Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Sci Adv ; 6(46)2020 11.
Article in English | MEDLINE | ID: mdl-33188020

ABSTRACT

Neurodegenerative disorders are caused by progressive neuronal loss, and there is no complete treatment available yet. Neuroinflammation is a common feature across neurodegenerative disorders and implicated in the progression of neurodegeneration. Dysregulated activation of microglia causes neuroinflammation and has been highlighted as a treatment target in therapeutic strategies. Here, we identified novel therapeutic candidate ALGERNON2 (altered generation of neurons 2) and demonstrate that ALGERNON2 suppressed the production of proinflammatory cytokines and rescued neurodegeneration in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced Parkinson's disease model. ALGERNON2 stabilized cyclinD1/p21 complex, leading to up-regulation of nuclear factor erythroid 2-related factor 2 (Nrf2), which contributes to antioxidative and anti-inflammatory responses. Notably, ALGERNON2 enhanced neuronal survival in other neuroinflammatory conditions such as the transplantation of induced pluripotent stem cell-derived dopaminergic neurons into murine brains. In conclusion, we present that the microglial potentiation of the p21-Nrf2 pathway can contribute to neuronal survival and provide novel therapeutic potential for neuroinflammation-triggered neurodegeneration.


Subject(s)
Microglia , Neurodegenerative Diseases , Animals , Disease Models, Animal , Dopaminergic Neurons/metabolism , Mice , Mice, Inbred C57BL , Microglia/metabolism , NF-E2-Related Factor 2/metabolism , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/therapy , Neuroinflammatory Diseases
3.
BJU Int ; 90(4): 462-6, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12175409

ABSTRACT

OBJECTIVE: To investigate the relationship between the expression of the 150-kDa oxygen-regulated protein (ORP150, which functions as a molecular chaperone in the endoplasmic reticulum for the folding and trafficking of newly synthesized proteins) and the aggressiveness of bladder cancer, and the expression of vascular endothelial growth factor (VEGF) and matrix metalloproteinases (MMPs), as the former is a secreting protein through the endoplasmic reticulum and the latter are closely involved in tumour invasion. MATERIALS AND METHODS: Thirty-nine cystectomy specimens, comprising 12 superficial (pT1) and 27 invasive (pT2-pT4) tumours, were immunohistochemically analysed using antibodies against ORP150, VEGF, MMP-1, MMP-2 and MMP-9. Staining was scored from 0 to 3, according to the ratio of positively staining cells. RESULTS: Staining was positive (score 1-3) for ORP150 in 10 of 12 superficial and 25 (93%) of the invasive tumours, with a significantly higher staining score for stage T4 than stage T1 tumours. The trend was the same for the staining score of MMP-2, and there was a significant correlation between ORP150 and MMP-2 expression. CONCLUSIONS: The expression of ORP150 was common in bladder cancer, with a tendency for greater expression in higher stages. The significant correlation between ORP150 and MMP-2 expression suggests that ORP150 acts as a molecular chaperone for MMP-2 secretion and thus tumour invasion.


Subject(s)
Proteins/metabolism , Urinary Bladder Neoplasms/diagnosis , Endothelial Growth Factors/metabolism , HSP70 Heat-Shock Proteins , Humans , Immunohistochemistry , Lymphokines/metabolism , Matrix Metalloproteinases/metabolism , Molecular Chaperones/metabolism , Neoplasm Invasiveness , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
4.
Mol Urol ; 5(2): 79-80, 2001.
Article in English | MEDLINE | ID: mdl-11690552

ABSTRACT

Heat shock proteins (HSPs)/stress proteins are molecular chaperones that are induced by various environmental and physiological stimuli. Evidence of the relations between the expression of HSPs and the regulation of cell growth or transformation has accumulated. The 150-kDa oxygen-regulated protein (ORP150), a new member of HSP family, functions as a molecular chaperone in the endoplasmic reticulum. We have examined whether transduced antisense ORP150 cDNA reduces tumorigenicity and angiogenicity. Relations between these stress proteins and cancer and possibilities for anticancer gene therapy are described.


