Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
Zhongguo Zhong Yao Za Zhi ; 48(18): 4893-4901, 2023 Sep.
Article in Chinese | MEDLINE | ID: mdl-37802831

ABSTRACT

Yiyi Fuzi Baijiang Powder(YFBP), originating from Synopsis of the Golden Chamber, is a classic prescription composed of Coicis Semen, Aconiti Lateralis Radix Praeparata, and Patriniae Herba for the treatment of abscesses and pus discharge. This article presented a systematic analysis of the clinical application of YFBP, including the indicated diseases, the number of cases, efficacy, dosage, administration methods, and compatibility with other drugs. The analysis reveals that YFBP has a wide range of clinical applications. It is commonly used, often with modifications or in combination with western medicine, for diseases in the fields of gastroente-rology, gynecology, urology, dermatology, and others. And most of the Traditional Chinese Medicine(TCM) evidence involved in these diseases are damp-heat evudence. The prescription shows rich variations in clinical administration methods, and most of which are the treatment of aqueous decoction of it. The therapeutic effect is also significant, and the total effective rate of clinical treatment is re-latively high. Additionally, this article summarized the pharmacological research on YFBP and found that it possessed various pharmacological effects, including anti-inflammatory, antioxidant, anticancer, and immune-modulating properties. Finally, correlation analysis was conducted on the main diseases, TCM types, prescription doses, pharmacological effects and action targets of YFBP, which to show the relationship between these five aspects in a visual form, reflecting the relationship between its clinical application and modern pharmacological effects. These findings provide a reference basis for further development and research on YFBP.


Subject(s)
Aconitum , Diterpenes , Drugs, Chinese Herbal , Powders , Drugs, Chinese Herbal/pharmacology , Medicine, Chinese Traditional
2.
Neurobiol Aging ; 34(1): 226-37, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22921154

ABSTRACT

Accumulation of amyloid-ß (Aß) is a key event mediating the cognitive deficits in Alzheimer's disease (AD) as Aß promotes synaptic dysfunction and triggers neuronal death. Recent evidence has linked the hormone leptin to AD as leptin levels are markedly attenuated in AD patients. Leptin is also a potential cognitive enhancer as it facilitates the cellular events underlying hippocampal learning and memory. Here we show that leptin prevents the detrimental effects of Aß(1-42) on hippocampal long-term potentiation. Moreover leptin inhibits Aß(1-42)-driven facilitation of long-term depression and internalization of the 2-amino-3-(5-methyl-3-oxo-1,2- oxazol-4-yl)propanoic acid (AMPA) receptor subunit, GluR1, via activation of PI3-kinase. Leptin also protects cortical neurons from Aß(1-42)-induced cell death by a signal transducer and activator of transcription-3 (STAT-3)-dependent mechanism. Furthermore, leptin inhibits Aß(1-42)-mediated upregulation of endophilin I and phosphorylated tau in vitro, whereas cortical levels of endophilin I and phosphorylated tau are enhanced in leptin-insensitive Zucker fa/fa rats. Thus leptin benefits the functional characteristics and viability of neurons that degenerate in AD. These novel findings establish that the leptin system is an important therapeutic target in neurodegenerative conditions.


Subject(s)
Amyloid beta-Peptides/pharmacology , Hippocampus/cytology , Leptin/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Peptide Fragments/pharmacology , Synapses/drug effects , Analysis of Variance , Animals , Animals, Newborn , Biophysics , Cell Death/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Dose-Response Relationship, Drug , Drug Interactions , Electric Stimulation , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Organ Culture Techniques , Patch-Clamp Techniques , Rats , Receptors, AMPA/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Synaptic Potentials/drug effects , Tetrazolium Salts , Thiazoles , tau Proteins/metabolism
3.
Biochem Insights ; 2010(3): 47-59, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-21852897

