13): 2281-90, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17077305

ABSTRACT

OBJECTIVE: Earlier we suggested that atheroma lesions constitute a "death zone" containing toxic materials that may cause dysfunction and demise of invading macrophages to prevent the removal of plaque materials. Here we have assessed the cytotoxic effects of nonfractionated gruel and insoluble (ceroid-like) material derived from advanced human atheroma. METHODS AND RESULTS: The insoluble material within advanced atherosclerotic plaque was isolated following protease K digestion and extensive extraction with aqueous and organic solvents. FTIR, Raman, and atomic absorption spectroscopy suggested that, despite its fluorescent nature, this material closely resembled hydroxyapatite and dentin, but also contained a significant amount of iron and calcium. When added to J774 cells and human macrophages in culture, this insoluble substance was phagocytosed, and progressive cell death followed. However, an even more cytotoxic activity was found in the atheromatous "gruel" that contains abundant carbonyls/aldehydes. Cell death caused by both crude gruel and ceroid could be blocked by preincubating cells with the lipophilic iron chelator salicylaldehyde isonicotinoyl hydrazone, apoferritin, BAPTA/AM, or sodium borohydride, indicating that cellular iron, calcium, and reactive aldehyde(s) are responsible for the observed cytotoxicity. CONCLUSIONS: Toxic materials within atheromatous lesions include both ceroid and even more cytotoxic lipidaceous materials. The cytotoxic effects of these plaque components may help explain the persistence of atherosclerotic lesions.


Subject(s)
Aorta, Abdominal/pathology , Atherosclerosis/pathology , Macrophages/pathology , Mammary Arteries/pathology , Animals , Arterial Occlusive Diseases/pathology , Cell Death , Cell Line , Culture Media , Humans , Mice , Rabbits
15.
Biochem J ; 394(Pt 1): 275-83, 2006 Feb 15.
Article in English | MEDLINE | ID: mdl-16236025

ABSTRACT

The introduction of apo-ferritin or the iron chelator DFO (desferrioxamine) conjugated to starch into the lysosomal compartment protects cells against oxidative stress, lysosomal rupture and ensuing apoptosis/necrosis by binding intralysosomal redox-active iron, thus preventing Fenton-type reactions and ensuing peroxidation of lysosomal membranes. Because up-regulation of MTs (metallothioneins) also generates enhanced cellular resistance to oxidative stress, including X-irradiation, and MTs were found to be capable of iron binding in an acidic and reducing lysosomal-like environment, we propose that these proteins might similarly stabilize lysosomes following autophagocytotic delivery to the lysosomal compartment. Here, we report that Zn-mediated MT up-regulation, assayed by Western blotting and immunocytochemistry, results in lysosomal stabilization and decreased apoptosis following oxidative stress, similar to the protection afforded by fluid-phase endocytosis of apo-ferritin or DFO. In contrast, the endocytotic uptake of an iron phosphate complex destabilized lysosomes against oxidative stress, but this was suppressed in cells with up-regulated MT. It is suggested that the resistance against oxidative stress, known to occur in MT-rich cells, may be a consequence of autophagic turnover of MT, resulting in reduced iron-catalysed intralysosomal peroxidative reactions.


Subject(s)
Lysosomes/metabolism , Metallothionein/metabolism , Oxidative Stress , Animals , Apoptosis , Autophagy , Cell Line , Iron/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice , Necrosis , Oxidation-Reduction , Oxidative Stress/drug effects , Protein Binding , Up-Regulation/drug effects , Zinc/pharmacology
16.
Mol Aspects Med ; 27(5-6): 471-82, 2006.
Article in English | MEDLINE | ID: mdl-16973208

ABSTRACT

Normal mitochondrial respiration is associated with a continuous production of superoxide and hydrogen peroxide, inevitably resulting in minor macromolecular damage. Damaged cellular components are not completely turned over by autophagy and other cellular repair systems, leading to a progressive age-related accumulation of biological "garbage" material, such as defective mitochondria, cytoplasmic protein aggregates and an intralysosomal undegradable material, lipofuscin. These changes primarily affect neurons, cardiac myocytes and other long-lived postmitotic cells that neither dilute this "garbage" by mitotic activity, nor are replaced by newly differentiated cells. Defective mitochondria are insufficient in ATP production and often generate increased amounts of reactive oxygen species, further enhancing oxidative stress. Lipofuscin-loaded lysosomes, in turn, poorly turn over mitochondria that gradually leads to the overload of long-lived postmitotic cells with "garbage" material, decreased adaptability and eventual cell death.


