Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Cell Biol ; 169(2): 331-9, 2005 Apr 25.
Article in English | MEDLINE | ID: mdl-15837797

ABSTRACT

Excessive accumulation of amyloid beta-peptide (Abeta) plays an early and critical role in synapse and neuronal loss in Alzheimer's Disease (AD). Increased oxidative stress is one of the mechanisms whereby Abeta induces neuronal death. Given the lessened susceptibility to oxidative stress exhibited by mice lacking p66Shc, we investigated the role of p66Shc in Abeta toxicity. Treatment of cells and primary neuronal cultures with Abeta caused apoptotic death and induced p66Shc phosphorylation at Ser36. Ectopic expression of a dominant-negative SEK1 mutant or chemical JNK inhibition reduced Abeta-induced JNK activation and p66Shc phosphorylation (Ser36), suggesting that JNK phosphorylates p66Shc. Abeta induced the phosphorylation and hence inactivation of forkhead transcription factors in a p66Shc-dependent manner. Ectopic expression of p66ShcS36A or antioxidant treatment protected cells against Abeta-induced death and reduced forkhead phosphorylation, suggesting that p66Shc phosphorylation critically influences the redox regulation of forkhead proteins and underlies Abeta toxicity. These findings underscore the potential usefulness of JNK, p66Shc, and forkhead proteins as therapeutic targets for AD.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Amyloid beta-Peptides/toxicity , Apoptosis/drug effects , Nuclear Proteins/metabolism , Signal Transduction/drug effects , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing/genetics , Alzheimer Disease/physiopathology , Amino Acid Substitution , Animals , Forkhead Transcription Factors , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Kinase 4 , Mice , Mitogen-Activated Protein Kinase Kinases/metabolism , Neurons/metabolism , Oxidation-Reduction/drug effects , PC12 Cells , Phosphorylation/drug effects , Point Mutation , Rats , Serine/metabolism , Shc Signaling Adaptor Proteins , Src Homology 2 Domain-Containing, Transforming Protein 1
2.
Free Radic Res ; 38(7): 729-37, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15453638

ABSTRACT

Singlet oxygen, an electronically excited form of molecular oxygen, is a primary mediator of the activation of stress-activated protein kinases elicited by ultraviolet A (UVA; 320-400 nm). Here, the effects of singlet oxygen (1O2) on the extracellular signal-regulated kinase (ERK) 1/2 and Akt/protein kinase B pathways were analyzed in human dermal fibroblasts. While basal ERK 1/2 phosphorylation was lowered in cells exposed to either 1O2, UVA or photodynamic treatment, Akt was moderately activated by photochemically generated 1O2 in a phosphoinositide 3-kinase (PI3K)-dependent fashion, resulting in the phosphorylation of glycogen synthase kinase-3 (GSK3). The activation of ERK 1/2 and Akt as induced by stimulation with epidermal growth factor (EGF) or platelet-derived growth factor (PDGF) was inhibited by 1O2 generated intracellularly upon photoexcitation of rose Bengal (RB). Photodynamic therapy (PDT)-induced apoptosis is known to be associated with increased formation of ceramides. Likewise, both 1O2 and UVA induced ceramide generation in human skin fibroblasts. The attenuation of EGF- and PDGF-induced activation of ERK 1/2 and Akt by 1O2 was mimicked by stimulation of fibroblasts with the cell-permeable C2-ceramide. Interestingly, EGF-induced tyrosine phosphorylation of the EGF receptor was strongly attenuated by 1O2 but unimpaired by C2-ceramide, implying that, although ceramide formation may mediate the above attenuation of ERK and Akt phosphorylation induced by 1O2, mechanisms beyond ceramide formation exist that mediate impairment of growth factor signaling by singlet oxygen. In summary, these data point to a novel mechanism of 1O2 toxicity: the known 1O2-induced activation of proapoptotic kinases such as JNK and p38 is paralleled by the prevention of activation of growth factor receptor-dependent signaling and of anti-apoptotic kinases, thus shifting the balance towards apoptosis.