ABSTRACT

The aim of our project was to study the effect of streptozotocin (STZ)-induced hyperglycemia on sciatic nerve morphology, blood plasma markers and immunohistochemical expression of RAGE (the Receptor for Advanced Glycation End-products), and its ligands-S100B and Carboxymethyl Lysine (CML)-advanced glycation endproduct (AGE) in the laboratory pig. Six months after STZ-injections, blood plasma measurements, morphometric analysis of sciatic nerve fiber density, immunofluorescent distribution of potential molecular neuropathy contributors, ELISA measurement of plasma AGE level and HPLC analysis of sciatic nerve levels of one of the pre-AGE and the glycolysis intermediate products-methyl-glyoxal (MG) were performed. The results of our study revealed that STZ-injected animals displayed elevated levels of plasma glucose, gamma glutamyl transferase (GGT) and triglycerides. The sciatic nerve of STZ-injected pigs revealed significantly lower numbers of small-diameter myelinated fibers, higher immunoreactivity for RAGE and S100B and increased levels of MG as compared to control animals. Our results correspond to clinical findings in human patients with hyperglycemia/diabetes-evoked peripheral neuropathy and suggest that the domestic pig may be a suitable large animal model for the study of mechanisms underlying hyperglycemia-induced neurological complications in the peripheral nerve and may serve as a relevant model for the pre-clinical assessment of candidate drugs in neuropathy.

4.
J Alzheimers Dis ; 16(4): 833-43, 2009.
Article in English | MEDLINE | ID: mdl-19387116

ABSTRACT

Receptor for Advanced Glycation Endproducts (RAGE) is a multiligand member of the immunoglobulin superfamily of cell surface molecules which serves as a receptor for amyloid-beta peptide (Abeta) on neurons, microglia, astrocytes, and cells of vessel wall. Increased expression of RAGE is observed in regions of the brain affected by Alzheimer's disease (AD), and Abeta-RAGE interaction in vitro leads to cell stress with the generation of reactive oxygen species and activation of downstream signaling mechanisms including the MAP kinase pathway. RAGE-mediated activation of p38 MAP kinase in neurons causes Abeta-induced inhibition of long-term potentiation in slices of entorhinal cortex. Increased expression of RAGE in an Abeta-rich environment, using transgenic mouse models, accelerates and accentuates pathologic, biochemical, and behavioral abnormalities compared with mice overexpressing only mutant amyloid-beta protein precursor. Interception of Abeta interaction with RAGE, by infusion of soluble RAGE, decreases Abeta content and amyloid load, as well as improving learning/memory and synaptic function, in a murine transgenic model of Abeta accumulation. These data suggest that RAGE may be a therapeutic target for AD.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Receptors, Immunologic/metabolism , Alzheimer Disease/physiopathology , Alzheimer Disease/therapy , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Brain/metabolism , Brain/pathology , Disease Models, Animal , Humans , Mice , Receptor for Advanced Glycation End Products
5.
Ann Med ; 41(6): 408-22, 2009.
Article in English | MEDLINE | ID: mdl-19322705

ABSTRACT

The multiligand receptor RAGE (receptor for advanced glycation end-products) is emerging as a central mediator in the immune/inflammatory response. Epidemiological evidence accruing in the human suggests upregulation of RAGE's ligands (AGEs, S100/calgranulins, high mobility group box-1 (HMGB1), and amyloid beta-peptide and beta-sheet fibrils) and the receptor itself at sites of inflammation and in chronic diseases such as diabetes and neurodegeneration. The consequences of ligand-RAGE interaction include upregulation of molecules implicated in inflammatory responses and tissue damage, such as cytokines, adhesion molecules, and matrix metalloproteinases. In this review, we discuss the localization of RAGE and its ligand families and the biological impact of this axis in multiple cell types implicated in chronic diseases. Lastly, we consider findings from animal model studies suggesting that although tissue-damaging effects ensue from recruitment of the ligand-RAGE interaction, in distinct settings, adaptive and repair/regeneration outcomes appear to override detrimental effects of RAGE. As RAGE blockade moves further into clinical development, clarifying the biology of RAGE garners ever-increasing importance.