Subject(s)
Prostatic Neoplasms/metabolism , Proteins/metabolism , Endothelial Growth Factors/genetics , Endothelial Growth Factors/metabolism , Genetic Therapy , Glucose/metabolism , HSP70 Heat-Shock Proteins , Humans , Lymphokines/genetics , Lymphokines/metabolism , Male , Molecular Chaperones/metabolism , Oligodeoxyribonucleotides, Antisense/metabolism , Prostatic Neoplasms/therapy , Proteins/genetics , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
5.
J Clin Invest ; 108(10): 1439-50, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11714735

ABSTRACT

A series of events initiated by glutamate-receptor interaction perturbs cellular homeostasis resulting in elevation of intracellular free calcium and cell death. Cells subject to such environmental change express stress proteins, which contribute importantly to maintenance of metabolic homeostasis and viability. We show that an inducible chaperone present in endoplasmic reticulum (ER), the 150-kDa oxygen-regulated protein (ORP150), is expressed both in the human brain after seizure attack and in mouse hippocampus after kainate administration. Using mice heterozygous for ORP150 deficiency, exposure to excitatory stimuli caused hippocampal neurons to display exaggerated elevation of cytosolic calcium accompanied by activation of mu-calpain and cathepsin B, as well as increased vulnerability to glutamate-induced cell death in vitro and decreased survival to kainate in vivo. In contrast, targeted neuronal overexpression of ORP150 suppressed each of these events and enhanced neuronal and animal survival in parallel with diminished seizure intensity. Studies using cultured hippocampal neurons showed that ORP150 regulates cytosolic free calcium and activation of proteolytic pathways causing cell death in neurons subject to excitatory stress. Our data underscore a possible role for ER stress in glutamate toxicity and pinpoint a key ER chaperone, ORP150, which contributes to the stress response critical for neuronal survival.


Subject(s)
Endoplasmic Reticulum/metabolism , Glutamic Acid/toxicity , Hippocampus/drug effects , Molecular Chaperones/metabolism , Neurons/drug effects , Proteins/metabolism , Animals , HSP70 Heat-Shock Proteins , Heterozygote , Hippocampus/cytology , Hippocampus/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Chaperones/genetics , Neurons/metabolism , Proteins/genetics
6.
J Clin Invest ; 108(1): 41-50, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11435456

ABSTRACT

Expression of angiogenic factors such as VEGF under conditions of hypoxia or other kinds of cell stress contributes to neovascularization during wound healing. The inducible endoplasmic reticulum chaperone oxygen-regulated protein 150 (ORP150) is expressed in human wounds along with VEGF. Colocalization of these two molecules was observed in macrophages in the neovasculature, suggesting a role of ORP150 in the promotion of angiogenesis. Local administration of ORP150 sense adenovirus to wounds of diabetic mice, a treatment that efficiently targeted this gene product to the macrophages of wound beds, increased VEGF antigen in wounds and accelerated repair and neovascularization. In cultured human macrophages, inhibition of ORP150 expression caused retention of VEGF antigen within the endoplasmic reticulum (ER), while overexpression of ORP150 promoted the secretion of VEGF into hypoxic culture supernatants. Taken together, these data suggest an important role for ORP150 in the setting of impaired wound repair and identify a key, inducible chaperone-like molecule in the ER. This novel facet of the angiogenic response may be amenable to therapeutic manipulation.


Subject(s)
Cell Hypoxia/physiology , Endothelial Growth Factors/physiology , Lymphokines/physiology , Molecular Chaperones/physiology , Neovascularization, Physiologic/physiology , Proteins/physiology , Transcription Factors , Adenoviridae/genetics , Animals , Cells, Cultured , Culture Media, Conditioned , DNA-Binding Proteins/physiology , Diabetes Complications , Diabetes Mellitus/genetics , Endoplasmic Reticulum/metabolism , Endothelial Growth Factors/biosynthesis , Endothelial Growth Factors/genetics , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Female , Fibroblast Growth Factor 2/physiology , Gene Expression Regulation/drug effects , Genetic Therapy , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , HSP70 Heat-Shock Proteins , Humans , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Lymphokines/biosynthesis , Lymphokines/genetics , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Neovascularization, Pathologic/physiopathology , Nuclear Proteins/physiology , Oxygen/pharmacology , Protein Transport , Proteins/genetics , RNA, Antisense/pharmacology , RNA, Messenger/metabolism , Rats , Recombinant Fusion Proteins/physiology , Single-Blind Method , Skin/blood supply , Skin/injuries , Transforming Growth Factor beta/physiology , Transforming Growth Factor beta1 , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Wound Healing/physiology
7.
Am J Pathol ; 158(5): 1685-94, 2001 May.
Article in English | MEDLINE | ID: mdl-11337366