Subject(s)
Ceramides/metabolism , Growth Substances/pharmacology , Signal Transduction/drug effects , Singlet Oxygen/pharmacology , Cells, Cultured , Ceramides/biosynthesis , Ceramides/pharmacology , Enzyme Activation/drug effects , ErbB Receptors/metabolism , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphotyrosine/drug effects , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , p38 Mitogen-Activated Protein Kinases/metabolism
3.
Exp Gerontol ; 39(5): 735-44, 2004 May.
Article in English | MEDLINE | ID: mdl-15130668

ABSTRACT

The endoplasmic reticulum (ER), as a processing plant for the folding and posttranslational modification of proteins, is exquisitely sensitive to changes in its internal environment. Various conditions, collectively termed 'ER stress', can perturb ER functions, leading to the activation of a complex response known as the unfolded protein response. Here, we investigated the response of hepatocytes derived from young (4-5 months) and aged (24-26 months) rats to two agents, thapsigargin (TG) and tunicamycin (TM), which act via different mechanisms to induce ER stress. Old hepatocytes displayed greater cell death than young cells following treatment with TG or TM, associated with higher expression of the pro-apoptotic gene gadd153 (also known as chop) and enhanced c-Jun N-terminal protein kinase (JNK) activation. Pharmacologic inhibition of JNK decreased the expression of TG-stimulated gadd153 in old cells and reduced their sensitivity to TG-induced cell death. Inhibition of p38, on the other hand, enhanced TG-induced gadd153 expression and JNK activation, and augmented TG-induced cell death. Additional experiments implicated the PERK/eIF-2 alpha signaling pathway as a contributor to the higher Gadd153 expression and JNK activation, and greater sensitivity of old cells to ER stress.


Subject(s)
Aging/physiology , CCAAT-Enhancer-Binding Proteins/analysis , Endoplasmic Reticulum/physiology , Hepatocytes/drug effects , Mitogen-Activated Protein Kinases/metabolism , Transcription Factors/analysis , Animals , Apoptosis/drug effects , CCAAT-Enhancer-Binding Proteins/genetics , Cells, Cultured , Endoplasmic Reticulum/drug effects , Enzyme Inhibitors/pharmacology , Gene Expression , Glycosylation/drug effects , Hepatocytes/physiology , JNK Mitogen-Activated Protein Kinases , Male , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Rats , Rats, Inbred F344 , Thapsigargin/pharmacology , Transcription Factor CHOP , Transcription Factors/genetics , Tunicamycin/pharmacology , p38 Mitogen-Activated Protein Kinases
4.
Free Radic Biol Med ; 35(3): 292-9, 2003 Aug 01.
Article in English | MEDLINE | ID: mdl-12885591

ABSTRACT

Aging is often characterized by reduced stress tolerance and decreased proliferative capacity, but little is known about the effects of aging on signaling pathways important in regulating these responses. Recent studies from our laboratory have implicated impairments in epidermal growth factor receptor (EGFR) signaling and extracellular signal-regulated kinase (ERK) activation to both effects in rat hepatocytes. Here we investigated the responsiveness of hepatocytes derived from young (4-5 months) and aged (24-29 months) mice to proliferative signals (low concentrations of H2O2 and epidermal growth factor [EGF] stimulation), and oxidant injury (high H2O2 concentrations). Old hepatocytes displayed lower levels of DNA synthesis in response to low H(2)O(2) concentrations (5-10 microM) and EGF stimulation, and reduced survival following treatment with high H2O2 concentrations (20-50 microM). Both effects were associated with reduced activation of ERK and diminished phosphorylation of EGFR tyrosine residue 1173. p38 was also activated by H2O2, but to a greater extent in old cells. Pharmacologic inhibition of ERK increased the sensitivity of young cells to H2O2-induced cell death, while inhibition of p38 decreased the sensitivity of old cells. Our findings suggest that impairments in common signaling events underlie age-related declines in proliferative capacity and oxidative stress tolerance in mouse hepatocytes, and that an imbalance in ERK and p38 activities contributes to the greater sensitivity of aged cells to H2O2.


