首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 15 毫秒
1.
A molecular mechanism of aluminium-induced Alzheimer's disease?   总被引:3,自引:0,他引:3  
An abundance of research has continued to link aluminium (Al) with Alzheimer's disease (AD) (Strong et al., J. Toxicol. Environ. Health 48 (1996) 599; Savory et al., J. Toxicol. Environ. Health 48 (1996) 615). Animals loaded with Al develop both symptoms and brain lesions that are similar to those found in AD. However, these animal models of Al intoxication are not representative of human exposure to Al. They have not addressed the significance of a truly chronic exposure to Al. If Al is a cause of AD it is effective at the level of our everyday exposure to the metal and AD will be one possible outcome of the life-long presence of a low, though burgeoning, brain Al burden. Individual susceptibility to AD will be as much to do with differences in brain physiology as with changes in our everyday exposure to the metal. There will be a chemical response and indeed biochemical/physiological response in the brain to Al. The question is whether brain Al homeostasis could impact upon brain function. In reviewing the recent literature covering the neurotoxicity of Al and, in particular, of the known and probable mechanisms involved in brain Al homeostasis I have identified a mechanism through which a truly chronic exposure to Al would bring about subtle and persistent changes in neurotransmission which, in time, could instigate the cascade of events known collectively as AD. This mechanism involves the potentiation of the activities of neurotransmitters by the action of Al-ATP at adenosine 5'-triphosphate (ATP) receptors in the brain.  相似文献   

2.
Aluminum (Al) is a metal toxin that has been implicated in the etiology of a number of diseases including Alzheimer's, Parkinson's, dialysis encephalopathy, and osteomalacia. Al has been shown to exert its effects by disrupting lipid membrane fluidity, perturbing iron (Fe), magnesium, and calcium homeostasis, and causing oxidative stress. However, the exact molecular targets of aluminum's toxicity have remained elusive. In the present review, we describe how the use of a systems biology approach in cultured hepatoblastoma cells (HepG2) allowed the identification of the molecular targets of Al toxicity. Mitochondrial metabolism is the main site of the toxicological action of Al. Fe-dependent and redox sensitive enzymes in the tricarboxylic acid (TCA) cycle and oxidative phosphorylation (OXPHOS) are dramatically decreased by Al exposure. In an effort to compensate for diminished mitochondrial function, Al-treated cells stabilize hypoxia inducible factor-1α (HIF-1α) to increase ATP production by glycolysis. Additionally, Al toxicity leads to an increase in intracellular lipid accumulation due to enhanced lipogenesis and a decrease in the β-oxidation of fatty acids. Central to these effects is the alteration of α-ketoglutarate (KG) homeostasis. In Al-exposed cells, KG is preferentially used to quench ROS leading to succinate accumulation and HIF-1α stabilization. Moreover, the channeling of KG to combat oxidative stress leads to a reduction of l-carnitine biosynthesis and a concomitant decrease in fatty acid oxidation. The fluidity and interaction of these metabolic modules and the implications of these findings in liver-related disorders are discussed herein.  相似文献   

3.
Prior to the identification of the various abnormal proteins deposited as fibrillar aggregates in the Alzheimer's disease (AD) brain, there was tremendous controversy over the importance of the various lesions with respect to primacy in the pathology of AD. Nevertheless, based on analogy to systemic amyloidosis, many investigators believed that the amyloid deposits in AD played a causal role and that characterization of these deposits would hold the key to understanding this complex disease. Indeed, in retrospect, it was the initial biochemical purifications of the approximately 4 kDa amyloid beta-peptide (Abeta) from amyloid deposits in the mid 1980s that launched a new era of AD research (Glenner and Wong, Biochem. Biophys. Res. Commun. 122 (1984) 1121-1135; Wong et al., Proc. Natl. Acad Sci. USA 82 (1985) 8729 8732; and Masters et al., Proc. Natl. Acad Sci. USA 82 (1985) 4245-4249). Subsequent studies of the biology of Abeta together with genetic studies of AD have all supported the hypothesis that altered Abeta metabolism leading to aggregation plays a causal role in AD. Although there remains controversy as to whether Abeta deposited as classic amyloid or a smaller, aggregated, form causes AD, the relevance of studying the amyloid deposits has certainly been proven. Despite the significant advances in our understanding of the role of Abeta in AD pathogenesis, many important aspects of Abeta biology remain a mystery. This review will highlight those aspects of Abeta biology that have led to our increased understanding of the pathogenesis of AD as well as areas which warrant additional study.  相似文献   