ABSTRACT

RA301/Tra2beta, a sequence-specific RNA-binding protein, was first cloned as a stress molecule in re-oxygenated astrocytes. In human vascular tissues, we have found enhanced RA301/Tra2beta expression in coronary artery with intimal thickening, and atherosclerotic aorta. Balloon injury to the rat carotid artery induced RA301/Tra2beta transcripts followed by expression of the antigen, which was detected in medial and neointimal vascular smooth muscle cells (VSMCs). In cultured VSMCs, hypoxia/re-oxygenation caused induction of RA301/Tra2beta and was accompanied by cell proliferation, both of which were blocked by the addition of either diphenyl iodonium, a NADPH oxidase inhibitor, PD98059, a mitogen-activated protein kinase kinase inhibitor, or antisense oligonucleotide for RA301/Tra2beta. Consistent with a link between RA301/Tra2beta and cell proliferation, platelet-derived growth factor also induced expression of RA301/Tra2beta in cultured VSMCS: These data suggest a possible role for RA301/Tra2beta in the regulation of VSMC proliferation, especially in the setting of hypoxia/re-oxygenation-induced cell stress.


Subject(s)
Carotid Arteries/metabolism , Muscle, Smooth, Vascular/cytology , Nerve Tissue Proteins/genetics , RNA-Binding Proteins/genetics , Adolescent , Adult , Aged , Animals , Biphenyl Compounds/pharmacology , Carotid Arteries/pathology , Cell Division/drug effects , Cell Division/physiology , DNA, Antisense/pharmacology , Dose-Response Relationship, Drug , Female , Flavonoids/pharmacology , Gene Expression , Humans , Infant , Male , Middle Aged , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Nerve Tissue Proteins/physiology , Onium Compounds/pharmacology , Oxygen/pharmacology , RNA Splicing , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/physiology , Rats , Rats, Sprague-Dawley , Serine-Arginine Splicing Factors , Superoxides/metabolism
8.
Cancer Res ; 61(10): 4206-13, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11358846

ABSTRACT

Expression of angiogenic factors such as vascular endothelial growth factor (VEGF) under conditions of cell stress involves both transcriptional and translational events, as well as an important role for inducible endoplasmic reticulum (ER) chaperones. Coexpression of VEGF and 150-kDa oxygen-regulated protein (ORP), a novel ER chaperone, in human glioblastoma suggested a link between angiogenesis and ORP150. C6 glioma cells stably transfected with ORP150 antisense displayed selectively reduced ORP150 expression. Tumors raised after inoculation of immunocompromised mice with ORP150 antisense C6 glioma transfectants demonstrated an initial phase of growth comparable to wild-type C6 glioma cells which was followed by marked regression within 8 days. Decreased density of platelet/endothelial cell adhesion molecule 1-positive structures within the tumor bed was consistent with reduced angiogenesis in C6 gliomas expressing ORP150 antisense, compared with tumors derived from C6 cells overexpressing ORP150 sense or vector controls. In vitro, inhibition of ORP150 expression decreased release of VEGF into culture supernatants; in ORP150 antisense transfectants, VEGF accumulated intracellularly within the ER. These findings demonstrate a critical role for the inducible ER chaperone ORP150 in tumor-mediated angiogenesis via processing of VEGF, and, thus, highlight a new facet of angiogenic mechanisms amenable to therapeutic manipulation in tumors.


Subject(s)
Endoplasmic Reticulum/metabolism , Neovascularization, Pathologic/metabolism , Proteins/physiology , Animals , Autoantibodies/biosynthesis , Autoantibodies/blood , Cell Division/physiology , Cell Hypoxia/physiology , DNA, Antisense/genetics , Endothelial Growth Factors/biosynthesis , Endothelial Growth Factors/metabolism , Glioblastoma/blood supply , Glioblastoma/immunology , Glioblastoma/metabolism , Golgi Apparatus/metabolism , HSP70 Heat-Shock Proteins , Humans , Lymphokines/biosynthesis , Lymphokines/metabolism , Mice , Neovascularization, Pathologic/immunology , Protein Biosynthesis , Protein Processing, Post-Translational/physiology , Proteins/genetics , Rats , Transfection , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
9.
Nat Med ; 7(3): 317-23, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11231630