Subject(s)
Cellular Senescence/physiology , Lysosomes/metabolism , Mitochondria/metabolism , Animals , Autophagy , Cell Proliferation , Humans , Mitosis
17.
Antioxid Redox Signal ; 8(1-2): 197-204, 2006.
Article in English | MEDLINE | ID: mdl-16487053

ABSTRACT

Normal metabolism is associated with unavoidable mild oxidative stress resulting in biomolecular damage that cannot be totally repaired or removed by cellular degradative systems, including lysosomes, proteasomes, and cytosolic and mitochondrial proteases. Consequently, irreversibly damaged and functionally defective structures (biological 'garbage') accumulate within long-lived postmitotic cells, such as cardiac myocytes and neurons, leading to progressive loss of adaptability and increased probability of death and characterizing a process called aging, or senescence. Intralysosomal 'garbage' is represented by lipofuscin (age pigment), an undegradable autophagocytosed material, while extralysosomal 'garbage' involves oxidatively modified cytosolic proteins, altered biomembranes, defective mitochondria and other organelles. In aged postmitotic cells, heavily lipofuscin-loaded lysosomes perform poorly, resulting in the enhanced accumulation of defective mitochondria, which in turn produce more reactive oxygen species causing additional damage (the mitochondrial-lysosomal axis theory). Potential anti-aging strategies may involve not only overall reduction of oxidative stress, but also the use of intralysosomal iron chelators hampering Fenton-type chemistry as well as the stimulation of cellular degradative systems.


Subject(s)
Aging/physiology , Oxidative Stress/physiology , Reactive Oxygen Species , Animals , Death , Humans , Lipofuscin/physiology , Lysosomes/physiology , Mitosis , Models, Biological
18.
FEBS J ; 273(13): 3106-17, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16762036

ABSTRACT

Oxidant-induced cell damage may be initiated by peroxidative injury to lysosomal membranes, catalyzed by intralysosomal low mass iron that appears to comprise a major part of cellular redox-active iron. Resulting relocation of lytic enzymes and low mass iron would result in secondary harm to various cellular constituents. In an effort to further clarify this still controversial issue, we tested the protective effects of two potent iron chelators--the hydrophilic desferrioxamine (dfo) and the lipophilic salicylaldehyde isonicotinoyl hydrazone (sih), using cultured lysosome-rich macrophage-like J774 cells as targets. dfo slowly enters cells via endocytosis, while the lipophilic sih rapidly distributes throughout the cell. Following dfo treatment, long-term survival of cells cannot be investigated because dfo by itself, by remaining inside the lysosomal compartment, induces apoptosis that probably is due to iron starvation, while sih has no lasting toxic effects if the exposure time is limited. Following preincubation with 1 mM dfo for 3 h or 10 microM sih for a few minutes, both agents provided strong protection against an ensuing approximately LD50 oxidant challenge by preventing lysosomal rupture, ensuing loss of mitochondrial membrane potential, and apoptotic/necrotic cell death. It appears that once significant lysosomal rupture has occurred, the cell is irreversibly committed to death. The results lend strength to the concept that lysosomal membranes, normally exposed to redox-active iron in high concentrations, are initial targets of oxidant damage and support the idea that chelators selectively targeted to the lysosomal compartment may have therapeutic utility in diminishing oxidant-mediated cell injury.


Subject(s)
Iron/pharmacology , Lysosomes/metabolism , Oxidative Stress , Aldehydes/pharmacology , Apoptosis , Cell Proliferation , Chelating Agents/pharmacology , Deferoxamine/pharmacology , Endocytosis , Hydrazones/pharmacology , Hydrogen Peroxide/chemistry , Hydrogen Peroxide/pharmacology , Iron/chemistry , Iron Chelating Agents/pharmacology , Lysosomes/chemistry , Membrane Potentials , Models, Biological , Necrosis , Oxidants/chemistry , Oxidants/metabolism
19.
APMIS ; 114(12): 874-83, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17207088