4.
Aluminium (Al) is a neurotoxic metal and Al exposure may be a factor in the aetiology of various neurodegenerative diseases such as Alzheimer's disease (AD). The major pathohistological findings in the AD brain are the presence of neuritic plaques containing beta-amyloid (Abeta) which may interfere with neuronal communication. Moreover, it has been observed that GRP78, a stress-response protein induced by conditions that adversely affect endoplasmic reticulum (ER) function, is reduced in the brain of AD patients. In this study, we investigated the correlation between the expression of Abeta and GRP78 in the brain cortex of mice chronically treated with aluminium sulphate. Chronic exposure over 12 months to aluminium sulphate in drinking water resulted in deposition of Abeta similar to that seen in congophilic amyloid angiopathy (CAA) in humans and a reduction in neuronal expression of GRP78 similar to what has previously been observed in Alzheimer's disease. So, we hypothesise that chronic Al administration is responsible for oxidative cell damage that interferes with ER functions inducing Abeta accumulation and neurodegenerative damage.  相似文献   

5.
In less than a decade, beginning with the demonstration by Floyd, Stadtman, Markesbery et al. of increased reactive carbonyls in the brains of patients with Alzheimer's disease (AD), oxidative damage has been established as a feature of the disease. Here, we review the types of oxidative damage seen in AD, sites involved, possible origin, relationship to lesions, and compensatory changes, and we also consider other neurodegenerative diseases where oxidative stress has been implicated. Although much data remain to be collected, the broad spectrum of changes found in AD are only seen, albeit to a lesser extent, in normal aging with other neurodegenerative diseases showing distinct spectrums of change.  相似文献   

6.
雌激素对淀粉样β蛋白代谢的调节和毒性缓解   总被引:2,自引:0,他引:2  
Zhang S  Yao T 《生理科学进展》2003,34(3):197-201
以淀粉样β蛋白为主的老年斑胞外沉积和神经元内神经原纤维缠结,是阿尔采末病(AD)特征性的病理学改变。近来,人们逐渐认可淀粉样蛋白假说,即认为淀粉样蛋白沉积是AD最初起因。研究人员正在寻找针对淀粉样β蛋白沉积的药物,雌激素是其中之一。初步的工作证明,雌激素能够调节淀粉样β蛋白前体代谢,减少淀粉样β蛋白生成,也能够减轻淀粉样B蛋白引起的免疫炎症反应、氧应激对细胞造成的损伤,和对抗细胞凋亡。  相似文献   

7.
Cole GM 《Neuron》2003,37(6):889-890
In this issue of Neuron, Kaur et al. demonstrate that iron chelation by ferritin transgene or the metal chelator clioquinol prevent oxidative damage and MPTP toxicity in mice. This raises the issue of specific iron chelators or clioquinol for control of oxidative damage in Parkinson's, Alzheimer's, and other neurodegenerative diseases, but not without safety concerns.  相似文献   