ABSTRACT

Oxygen-regulated protein 150 kD (ORP150) is a novel endoplasmic-reticulum-associated chaperone induced by hypoxia/ischemia. Although ORP150 was sparingly upregulated in neurons from human brain undergoing ischemic stress, there was robust induction in astrocytes. Cultured neurons overexpressing ORP150 were resistant to hypoxemic stress, whereas astrocytes with inhibited ORP150 expression were more vulnerable. Mice with targeted neuronal overexpression of ORP150 had smaller strokes compared with controls. Neurons with increased ORP150 demonstrated suppressed caspase-3-like activity and enhanced brain-derived neurotrophic factor (BDNF) under hypoxia signaling. These data indicate that ORP150 is an integral participant in ischemic cytoprotective pathways.


Subject(s)
Cell Death/physiology , Cell Hypoxia , Neurons/pathology , Proteins/physiology , Animals , Brain/cytology , Brain/metabolism , Brain-Derived Neurotrophic Factor/metabolism , HSP70 Heat-Shock Proteins , Humans , Mice , Neurons/metabolism , Proteins/antagonists & inhibitors
10.
J Biol Chem ; 275(35): 27100-9, 2000 Sep 01.
Article in English | MEDLINE | ID: mdl-10869339

ABSTRACT

Amyloid beta-peptide-binding alcohol dehydrogenase (ABAD) is a member of the family of short chain dehydrogenase/reductases whose distinctive properties include the capacity to bind amyloid beta-peptide and enzymatic activity toward a broad array of substrates including n-isopropanol and beta-estradiol. In view of the wide substrate specificity of ABAD and its high activity on l-beta-hydroxyacyl-CoA derivatives, we asked whether it might also catalyze the oxidation of the ketone body d-3-hydroxybutyrate. This was indeed the case, and oxidation proceeded with K(m) of approximately 4.5 mm and V(max) of approximately 4 nmol/min/mg protein. When placed in medium with d-beta-hydroxybutyrate as the principal energy substrate, COS cells stably transfected to overexpress wild-type ABAD (COS/wtABAD) better maintained 3-(4, 5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide reduction, cellular energy charge, and morphologic phenotype compared with COS/vector cells. Using a severe model of metabolic perturbation, transgenic mice with targeted neuronal expression of ABAD subjected to transient middle cerebral artery occlusion showed strokes of smaller volume and lower neurologic deficit scores in parallel with increased brain ATP and decreased lactate, compared with nontransgenic controls. These data suggest that ABAD contributes to the protective response to metabolic stress, especially in the setting of ischemia.


Subject(s)
Alcohol Dehydrogenase/metabolism , Amyloid beta-Peptides/metabolism , Oxidative Stress , Peptide Fragments/metabolism , 3-Hydroxybutyric Acid/metabolism , Alcohol Dehydrogenase/genetics , Amyloid beta-Peptides/genetics , Animals , COS Cells , Magnetic Resonance Spectroscopy , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Peptide Fragments/genetics , Stroke/metabolism , Up-Regulation
11.
Nature ; 405(6784): 354-60, 2000 May 18.
Article in English | MEDLINE | ID: mdl-10830965

ABSTRACT

The receptor for advanced glycation end products (RAGE), a multi-ligand member of the immunoglobulin superfamily of cell surface molecules, interacts with distinct molecules implicated in homeostasis, development and inflammation, and certain diseases such as diabetes and Alzheimer's disease. Engagement of RAGE by a ligand triggers activation of key cell signalling pathways, such as p21ras, MAP kinases, NF-kappaB and cdc42/rac, thereby reprogramming cellular properties. RAGE is a central cell surface receptor for amphoterin, a polypeptide linked to outgrowth of cultured cortical neurons derived from developing brain. Indeed, the co-localization of RAGE and amphoterin at the leading edge of advancing neurites indicated their potential contribution to cellular migration, and in pathologies such as tumour invasion. Here we demonstrate that blockade of RAGE-amphoterin decreased growth and metastases of both implanted tumours and tumours developing spontaneously in susceptible mice. Inhibition of the RAGE-amphoterin interaction suppressed activation of p44/p42, p38 and SAP/JNK MAP kinases; molecular effector mechanisms importantly linked to tumour proliferation, invasion and expression of matrix metalloproteinases.