Subject(s)
Aging , Oxidative Stress , Animals , Antimetabolites/pharmacology , Bromodeoxyuridine/pharmacology , Cell Division , Cell Survival , DNA/biosynthesis , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , ErbB Receptors/metabolism , Free Radicals , Hepatocytes/metabolism , Hydrogen Peroxide/pharmacology , Kinetics , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Signal Transduction , Time Factors , p38 Mitogen-Activated Protein Kinases
5.
Exp Gerontol ; 38(4): 431-9, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12670630

ABSTRACT

It is well established that the proliferative potential of the liver declines with aging. Epidermal growth factor (EGF)-stimulated DNA synthesis is reduced in hepatocytes from aged rats relative to young rats, and this reduction correlates with diminished activation of the extracellular signal-regulated kinase (ERK) pathway and lower phosphorylation of the EGF receptor on residue Y1173. Calorie restriction (CR) can increase rodent life span and retard many age-associated declines in physiologic function, but its influence on cell proliferation is unknown. Here, we investigated the effects of long-term CR on proliferation of hepatocytes derived from young and aged rats following in vitro stimulation with either low-dose hydrogen peroxide or EGF. CR reduced the proliferative response of hepatocytes derived from young hosts, but long-term CR was associated with enhanced proliferation in aged cells relative to that of ad libitum (AL)-fed animals. ERK activation mirrored the effects of CR on proliferation, in that young CR cells exhibited lower ERK activation than young AL cells, but old CR cells showed higher ERK activation than old AL cells. Finally, a decline in EGF receptor phosphorylation on Y1173, which normally occurs with aging, was absent in cells of old hosts maintained on long-term CR, supporting the view that alterations in this early signaling event underlie the age-related decline in proliferative potential in rat hepatocytes.


Subject(s)
Aging/physiology , Cell Cycle Proteins , Dietary Carbohydrates/administration & dosage , Hepatocytes/cytology , Phosphoprotein Phosphatases , Animals , Blotting, Northern/methods , Blotting, Western/methods , Cell Division , Cells, Cultured , DNA/biosynthesis , Dual Specificity Phosphatase 1 , Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Genes, jun , Hepatocytes/metabolism , Hydrogen Peroxide/pharmacology , Immediate-Early Proteins/genetics , Male , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Precipitin Tests/methods , Protein Phosphatase 1 , Protein Tyrosine Phosphatases/genetics , RNA, Messenger/analysis , Rats , Rats, Inbred F344 , Stimulation, Chemical
6.
Mol Cell Biol ; 23(7): 2407-14, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12640124

ABSTRACT

The inhibitory effect of caveolin on the cellular response to growth factor stimulation is well established. Given the significant overlap in signaling pathways involved in regulating cell proliferation and stress responsiveness, we hypothesized that caveolin would also affect a cell's ability to respond to environmental stress. Here we investigated the ability of caveolin-1 to modulate the cellular response to sodium arsenite and thereby alter survival of the human cell lines 293 and HeLa. Cells stably transfected with caveolin-1 were found to be much more sensitive to the toxic effects of sodium arsenite than either untransfected parental cells or parental cells transfected with an empty vector. Unexpectedly, the caveolin-overexpressing cells also exhibited a significant activation of the phosphatidylinositol 3-kinase (PI3K)/Akt pathway, which additional studies suggested was likely due to decreased neutral sphingomyelinase activity and ceramide synthesis. In contrast to its extensively documented antiapoptotic influence, the elevated activity of Akt appears to be important in sensitizing caveolin-expressing cells to arsenite-induced toxicity, as both pretreatment of cells with the PI3K inhibitor wortmannin and overexpression of a dominant-negative Akt mutant markedly improved the survival of arsenite-treated cells. This death-promoting influence of the PI3K/Akt pathway in caveolin-overexpressing cells appeared not to be unique to sodium arsenite, as wortmannin pretreatment also resulted in increased survival in the presence of H(2)O(2). In summary, our results indicate that caveolin-induced upregulation of the PI3K/Akt signaling pathway, which appears to be a death signal in the presence of arsenite and H(2)O(2), sensitizes cells to environmental stress.