8.
Oxidative injury of the root elongation zone is a primary event in aluminum (Al) toxicity in plants, but the injuring species remain unidentified. We verified the hypothesis that lipid peroxide-derived aldehydes, especially highly electrophilic α,β-unsaturated aldehydes (2-alkenals), participate in Al toxicity. Transgenic tobacco (Nicotiana tabacum) overexpressing Arabidopsis (Arabidopsis thaliana) 2-alkenal reductase (AER-OE plants), wild-type SR1, and an empty vector-transformed control line (SR-Vec) were exposed to AlCl3 on their roots. Compared with the two controls, AER-OE plants suffered less retardation of root elongation under AlCl3 treatment and showed more rapid regrowth of roots upon Al removal. Under AlCl3 treatment, the roots of AER-OE plants accumulated Al and H2O2 to the same levels as did the sensitive controls, while they accumulated lower levels of aldehydes and suffered less cell death than SR1 and SR-Vec roots. In SR1 roots, AlCl3 treatment markedly increased the contents of the highly reactive 2-alkenals acrolein, 4-hydroxy-(E)-2-hexenal, and 4-hydroxy-(E)-2-nonenal and other aldehydes such as malondialdehyde and formaldehyde. In AER-OE roots, accumulation of these aldehydes was significantly less. Growth of the roots exposed to 4-hydroxy-(E)-2-nonenal and (E)-2-hexenal were retarded more in SR1 than in AER-OE plants. Thus, the lipid peroxide-derived aldehydes, formed downstream of reactive oxygen species, injured root cells directly. Their suppression by AER provides a new defense mechanism against Al toxicity.Aluminum (Al) is the most abundant metal in the earth''s crust and is a major factor limiting plant growth and productivity in acid soils, which cover about 50% of the world''s potentially arable land surface (Kochian, 1995; Kochian et al., 2004). The primary site of Al accumulation and toxicity is the root meristem, and inhibition of root elongation is the most notable symptom of Al toxicity (Delhaize and Ryan, 1995; Yamamoto et al., 2003). Al causes various adverse effects, such as disruption of signal transduction pathways, inhibition of cell division and ion fluxes, disruption of cytoskeletal dynamics, induced generation of reactive oxygen species (ROS), and disturbance of plasma membrane stability and function (Jones and Kochian, 1995; Blancaflor et al., 1998; Yamamoto et al., 2001, 2002; Kochian et al., 2004; Ma et al., 2007). Of all these toxic effects, the generation of ROS is observed rapidly and sustainably in roots after Al exposure. Al-induced generation of ROS has been shown in maize (Zea mays) and Allium cepa roots (Jones et al., 2006; Achary et al., 2008). Tahara et al. (2008) showed that ROS generated to a greater degree in Al-sensitive species than in Al-tolerant species. Yamamoto et al. (2002, 2003) have shown a correlation between ROS level and inhibition of growth capacity in cultured tobacco (Nicotiana tabacum) cells. Furthermore, ROS generation increases with increasing Al concentration (Achary et al., 2008; Xue et al., 2008). Generation of ROS appears to be a cause, rather than a result, of Al-induced cell injury, because high ROS scavenging ability resulted in enhanced Al tolerance (Devi et al., 2003; Ezaki et al., 2008). In addition, overexpression of genes encoding antioxidant enzymes (peroxidase and superoxide dismutase) conferred Al tolerance to the transgenic plants (Ezaki et al., 2000; Basu et al., 2001). Thus, ROS appears to be the primary factors that cause growth inhibition in Al-stressed roots.Downstream of ROS generation, lipid peroxidation is a common symptom of Al toxicity (Yamamoto et al., 2001), and it increases with increasing Al concentration (Achary et al., 2008). From animal cell studies, it is now recognized that the toxicity of lipid peroxide (LOOH) is largely ascribable to LOOH-derived aldehydes. In particular, α,β-unsaturated aldehydes, such as 4-hydroxy-(E)-2-nonenal (HNE) and acrolein, are strong electrophiles and readily modify proteins and nucleic acids (Esterbauer et al., 1991; Taylor et al., 2002; O''Brien et al., 2005; Møller et al., 2007). HNE causes depletion of glutathione, a decrease in protein thiols, disturbance of calcium homeostasis, inhibition of DNA, RNA, and protein synthesis, lactate release, morphological changes of cells, and finally leading to cell death (Esterbauer et al., 1991; Burcham, 1998). Increase of HNE has been observed in a wide range of human diseases, including Alzheimer''s disease, Parkinson''s disease, and mitochondrial complex 1 deficiency (Poli and Schaur, 2000).In plants, too, a close correlation between the level of LOOH-derived aldehydes (determined as thiobarbituric acid-reactive substances [TBARS]) and cellular damage has been shown under environmental stresses caused by heat, chilling, UV-B radiation, salinity, heavy metals, and Al (Ma et al., 2007; Ezaki et al., 2008). Their involvement in cellular damage has been demonstrated by the protective effects of the aldehyde-scavenging enzymes aldehyde dehydrogenase (Sunkar et al., 2003; Kotchoni et al., 2006) and aldehyde reductase (Oberschall et al., 2000; Hideg et al., 2003; Hegedüs et al., 2004) to confer tolerance against various environmental stresses when they were overexpressed in plants. In barley (Hordeum vulgare) roots, the formation of HNE in association with Al treatment was observed (Sakihama and Yamasaki, 2002). Occurrence of HNE in Arabidopsis (Arabidopsis thaliana) leaves under oxidative stress has been also deduced by detection of modified proteins in the mitochondria (Winger et al., 2007). HNE rapidly inhibited respiration in isolated potato (Solanum tuberosum) mitochondria by inactivating pyruvate dehydrogenase, 2-oxoglutarate dehydrogenase, NAD-malic enzyme (Millar and Leaver, 2000), and alternative oxidase (Winger et al., 2005). HNE and other 2-alkenals also inactivated photosynthesis in isolated chloroplasts (Mano et al., 2009). Arabidopsis contains 2-alkenal reductase (AER; E.C. 1.3.1.74) that catalyzes the reduction of the α,β-unsaturated bond of 2-alkenals to produce n-alkanals (Mano et al., 2002). Overexpression of AER in tobacco (Mano et al., 2005) and in Arabidopsis (Papdi et al., 2008) improved the tolerance to photooxidative stress and NaCl stress, respectively. Thus, accumulated observation indicates that LOOH-derived aldehydes, especially 2-alkenals, are commonly involved in oxidative damage in plant cells. Considering the critical importance of ROS in Al toxicity to roots, it is expected that 2-alkenals are produced and mediate damage in the stressed root cells.To evaluate the roles of LOOH-derived aldehydes in root injury under Al stress, we employed transgenic tobacco plants that overexpress the AER gene (AER-OE plants; Mano et al., 2005). With Al treatment, the roots of AER-OE accumulated Al and H2O2 to the same levels as those of the wild type, but they showed resistance to inhibition of elongation. Aldehyde analysis revealed that the Al treatment increased the contents of several toxic aldehydes, including HNE and acrolein in wild-type plants, but these aldehydes were significantly suppressed in the AER-OE plants. On the basis of these results, we propose that the inhibition of root growth by Al ions is induced by toxic aldehydes generated with ROS.  相似文献   