Subject(s)
Carrier Proteins/physiology , High Mobility Group Proteins/physiology , MAP Kinase Signaling System , Neoplasm Invasiveness , Neoplasm Metastasis , Receptors, Immunologic/physiology , Animals , Bromodeoxyuridine/metabolism , Carrier Proteins/antagonists & inhibitors , HMGB1 Protein , High Mobility Group Proteins/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Mice, Nude , Mice, SCID , Neoplasm Transplantation , Rats , Receptor for Advanced Glycation End Products , Receptors, Immunologic/antagonists & inhibitors , Receptors, Immunologic/genetics , Transfection , Tumor Cells, Cultured
12.
Am J Physiol Cell Physiol ; 278(6): C1172-82, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10837345

ABSTRACT

To assess the participation of the 150-kDa oxygen-regulated protein (ORP150) in protein transport, its function in Madin-Darby canine kidney (MDCK) cells was studied. Exposure of MDCK cells to hypoxia resulted in an increase of ORP150 antigen and increased binding of ORP150 to GP80/clusterin (80-kDa glycoprotein), a natural secretory protein in this cell line. In ORP150 antisense transformant MDCK cells, GP80 was retained within the endoplasmic reticulum after exposure to hypoxia. Metabolic labeling showed the delay of GP80 maturation in antisense transformants in hypoxia, whereas its matured form was detected in wild-type cells, indicating a role of ORP150 in protein transport, especially in hypoxia. The affinity chromatographic analysis of ORP150 suggested its ability to bind to ATP-agarose. Furthermore, the ATP hydrolysis analysis showed that ORP150 can release GP80 at a lower ATP concentration. These data indicate that ORP150 may function as a unique molecular chaperone in renal epithelial cells by facilitating protein transport/maturation in an environment where less ATP is accessible.


Subject(s)
Cell Hypoxia/physiology , Molecular Chaperones/physiology , Proteins/physiology , Adenosine Triphosphate/metabolism , Animals , Cell Line , Cell Survival , Clusterin , Dogs , Energy Metabolism , Glycoproteins/metabolism , HSP70 Heat-Shock Proteins , Humans , Kidney , Membrane Glycoproteins/metabolism , Oligodeoxyribonucleotides, Antisense/pharmacology , Proteins/genetics , Receptors, Interleukin-6 , Recombinant Proteins/metabolism , Transfection
14.
J Cell Biol ; 147(6): 1195-204, 1999 Dec 13.
Article in English | MEDLINE | ID: mdl-10601334

ABSTRACT

Application of differential display to cultured rat astrocytes subjected to hypoxia allowed cloning of a novel cDNA, termed stress-associated endoplasmic reticulum protein 1 (SERP1). Expression of SERP1 was enhanced in vitro by hypoxia and/or reoxygenation or other forms of stress, causing accumulation of unfolded proteins in endoplasmic reticulum (ER) stress, and in vivo by middle cerebral artery occlusion in rats. The SERP1 cDNA encodes a 66-amino acid polypeptide which was found to be identical to ribosome-associated membrane protein 4 (RAMP4) and bearing 29% identity to yeast suppressor of SecY 6 protein (YSY6p), suggesting participation in pathways controlling membrane protein biogenesis at ER. In cultured 293 cells subjected to ER stress, overexpression of SERP1/RAMP4 suppressed aggregation and/or degradation of newly synthesized integral membrane proteins, and subsequently, facilitated their glycosylation when the stress was removed. SERP1/RAMP4 interacted with Sec61alpha and Sec61beta, which are subunits of translocon, and a molecular chaperon calnexin. Furthermore, Sec61alpha and Sec61beta, but not SERP1/RAMP4, were found to associate with newly synthesized integral membrane proteins under stress. These results suggest that stabilization of membrane proteins in response to stress involves the concerted action of a rescue unit in the ER membrane comprised of SERP1/RAMP4, other components of translocon, and molecular chaperons in ER.