Subject(s)
Arsenites/toxicity , Caveolins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Signal Transduction/physiology , Androstadienes/pharmacology , Blotting, Western , Caveolin 1 , Caveolins/genetics , Cell Line , Cell Survival/drug effects , Cell Survival/genetics , Ceramides/metabolism , Enzyme Activation/drug effects , Enzyme Activation/genetics , Enzyme Inhibitors/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Genes, Dominant , HeLa Cells , Humans , Oxidants/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Signal Transduction/drug effects , Transfection , Wortmannin
7.
J Biol Chem ; 278(19): 16726-31, 2003 May 09.
Article in English | MEDLINE | ID: mdl-12609979

ABSTRACT

Aging is generally accompanied by reduced tolerance to oxidative stress and altered responsiveness to proliferative signals. We have shown that hepatocytes derived from aged rats (24-26 months) exhibit greater sensitivity to H(2)O(2) treatment and reduced proliferation following epidermal growth factor (EGF) treatment than cells of young adult rats (5-6 months). Here we examined the effects of aging and calorie restriction (CR) on expression of the oxidative stress-inducible and pro-apoptotic gene gadd153 (chop) in these hepatocytes, and we investigated its influence on sensitivity to oxidants. We show that aging was associated with elevated expression of gadd153, both basally and in response to H(2)O(2) treatment. CR, which attenuates age-associated declines in stress tolerance, prevented the age-related increase in gadd153 expression. EGF treatment also resulted in gadd153 induction in old cells. This effect was absent in young cells and in old cells of CR rats. gadd153 induction by EGF was reactive oxygen species-dependent and correlated with heightened sensitivity to subsequent H(2)O(2) treatment, suggesting that elevated Gadd153 contributes to the greater sensitivity of EGF-pretreated old cells to oxidative stress. Additional support for this hypothesis was provided by experiments with Rat1 fibroblasts in which conditional expression of Gadd153 conferred increased sensitivity to H(2)O(2). We propose a model whereby the diminished ability of old hepatocytes to overcome an EGF-triggered reactive oxygen species load leads to induction of the proapoptotic gene gadd153, which, in turn, sensitizes the cells to oxidant injury. Our findings point to gadd153 expression levels as an important factor in liver aging.


Subject(s)
Aging/genetics , CCAAT-Enhancer-Binding Proteins/genetics , Liver/metabolism , Oxidative Stress/genetics , Transcription Factors/genetics , Aging/metabolism , Aging/pathology , Animals , Apoptosis/genetics , CCAAT-Enhancer-Binding Proteins/biosynthesis , Gene Expression Regulation , Liver/pathology , Male , Rats , Rats, Inbred F344 , Transcription Factor CHOP , Transcription Factors/biosynthesis
8.
J Biol Chem ; 278(18): 15558-64, 2003 May 02.
Article in English | MEDLINE | ID: mdl-12601012

ABSTRACT

The unfolded protein response, which is activated in response to the loss of endoplasmic reticulum (ER) Ca(2+) homeostasis and/or the accumulation of misfolded, unassembled, or aggregated proteins in the ER lumen, involves both transcriptional and translational regulation. In the current studies we sought to identify novel ER stress-induced genes by conducting microarray analysis on tunicamycin-treated cells. We identified P58(IPK), an inhibitor of the interferon-induced double-stranded RNA-activated protein kinase, as induced during ER stress. Additional studies suggested that p58(IPK) induction was mediated via ATF6 and that P58(IPK) played a role in down-regulating the activity of the pancreatic eIF2 kinase/eukaryotic initiation factor 2alpha (eIF2alpha)-like ER kinase/activation transcription factor (ATF) 4 pathway. Modulation of P58(IPK) levels altered the phosphorylation status of eIF2alpha, and thereby affected expression of its downstream targets, ATF4 and Gadd153. Overexpression of P58(IPK) inhibited eIF2alpha phosphorylation and reduced ATF4 and Gadd153 protein accumulation, whereas silencing of P58(IPK) expression enhanced pancreatic eIF2alpha-like ER kinase and eIF2alpha phosphorylation and increased ATF4 and Gadd153 accumulation. These findings implicate P58(IPK) as an important component of a negative feedback loop used by the cell to inhibit eIF2alpha signaling, and thus attenuate the unfolded protein response.