9.
10.
微生物铝毒和耐铝机制的研究现状   总被引:3,自引:0,他引:3  
铝是地球上含量最为丰富的金属元素 ,在酸性条件下 ,主要以Al3 存在。Al3 作为一种严重的环境毒剂 ,已经在众多模式生物中所证明。近年来 ,许多生物学家已日益注意到铝毒和耐铝性在环境科学与生命科学领域的重要性。结合研究工作 ,综述了微生物铝毒害和耐铝的机制。微生物通过①增强分泌有机酸与Al3 螯合 ,②超表达Mg2 通道蛋白 ,增强细胞转运吸收Mg2 ,③通过线粒体ATPase和液泡ATPase协同作用将Al3 隔离于液泡内 ,以及④通过氧化胁迫改变、调节Al3 毒害和耐铝性 ,减缓Al3 对细胞的毒害。  相似文献   

11.
Aluminum (Al), a known environmental toxicant, has been linked to a variety of pathological conditions such as dialysis dementia, osteomalacia, Alzheimer's disease, and Parkinson's disease. However, its precise role in the pathogenesis of these disorders is not fully understood. Using hepatocytes as a model system, we have probed the impact of this trivalent metal on the aerobic energy-generating machinery. Here we show that Al-exposed hepatocytes were characterized by lipid and protein oxidation and a dysfunctional tricarboxylic acid (TCA) cycle. BN-PAGE, SDS-PAGE, and Western blot analyses revealed a marked decrease in activity and expression of succinate dehydrogenase (SDH), alpha-ketoglutarate dehydrogenase (KGDH), isocitrate dehydrogenase-NAD+ (IDH), fumarase (FUM), aconitase (ACN), and cytochrome c oxidase (Cyt C Ox). 13C-NMR and HPLC studies further confirmed the disparate metabolism operative in control and Al-stressed cells and provided evidence for the accumulation of succinate in the latter cultures. In conclusion, these results suggest that Al toxicity promotes a dysfunctional TCA cycle and impedes ATP production, events that may contribute to various Al-induced abnormalities.  相似文献   