Subject(s)
Cell Hypoxia/physiology , Membrane Proteins/metabolism , Amino Acid Sequence , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Calcium-Binding Proteins/metabolism , Calnexin , Cell Line , Cloning, Molecular , Endoplasmic Reticulum/metabolism , Glycoproteins/biosynthesis , Glycoproteins/genetics , Glycoproteins/metabolism , Glycosylation , Homeostasis/physiology , Humans , Male , Membrane Proteins/biosynthesis , Membrane Proteins/chemistry , Membrane Proteins/genetics , Molecular Chaperones/metabolism , Molecular Sequence Data , Protein Binding , Protein Denaturation , Protein Renaturation , RNA, Messenger/analysis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , SEC Translocation Channels , Sequence Homology, Amino Acid
15.
J Neurochem ; 72(6): 2498-505, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10349860

ABSTRACT

Mutations in the presenilin-1 (PS-1) and presenilin-2 (PS-2) genes account for the majority of cases of early-onset familial Alzheimer's disease (AD). Alternative splicing forms of the PS-1 and PS-2 gene products have previously been reported in fibroblast and brain tissue from both familial and sporadic AD patients, as well as from normal tissues and cell lines. We demonstrate here unusual alternative splicing of the PS-2 gene that leads to the generation of mRNA lacking exon 5 in human brain tissue. This product was more frequently detected in brain tissue from sporadic AD patients (70.0%; 21 of 30) than from normal age-matched controls (17.6%; three of 17). In cultured neuroblastoma cells, this splice variant was generated in hypoxia but not under other forms of cellular stress. Hypoxia-mediated induction of this splice variant was blocked by pretreatment of neuroblastoma cells with the protein synthesis inhibitor cycloheximide or antioxidants such as N-acetylcysteine and diphenyl iodonium, suggesting that hypoxia-mediated oxidant stress might, at least in part, underlie the alternative splicing of PS-2 mRNA through de novo protein synthesis. Furthermore, the stable transfectants of this splice variant produced the N-terminal part of PS-2 protein (15 kDa) and were more susceptible to cellular stresses than control transfectants. These results suggest the possibility that altered presenilin gene products in stress conditions may also participate in the pathogenesis of AD.


Subject(s)
Alternative Splicing , Alzheimer Disease/genetics , Genetic Variation , Membrane Proteins/genetics , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Amino Acid Sequence , Apolipoprotein E4 , Apolipoproteins E/genetics , Base Sequence , Humans , Lipid Peroxidation , Membrane Proteins/biosynthesis , Middle Aged , Molecular Sequence Data , Neuroblastoma , Polymerase Chain Reaction , Presenilin-1 , Presenilin-2 , Reactive Oxygen Species/physiology , Recombinant Proteins/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Tumor Cells, Cultured
16.
Brain Res Mol Brain Res ; 74(1-2): 26-34, 1999 Dec 10.
Article in English | MEDLINE | ID: mdl-10640673

ABSTRACT

Based on the neurotrophic properties of astrocytes in response to ischemia, the current work focuses on the mechanism for cultured astrocytes to adapt to a hypoxic environment. Intracellular glucose levels in primary cultured rat astrocytes exposed to hypoxia fell by 30% within 24 h, in parallel with a decrease in glycogen stores. Glycolytic metabolism was crucial for cell survival during hypoxia, as 2-deoxyglucose resulted in rapid ATP depletion and cell death. The mechanism for maintaining glucose levels under these conditions appeared to be mobilization of glycogen stores, rather than increased extracellular uptake of glucose, as gluconolactone (an inhibitor of beta1-4 amyloglucosidase) induced a rapid fall in cellular ATP in cultures subjected to hypoxia, whereas cytochalasin B was without affect. Addition of cycloheximide diminished the viability of astrocytes in hypoxia, suggesting an obligatory role of de-novo gene expression to respond to hypoxia. Consistently, the results of differential display suggested the induction of glycolytic enzymes, including aldolase A (EC 4.1.2.13), hexokinase II (ATP: D-hexose 6-phosphotransferase, EC 2.7.1.1), and triosephosphate isomerase (EC 5.3.1.1) in the hypoxic culture. Marked induction of these glycolytic enzymes in hypoxic astrocytes was confirmed by Northern blot analysis. These data provide a theoretical basis to understand the ability of astrocytes to tolerate ischemic condition.