Subject(s)
Eukaryotic Initiation Factor-2/physiology , Repressor Proteins/physiology , Activating Transcription Factor 6 , Apoptosis , CCAAT-Enhancer-Binding Proteins/biosynthesis , DNA-Binding Proteins/physiology , Endoplasmic Reticulum/metabolism , Gene Silencing , HSP40 Heat-Shock Proteins , Humans , Oligonucleotide Array Sequence Analysis , Phosphorylation , Protein Folding , RNA, Small Interfering , Repressor Proteins/genetics , Transcription Factor CHOP , Transcription Factors/biosynthesis , Transcription Factors/physiology , eIF-2 Kinase/metabolism
9.
J Neurochem ; 83(3): 673-81, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12390529

ABSTRACT

Mutations in the presenilin-1 (PS1) gene cause early onset familial Alzheimer's disease (FAD) by a mechanism believed to involve perturbed endoplasmic reticulum (ER) function and altered proteolytic processing of the amyloid precursor protein. We investigated the molecular mechanisms underlying cell death and ER dysfunction in cultured cells and knock-in mice expressing FAD PS1 mutations. We report that PS1 mutations cause a marked increase in basal protein levels of the pro-apoptotic transcription factor Gadd153. PS1 mutations increase Gadd153 protein translation without affecting mRNA levels, while decreasing levels of the anti-apoptotic protein Bcl-2. Moreover, an exaggerated Gadd153 response to stress induced by ER stress agents was observed in PS1 mutant cells. Cell death in response to ER stress is enhanced by PS1 mutations, and this endangering effect is attenuated by anti-sense-mediated suppression of Gadd153 production. An abnormality in the translational regulation of Gadd153 may sensitize cells to the detrimental effects of ER stress and contribute to the pathogenic actions of PS1 mutations in FAD.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , Membrane Proteins/genetics , Transcription Factors/metabolism , Alzheimer Disease/metabolism , Animals , Apoptosis/physiology , CCAAT-Enhancer-Binding Proteins/antagonists & inhibitors , CCAAT-Enhancer-Binding Proteins/genetics , Calcium/metabolism , Calcium-Transporting ATPases/antagonists & inhibitors , Cell Line , Cerebral Cortex/metabolism , Clone Cells , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Hippocampus/metabolism , Humans , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Mutation , Oligonucleotides, Antisense/pharmacology , PC12 Cells , Presenilin-1 , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/metabolism , Rats , Stress, Physiological/metabolism , Thapsigargin/pharmacology , Transcription Factor CHOP , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Up-Regulation/drug effects , Up-Regulation/physiology
10.
Biochem Pharmacol ; 64(5-6): 999-1005, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12213598

ABSTRACT

Accumulation of oxidative damage is believed to be a major contributor to the decline in physiologic function that characterizes mammalian aging, and recent studies suggest that how well you respond to acute oxidative stress is an important factor in determining longevity. Oxidant injury elicits a wide spectrum of responses ranging from proliferation to cell death. The particular outcome observed largely reflects the severity of the stress encountered and the relative degree of activation of various signal transduction pathways aimed at enhancing survival or inducing cell death. Herein we examine the relationship between pathways important in supporting cell survival in response to oxidant injury and those involved in regulating proliferation. We review evidence indicating that [Curr. Opin. Cell Biol. 10 (1998) 248] common pathways are indeed involved in regulating these responses, and [Physiol. Rev. 82 (2002) 47] alterations in shared signaling events likely account for the age-related decline in the ability of cells to respond to both proliferative signals and oxidant stimuli.


Subject(s)
Aging/physiology , Growth Substances/metabolism , Oxidative Stress/physiology , Signal Transduction/physiology , Animals , Cell Division/physiology , Eating , Humans , Longevity , Receptors, Growth Factor/metabolism
11.
J Cell Physiol ; 192(1): 1-15, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12115731

ABSTRACT

Reactive oxygen species (ROS), whether produced endogenously as a consequence of normal cell functions or derived from external sources, pose a constant threat to cells living in an aerobic environment as they can result in severe damage to DNA, protein, and lipids. The importance of oxidative damage to the pathogenesis of many diseases as well as to degenerative processes of aging has becoming increasingly apparent over the past few years. Cells contain a number of antioxidant defenses to minimize fluctuations in ROS, but ROS generation often exceeds the cell's antioxidant capacity, resulting in a condition termed oxidative stress. Host survival depends upon the ability of cells and tissues to adapt to or resist the stress, and repair or remove damaged molecules or cells. Numerous stress response mechanisms have evolved for these purposes, and they are rapidly activated in response to oxidative insults. Some of the pathways are preferentially linked to enhanced survival, while others are more frequently associated with cell death. Still others have been implicated in both extremes depending on the particular circumstances. In this review, we discuss the various signaling pathways known to be activated in response to oxidative stress in mammalian cells, the mechanisms leading to their activation, and their roles in influencing cell survival. These pathways constitute important avenues for therapeutic interventions aimed at limiting oxidative damage or attenuating its sequelae.