12.
Studies of aluminum neurobehavioral toxicity in the intact mammal   总被引:5,自引:0,他引:5  
Summary 1. Aluminum (Al) has been implicated in neurotoxic syndromes in several conditions, including Alzheimer's disease (AD). The developmental stage of the mammalian brain most susceptible to Al was determined in rabbits systemically exposed to Al during the prenatal, postnatal, or second month or for 1 month as adults or as aged subjects. Eyeblink reflex classical conditioning showed an Al-induced learning deficit only in the adult and aged rabbits.2. 4-Aminopyridine, which was reported to improve learning in AD subjects, attenuated this Al-induced learning deficit.3. Conditioned eyeblink acquisition is slower in AD subjects than controls, supporting the Al-loaded rabbit as a model of some AD effects.4. To determine if the Al-loaded rabbit modeled the AD cholinergic deficit, acetylcholine (Ach) overflow was measured in rabbit hippocampus using microdialysis. Aluminum pretreatment reduced basal and potassium-stimulated Ach overflow compared to controls.5. Acetylcholine overflow increased as control rabbits acquired the conditioned eyeblink reflex, then subsequently decreased, although conditioned eyeblink performance continued. In contrast, Al-loaded rabbits showed a delay in conditioned eyeblink acquisition and greatly attenuated Ach overflow. The Al-induced attenuation of Ach overflow may contribute to the Al-induced learning deficit.6. Brain Al entry was studied using microdialysis of blood, brain, and lateral ventricle. Aluminum rapidly entered the brain and lateral ventricle. Frontal cortical Al was greater than lateral ventricular Al, suggesting that Al primarily enters the brain through the cerebral microvasculature.7. The brain/blood Al ratio was always significantly less than 1. This ratio was influenced by the Al form administered, brain site and animal species. Thus, there appears to be an active process moving Al out of brain extracellular fluid (ECF).8. Brain and blood dialysate Ach concentrations were not different after cyanide addition to the dialysate, supporting the conclusion that an active process moves Al out of brain ECF.  相似文献   

13.
We have previously reported that amyloid Abeta, the major component of senile plaques in Alzheimer's disease (AD), binds Cu with high affinity via histidine and tyrosine residues [Atwood, C. S., et al. (1998) J. Biol. Chem. 273, 12817-12826; Atwood, C. S., et al. (2000) J. Neurochem. 75, 1219-1233] and produces H(2)O(2) by catalyzing the reduction of Cu(II) or Fe(III) [Huang, X., et al. (1999) Biochemistry 38, 7609-7616; Huang, X., et al. (1999) J. Biol. Chem. 274, 37111-37116]. Incubation with Cu induces the SDS-resistant oligomerization of Abeta [Atwood, C. S., et al. (2000) J. Neurochem. 75, 1219-1233], a feature characteristic of neurotoxic soluble Abeta extracted from the AD brain. Since residues coordinating Cu are most vulnerable to oxidation, we investigated whether modifications of these residues were responsible for Abeta cross-linking. SDS-resistant oligomerization of Abeta caused by incubation with Cu was found to induce a fluorescence signal characteristic of tyrosine cross-linking. Using ESI-MS and a dityrosine specific antibody, we confirmed that Cu(II) (at concentrations lower than that associated with amyloid plaques) induces the generation of dityrosine-cross-linked, SDS-resistant oligomers of human, but not rat, Abeta peptides. The addition of H2O2 strongly promoted Cu-induced dityrosine cross-linking of Abeta1-28, Abeta1-40, and Abeta1-42, suggesting that the oxidative coupling is initiated by interaction of H2O2 with a Cu(II) tyrosinate. The dityrosine modification is significant since it is highly resistant to proteolysis and is known to play a role in increasing structural strength. Given the elevated concentration of Cu in senile plaques, our results suggest that Cu interactions with Abeta could be responsible for causing the covalent cross-linking of Abeta in these structures.  相似文献   