Subject(s)
Astrocytes/metabolism , Enzymes/metabolism , Glucose/metabolism , Hypoxia/physiopathology , Animals , Animals, Newborn , Antimetabolites/pharmacology , Astrocytes/cytology , Astrocytes/enzymology , Blotting, Northern , Cell Survival/drug effects , Cells, Cultured , Cycloheximide/pharmacology , DNA, Complementary/genetics , DNA, Complementary/isolation & purification , Deoxyglucose/pharmacology , Enzyme Induction , Enzymes/genetics , Fructose-Bisphosphate Aldolase/genetics , Fructose-Bisphosphate Aldolase/metabolism , Gene Expression Regulation , Glucose/pharmacokinetics , Glycogen/metabolism , Glycolysis , Hexokinase/genetics , Hexokinase/metabolism , Oxygen/pharmacology , Protein Synthesis Inhibitors/pharmacology , RNA/genetics , RNA/metabolism , Rats , Rats, Sprague-Dawley , Triose-Phosphate Isomerase/genetics , Triose-Phosphate Isomerase/metabolism
17.
J Neurochem ; 70(2): 550-7, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9453549

ABSTRACT

Protein synthesis is important in the readaptive processes for cultured astrocytes after hypoxia and subsequent reoxygenation. We have identified 72-kDa inducible heat shock protein (HSP72) as a major stress protein in reoxygenated astrocytes. To assess the mechanism for reoxygenation-mediated induction of HSP72, a reporter gene that consists of a human HSP promoter fused to the luciferase gene was transfected into cultured astrocytes. Analysis of cellular energy nucleotides showed an increase of the ADP/ATP ratio after reoxygenation, which synchronized with activation of the HSP promoter. Activation of the HSP promoter was also observed after an addition of iodoacetic acid to hypoxic astrocytes, which reached the maximum when the ADP/ATP ratio reached 50%, but further decline in the energy profile caused inactivation of this promoter. Inhibition of protein synthesis after reoxygenation resulted in temporary restoration of the energy profile and suppression of the DNA binding activity of the heat shock factor. Addition of quercetin greatly decreased the [3H]leucine incorporation in the polysome fraction without any effect on the mature mRNA formation. These data suggest that the energy depletion in reoxygenation triggers induction of HSP72 after reoxygenation, which may act as a pivotal mediator in the stress response of reoxygenated astrocytes by facilitating protein synthesis.


Subject(s)
Adenine Nucleotides/metabolism , Astrocytes/metabolism , Energy Metabolism/physiology , Heat-Shock Proteins/biosynthesis , Aerobiosis , Animals , Animals, Newborn , Astrocytes/cytology , Astrocytes/drug effects , Cell Hypoxia , Cells, Cultured , Energy Metabolism/drug effects , Genes, Reporter , HSP72 Heat-Shock Proteins , Humans , Luciferases/biosynthesis , Polyribosomes/drug effects , Polyribosomes/metabolism , Promoter Regions, Genetic , Quercetin/pharmacology , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/biosynthesis , Transfection , Ubiquitins/biosynthesis
18.
J Biol Chem ; 272(28): 17810-4, 1997 Jul 11.
Article in English | MEDLINE | ID: mdl-9211935

ABSTRACT

Advanced glycation end products (AGEs) exert their cellular effects on cells by interacting with specific cellular receptors, the best characterized of which is the receptor for AGE (RAGE). The transductional processes by which RAGE ligation transmits signals to the nuclei of cells is unknown and was investigated. AGE-albumin, a prototypic ligand, activated p21(ras) in rat pulmonary artery smooth muscle cells that express RAGE, whereas nonglycated albumin was without effect. MAP kinase activity was enhanced at concentrations of AGE-albumin, which activated p21(ras) and NF-kappaB. Depletion of intracellular glutathione rendered cells more sensitive to AGE-mediated activation of this signaling pathway. In contrast, signaling was blocked by preventing p21(ras) from associating with the plasma membrane or mutating Cys118 on p21(ras) to Ser. Signaling was receptor-dependent, because it was prevented by blocking access to RAGE with either anti-RAGE IgG or by excess soluble RAGE. These data suggest that RAGE-mediated induction of cellular oxidant stress triggers a cascade of intracellular signals involving p21(ras) and MAP kinase, culminating in transcription factor activation. The molecular mechanism that triggers this pathway likely involves oxidant modification and activation of p21(ras).