ABSTRACT

This study aimed to determine the effects of advanced glycation end products (AGEs) on endothelial cytoskeleton morphology and permeability, and to detect the underlying signaling mechanisms involved in these responses. Cultured endothelial cells (ECs) were exposed to AGE-modified human serum albumin (AGE-HSA), and EC cytoskeletal changes were evaluated by observing fluorescence of F-actin following ligation with labeled antibodies. Endothelial permeability was detected by measuring the flux of TRITC-albumin across the EC monolayers. To explore the signaling pathways behind AGE-induced EC alteration, ECs were treated with either soluble anti-AGE receptor (RAGE) IgG, or the MAPK inhibitors PD98059 and SB203580 before AGE-HSA administration. To further elucidate possible involvement of the ERK and p38 pathways in AGE-induced EC changes, adenovirus-carried recombinant constitutive dominant-negative forms of upstream ERK and p38 kinases, namely MEK1(A) and MKK6b(A), were pre-infected into ECs 24 h prior to AGE-HSA exposure. AGE-HSA induced actin cytoskeleton rearrangement, as well as EC hyperpermeability, in a dose and time-dependent manner. The effects were attenuated in cells pretreated with anti-RAGE IgG, PD98059 or SB203580, respectively. EC pre-infection with MEK1(A) and MKK6b(A) also alleviated the effect of AGEs. Furthermore, adenovirus-mediated administration of activated forms of either MEK1 or MKK6b alone induced rearrangement of F-actin and hyperpermeability. The results indicate that ERK and p38 MAPK play important roles in the mediation of AGE-induced EC barrier dysfunction associated with morphological changes of the F-actin.


Subject(s)
Actins/metabolism , Endothelial Cells/metabolism , Glycation End Products, Advanced/pharmacology , Serum Albumin/pharmacology , Capillary Permeability/drug effects , Cell Line , Cytoskeleton/metabolism , Endothelial Cells/cytology , Endothelial Cells/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Flavonoids/pharmacology , Humans , Imidazoles/pharmacology , MAP Kinase Signaling System/drug effects , Microscopy, Fluorescence , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Receptor for Advanced Glycation End Products , Receptors, Immunologic/immunology , Serum Albumin, Human , p38 Mitogen-Activated Protein Kinases/metabolism
20.
Chem Biol Interact ; 163(1-2): 29-37, 2006 Oct 27.
Article in English | MEDLINE | ID: mdl-16737690

ABSTRACT

Aging (senescence) is characterized by a progressive accumulation of macromolecular damage, supposedly due to a continuous minor oxidative stress associated with mitochondrial respiration. Aging mainly affects long-lived postmitotic cells, such as neurons and cardiac myocytes, which neither divide and dilute damaged structures, nor are replaced by newly differentiated cells. Because of inherent imperfect lysosomal degradation (autophagy) and other self-repair mechanisms, damaged structures (biological "garbage") progressively accumulate within such cells, both extra- and intralysosomally. Defective mitochondria and aggregated proteins are the most typical forms of extralysosomal "garbage", while lipofuscin that forms due to iron-catalyzed oxidation of autophagocytosed or heterophagocytosed material, represents intralysosomal "garbage". Based on findings that autophagy is diminished in lipofuscin-loaded cells and that cellular lipofuscin content positively correlates with oxidative stress and mitochondrial damage, we have proposed the mitochondrial-lysosomal axis theory of aging, according to which mitochondrial turnover progressively declines with age, resulting in decreased ATP production and increased oxidative damage. Due to autophagy of ferruginous material, lysosomes contain a pool of redox-active iron, which makes these organelles particularly susceptible to oxidative damage. Oxidant-mediated destabilization of lysosomal membranes releases hydrolytic enzymes to the cytosol, eventuating in cell death (either apoptotic or necrotic depending on the magnitude of the insult), while chelation of the intralysosomal pool of redox-active iron prevents these effects. In relation to the onset of oxidant-induced apoptosis, but after the initiating lysosomal rupture, cytochrome c is released from mitochondria and caspases are activated. Mitochondrial damage follows the release of lysosomal hydrolases, which may act either directly or indirectly, through activation of phospholipases or pro-apoptotic proteins such as Bid. Additional lysosomal rupture seems to be a consequence of a transient oxidative stress of mitochondrial origin that follows the attack by lysosomal hydrolases and/or phospholipases, creating an amplifying loop system.


Subject(s)
Aging/physiology , Apoptosis/physiology , Lysosomes/physiology , Mitochondria/physiology , Animals , Cell Communication/physiology , Humans , Oxidation-Reduction , Oxidative Stress/physiology
SELECTION OF CITATIONS
SEARCH DETAIL