14.
A growing body of evidence supports a central role for biometals in neurodegenerative disorders. Biometals induce oxidative stress through the generation of reactive oxygen species and contribute to neuronal cell dysfunction in Alzheimer's disease (AD), prion disorders and Parkinson's disease (PD). Therapies based on modulation of biometal metabolism are currently being developed and the metal ligand, 5-chloro-7-iodo-8-hydroxyquinoline (clioquinol or CQ) has been investigated for the treatment of AD. CQ has also shown therapeutic benefits in an animal model of PD. However, little is known about the neuroprotective processes of CQ in vivo. In this study, we examined the effect of CQ in BE(2)-M17 human neuroblastoma cells exposed to increased oxidative stress (hydrogen peroxide (H2O2) treatment). Although CQ alone induced a moderate toxic effect on cells, when added to H2O2-treated M17 cells, CQ induced a significant inhibition of H2O2 toxicity. This correlated with up-regulation of phosphoinositol-3-kinase (PI3K) activity in CQ-treated cells. The protective action of CQ was not observed in murine N2a neuroblastoma cells treated with H2O2 and this cell-line did not reveal CQ-mediated increases in PI3K activation. The protective effect was specific for CQ and was not induced by a number of different metal ligands. Inhibition of PI3K activity with LY294002 prevented CQ protection against H2O2 toxicity, demonstrating a crucial role for CQ activation of PI3K in protection against oxidative stress. Furthermore, CQ inhibited H2O2-mediated up-regulation of p53 activity in the M17 cells and this was dependent on PI3K activation. Our studies demonstrate that in human M17 cells, CQ can protect against oxidative stress by activating the PI3K-dependent survival pathway and blocking p53-mediated cell death. These findings have important implications for the development of protective metal ligand-based therapies for treatment of disorders involving oxidative stress.  相似文献   

15.
Aluminum (Al), oxidative stress and impaired cholinergic functions have all been related to Alzheimer's disease (AD). The present study evaluates the effect of aluminum on acetylcholinesterase (AChE) and lipid peroxidation in the mouse brain. Mice were loaded by gavage with Al 0.1 mmol/kg/day 5 days per week during 12 weeks. The mice were divided into four groups: (1) control; (2) 10 mg/mL of citrate solution; (3) 0.1 mmol/kg of Al solution; (4) 0.1 mmol/kg of Al plus 10 mg/mL of citrate solution. AChE activity was determined in the hippocampus, striatum, cortex, hypothalamus and cerebellum and lipid peroxidation was determined in the hippocampus, striatum and cortex. An increase of AChE activity was observed in the fourth group (Al + Ci) in the hippocampus (36%), striatum (54%), cortex (44%) and hypothalamus (22%) (p<0.01). The third group (Al) presented a decrease of AChE activity in the hypothalamus (20%) and an enhancement in the striatum (27%). Lipid peroxidation, measured by TBARS (thiobarbituric acid reactive substances), was elevated in the hippocampus and cerebral cortex when compared with the control (p < 0.01). The effect of aluminum on AChE activity may be due to a direct neurotoxic effect of the metal or perhaps a disarrangement of the plasmatic membrane caused by increased lipid peroxidation.  相似文献   

16.
A growing body of evidence suggests a relationship between oxidative stress and beta-amyloid (Abeta) peptide accumulation, a hallmark in the pathogenesis of Alzheimer's disease (AD). However, a direct causal relationship between oxidative stress and Abeta pathology has not been established in vivo. Therefore, we crossed mice with a knockout of one allele of manganese superoxide dismutase (MnSOD), a critical antioxidant enzyme, with Tg19959 mice, which overexpress a doubly mutated human beta-amyloid precursor protein (APP). Partial deficiency of MnSOD, which is well established to cause elevated oxidative stress, significantly increased brain Abeta levels and Abeta plaque burden in Tg19959 mice. These results indicate that oxidative stress can promote the pathogenesis of AD and further support the feasibility of antioxidant approaches for AD therapy.  相似文献   

17.
Following our previous finding that the sulfhydryl-oxidising chemical diamide induced a marked elevation of cellular Al(3+) (Wu et al., Int J Mol Sci, 12:8119-8132, 2011), a further investigation into the underlying molecular mechanism was carried out, using the eukaryotic model organism Saccharomyces cerevisiae. The effects of non-toxic dose of diamide (0.8?mM) and a mild dose of aluminium sulphate (Al(3+)) (0.4?mM) were determined prior to the screening of gene deletion mutants. A total of 81 deletion mutants were selected for this study according to the available screening data against Al(3+) only (Kakimoto et al., BioMetals, 18: 467-474, 2005) and diamide only (Thorpe et al., Proc Natl Acad Sci USA, 101: 6564-6569, 2004). On the basis of our screening data and the cluster analysis, a cluster containing the gene deletions (rpe1?, sec72?, pdr5? and ric1?) was found to be specifically sensitive to the mixture of diamide and Al(3+). However gnp1?, mch5? and ccc1? mutants were resistant. Dithiothreitol (DTT) and ascorbate markedly reversed the diamide-induced Al(3+) toxicity. Inductively-coupled plasma optical emission spectrometry demonstrated that DTT reduced the intracellular Al(3+) content in diamide/Al(3+)-treated yeast cells six-fold compared to the non-DTT controls. These data together revealed that the pleiotropic drug resistance transporter (Pdr5p) and vacuolar/vesicular transport-related proteins (Ric1p and Sec72p) are the targets of diamide. A dysfunctional membrane-bound Pdr5p terminates the detoxification pathway for Al(3+) at the final step, leading to intracellular Al(3+) accumulation and hence toxicity. As Al(3+) toxicity has been a problem in agriculture and human health, this study has provided a significant step forward in understanding Al(3+) toxicity.  相似文献   