Subject(s)
Alkyl and Aryl Transferases , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Glycation End Products, Advanced/metabolism , Oxidative Stress , Transferases/metabolism , Animals , Enzyme Activation , Glutathione/metabolism , PC12 Cells , Rats , Receptor for Advanced Glycation End Products , Receptors, Immunologic/metabolism , Signal Transduction
19.
J Periodontal Res ; 31(7): 508-15, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8915955

ABSTRACT

We hypothesized that one mechanism underlying advanced periodontal disease in diabetes may involve oxidant stress in the gingiva, induced by the effects of Advanced Glycation Endproducts (AGEs), the irreversible products of non-enzymatic glycation and oxidation of proteins and lipids which accumulate in diabetic plasma and tissue. Infusion of AGE albumin, a prototypic ligand, into mice resulted in increased generation of thiobarbituric acid reactive substances (TBARS) compared with infusion of non-glycated albumin in the gingiva, as well as in the lung, kidney and brain. Pretreatment of the animals with the antioxidants probucol or N-acetylcysteine (NAC) prevented the generation of TBARS in the gingiva. Affinity-purified antibody to AGEs demonstrated increased immunoreactivity for AGEs in the vasculature and connective tissues of the gingiva in streptozotocin-induced diabetic mice compared to non-diabetic controls. Increased immunoreactivity for AGEs was also demonstrated in the gingiva of diabetic humans compared with non-diabetic individuals via immunohistochemistry and ELISA. Consistent with these data, immunohistochemistry for heme oxygenase-1, a marker of enhanced oxidant stress, was increased in the gingival vasculature of diabetic mice and humans compared with non-diabetic controls. These data suggest that AGEs present in diabetic gingiva may be associated with a state of enhanced oxidant stress, a potential mechanism for accelerated tissue injury.


Subject(s)
Diabetes Complications , Diabetes Mellitus/metabolism , Glycation End Products, Advanced/metabolism , Oxidative Stress/physiology , Periodontitis/etiology , Adult , Animals , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Enzyme-Linked Immunosorbent Assay , Gingiva/metabolism , Glycation End Products, Advanced/administration & dosage , Glycation End Products, Advanced/blood , Heme Oxygenase (Decyclizing)/biosynthesis , Humans , Mice , Periodontitis/metabolism , Streptozocin , Thiobarbituric Acid Reactive Substances/metabolism
20.
J Clin Invest ; 98(5): 1088-94, 1996 Sep 01.
Article in English | MEDLINE | ID: mdl-8787669

ABSTRACT

An important component of amyloid fibrils in dialysis-related amyloidosis is a form of beta2microglobulin modified with advanced glycation end products (AGEs) of the Maillard reaction, known as AGE-beta2M. We demonstrate here that the interaction of AGE-beta2M with mononuclear phagocytes (MPs), cells important in the pathogenesis of the inflammatory arthropathy of dialysis-related amyloidosis, is mediated by the receptor for AGEs, or RAGE. 125I-AGE-beta2M bound to immobilized RAGE or to MPs in a specific, dose-dependent manner (Kd approximately 53.5 and approximately 81.6 nM, respectively), a process inhibited in the presence of RAGE blockade. AGE-beta2M-mediated monocyte chemotaxis was prevented by excess sRAGE or anti-RAGE IgG. Induction of tumor necrosis factor-alpha (TNF) expression by MPs exposed to AGE-beta2M resulted from engagement of RAGE, as appearances of TNF transcripts and TNF antigen release into culture supernatants were prevented by addition of sRAGE, a process mediated, at least in part, by oxidant stress. AGE-beta2M reduced cytochrome c and the elaboration of TNF by MPs was inhibited by N-acetylcysteine. Consistent with these data, immunohistochemical studies of AGE-laden amyloid deposits of a long-term hemodialysis patient revealed positive staining for RAGE in the MPs infiltrating these lesions. These data indicate that RAGE is a central binding site for AGEs formed in vivo and suggest that AGE-beta2M-MP-RAGE interaction likely contributes to the initiation of an inflammatory response in amyloid deposits of long-term hemodialysis patients, a process which may ultimately lead to bone and joint destruction.


Subject(s)
Amyloidosis/etiology , Glycation End Products, Advanced/metabolism , Joint Diseases/etiology , Leukocytes, Mononuclear/metabolism , Phagocytes/metabolism , Receptors, Immunologic/metabolism , beta 2-Microglobulin/metabolism , Antigens, CD/isolation & purification , Antigens, Differentiation, Myelomonocytic/isolation & purification , Cell Movement , Humans , Oxidation-Reduction , Oxidative Stress , Protein Binding , Receptor for Advanced Glycation End Products , Renal Dialysis/adverse effects , Signal Transduction , Skin/pathology , Tumor Necrosis Factor-alpha/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...