Subject(s)
Cell Physiological Phenomena , Oxidative Stress/physiology , Protein Serine-Threonine Kinases , Signal Transduction/physiology , Animals , Cell Survival/physiology , Heat-Shock Proteins/metabolism , Humans , Isoenzymes/physiology , Mitogen-Activated Protein Kinases/physiology , NF-kappa B/physiology , Phosphatidylinositol 3-Kinases/physiology , Phospholipase C gamma , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-akt , Tumor Suppressor Protein p53/physiology , Type C Phospholipases/physiology
12.
Cancer Res ; 62(11): 3257-63, 2002 Jun 01.
Article in English | MEDLINE | ID: mdl-12036942

ABSTRACT

Although the c-Jun NH(2)-terminal kinase (JNK) pathway has been implicated in mediating cell growth and transformation, its downstream effectors remain to be identified. Using JNK2 antisense oligonucleotides (JNK2AS), we uncovered previously a role for JNK2 in regulating cell cycle progression and survival of human PC3 prostate carcinoma cells. Here, to identify genes involved in implementing JNK2-mediated effects, we have analyzed global gene expression changes in JNK2-deprived PC3 cells using Serial Analysis of Gene Expression. More than 40,000 tags each were generated from control and PC3-JNK2AS libraries, corresponding to 15,999 and 20,698 unique transcripts, respectively. Transcripts corresponding to transcription factors, stress-induced genes, and apoptosis-related genes were up-regulated in the PC3-JNK2AS library, revealing a significant stress response after the inhibition of JNK2 expression. Genes involved in DNA repair, mRNA turnover, and drug resistance were found to be down-regulated by inhibition of JNK2 expression, further highlighting the importance of JNK2 signaling in regulating cell homeostasis and tumor cell growth.


Subject(s)
Mitogen-Activated Protein Kinases/physiology , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics , Apoptosis/drug effects , Apoptosis/physiology , Blotting, Northern , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Division/physiology , Gene Expression/drug effects , Gene Expression Profiling , Humans , JNK Mitogen-Activated Protein Kinases , Male , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/genetics , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/pharmacology , Prostatic Neoplasms/pathology , Signal Transduction/physiology
13.
FASEB J ; 16(1): 114-6, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11709495

ABSTRACT

Oxidative stress is believed to be an important factor in the development of age-related diseases, and studies in lower organisms have established links between oxidative stress tolerance and longevity. We have hypothesized that aging is associated with a reduced ability to mount acute host defenses to oxidant injury, which increases the vulnerability of aged cells to stress. We tested this hypothesis by using primary hepatocytes from young (4-6 months) and aged (24-26 months) rats. Old hepatocytes were more sensitive to H2O2-induced apoptosis than were young cells. Lower survival is associated with reduced activations of extracellular signal-regulated kinase (ERK) and Akt kinase, both of which protect against oxidant injury. That reduced ERK and Akt activities contribute to lower survival of aged cells was supported by additional findings. First, pharmacologic inhibition of ERK and Akt activation in young cells markedly increased their sensitivity to H2O2. Second, caloric restriction, which increases rodent life span and delays the onset of many age-related declines in physiologic function, prevented loss in ERK and Akt activation by H2O2 and enhanced survival of old hepatocytes to levels similar to those of young cells. Strategies aimed at boosting these host responses to acute oxidant injury could have significant anti-aging benefits.


Subject(s)
Aging , Mitogen-Activated Protein Kinases/metabolism , Oxidative Stress , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Animals , Cell Survival , Cells, Cultured , Chromones/pharmacology , Drug Synergism , Energy Intake , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Hepatocytes/drug effects , Hepatocytes/enzymology , Hydrogen Peroxide/pharmacology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Models, Biological , Morpholines/pharmacology , Oxidants/pharmacology , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins c-akt , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...