18.
Alzheimer's disease (AD) is a late-onset dementia that is characterized by the loss of memory and an impairment of multiple cognitive functions. Advancements in molecular, cellular, and animal model studies have revealed that the formation of amyloid beta (Abeta) and other derivatives of the amyloid precursor protein (APP) are key factors in cellular changes in the AD brain, including the generation of free radicals, oxidative damage, and inflammation. Recent molecular, cellular, and gene expression studies have revealed that Abeta enters mitochondria, induces the generation of free radicals, and leads to oxidative damage in post-mortem brain neurons from AD patients and in brain neurons from cell models and transgenic mouse models of AD. In the last three decades, tremendous progress has been made in mitochondrial research and has provided significant findings to link mitochondrial oxidative damage and neurodegenerative diseases such as AD. Researchers in the AD field are beginning to recognize the possible involvement of a mutant APP and its derivatives in causing mitochondrial oxidative damage in AD. This article summarizes the latest research findings on the generation of free radicals in mitochondria and provides a possible model that links Abeta proteins, the generation of free radicals, and oxidative damage in AD development and progression.  相似文献   

19.
Prostate cancer is one of the most common malignancies.The development and progression of prostate cancer are driven by a series of genetic and epigenetic events including gene amplification that activates oncogenes and chromosomal deletion that inactivates tumor suppressor genes.Whereas gene amplification occurs in human prostate cancer,gene deletion is more common,and a large number of chromosomal regions have been identified to have frequent deletion in prostate cancer,suggesting that tumor suppressor inactivation is more common than oncogene activation in prostatic carcinogenesis (Knuutila et al.,1998,1999;Dong,2001).Among the most frequently deleted chromosomal regions in prostate cancer,target genes such as NKX3-1 from 8p21,PTENfrom 10q23 andATBF1 from 16q22 have been identified by different approaches (He et al.,1997;Li et al.,1997;Sun et al.,2005),and deletion of these genes in mouse prostates has been demonstrated to induce and/or promote prostatic carcinogenesis.For example,knockout of Nkx3-1 in mice induces hyperplasia and dysplasia (Bhatia-Gaur et al.,1999;Abdulkadir et al.,2002) and promotes prostatic tumorigenesis (Abate-Shen et al.,2003),while knockout of Pten alone causes prostatic neoplasia (Wang et al.,2003).Therefore,gene deletion plays a causal role in prostatic carcinogenesis (Dong,2001).  相似文献   

20.
Iron (Fe) and aluminum (Al) have been implicated in the pathogenesis of Alzheimer's disease (AD). In this study, we examined neuronal and glial cells to clarify which contributes most to metal accumulation after internalization through the transferrin-independent iron uptake (Tf-IU) systems in primary neuronal and glial predominant (NP and GP) cells from rat cerebral cortex, which affect the accumulation of transition metals in a variety of cultured cells. Al more significantly upregulated the Tf-IU activity in GP cells than in NP cells. GP cells were more resistant to Fe and Al exposure than NP cells. However, a chemiluminescence analysis specific for reactive oxygen species (ROS) showed that ROS levels in Fe- or Al-loaded NP cells were twice as high as in Fe- or Al-loaded GP cells. Northern blot analysis and gel retardation assay showed that the Al and Fe exposure taken up by the cells suppress Tf receptor mRNA expression to a greater extent in GP than NP cells, indicating that Al and Fe more markedly accumulate in glial than in neuronal cells. These results suggest that glial cells rather than neuronal cells contribute to the metal accumulation and are more resistant to oxidative stress caused by metals than neuronal cells. The present study may help to explain the pathogenesis of neurodegeneration in AD disorders caused by metal-generated oxidative stress.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号