Subject(s)
Diabetes Complications/metabolism , Glycation End Products, Advanced/metabolism , Inflammation/metabolism , Neurodegenerative Diseases/metabolism , Amyloid beta-Peptides/metabolism , Animals , Dendrites/metabolism , Glycation End Products, Advanced/immunology , HMGB1 Protein/metabolism , Humans , Leukocyte L1 Antigen Complex/metabolism , Ligands , Macrophages/metabolism , Mice , Monocytes/metabolism , Neutrophils/metabolism , Rats , Up-Regulation
7.
J Alzheimers Dis ; 17(1): 59-68, 2009.
Article in English | MEDLINE | ID: mdl-19221410

ABSTRACT

Oligomeric amyloid-beta (Abeta) interferes with long-term potentiation (LTP) and cognitive processes, suggesting that Abeta peptides may play a role in the neuronal dysfunction which characterizes the early stages of Alzheimer's disease (AD). Multiple lines of evidence have highlighted RAGE (receptor for advanced glycation end-products) as a receptor involved in Abeta-induced neuronal and synaptic dysfunction. In the present study, we investigated the effect of oligomeric soluble Abeta1-42 on LTP elicited by the stimulation of different intracortical pathways in the mouse visual cortex. A variety of nanomolar concentrations (20-200 nM) of Abeta1-42 were able to inhibit LTP in cortical layer II-III induced by either white matter (WM-Layer II/III) or the layer II/III (horizontal pathway) stimulation, whereas the inhibition of LTP was more susceptible to Abeta1-42, which occurred at 20 nM of Abeta, when stimulating layer II-III horizontal pathway. Remarkably, cortical slices were resistant to nanomolar Abeta1-42 in the absence of RAGE (genetic deletion of RAGE) or blocking RAGE by RAGE antibody. These results indicate that nanomolar Abeta inhibits LTP expression in different neocortical circuits. Crucially, it is demonstrated that Abeta-induced reduction of LTP in different cortical pathways is mediated by RAGE.


Subject(s)
Amyloid beta-Peptides/pharmacology , Long-Term Potentiation/drug effects , Mitogen-Activated Protein Kinases/metabolism , Nerve Net/physiology , Peptide Fragments/pharmacology , Visual Cortex/physiology , Analysis of Variance , Animals , Animals, Newborn , Antibodies/pharmacology , Biophysics , Dose-Response Relationship, Drug , Electric Stimulation/methods , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinases/deficiency , Mitogen-Activated Protein Kinases/immunology , Neural Pathways/physiology , Visual Cortex/drug effects
8.
Nat Med ; 14(10): 1097-105, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18806802

ABSTRACT

Cyclophilin D (CypD, encoded by Ppif) is an integral part of the mitochondrial permeability transition pore, whose opening leads to cell death. Here we show that interaction of CypD with mitochondrial amyloid-beta protein (Abeta) potentiates mitochondrial, neuronal and synaptic stress. The CypD-deficient cortical mitochondria are resistant to Abeta- and Ca(2+)-induced mitochondrial swelling and permeability transition. Additionally, they have an increased calcium buffering capacity and generate fewer mitochondrial reactive oxygen species. Furthermore, the absence of CypD protects neurons from Abeta- and oxidative stress-induced cell death. Notably, CypD deficiency substantially improves learning and memory and synaptic function in an Alzheimer's disease mouse model and alleviates Abeta-mediated reduction of long-term potentiation. Thus, the CypD-mediated mitochondrial permeability transition pore is directly linked to the cellular and synaptic perturbations observed in the pathogenesis of Alzheimer's disease. Blockade of CypD may be a therapeutic strategy in Alzheimer's disease.


Subject(s)
Alzheimer Disease/prevention & control , Cyclophilins/deficiency , Learning , Memory , Mitochondria/metabolism , Neurons/physiology , Alzheimer Disease/drug therapy , Alzheimer Disease/etiology , Amyloid beta-Peptides/metabolism , Animals , Apoptosis , Calcium/metabolism , Cyclophilins/antagonists & inhibitors , Cyclophilins/physiology , Disease Models, Animal , Humans , Membrane Potential, Mitochondrial , Mice , Mitochondrial Membrane Transport Proteins , Mitochondrial Permeability Transition Pore , Reactive Oxygen Species/metabolism , Synapses/physiology
9.
Neurochem Int ; 52(7): 1358-64, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18387708

ABSTRACT

Ovarian hormone decline after menopause may influence cognitive performance and increase the risk for Alzheimer's disease (AD) in women. Amyloid-beta peptide (Abeta) has been proposed to be the primary cause of AD. In this study, we examined whether ovariectomy (OVX) could affect the levels of cofactors Abeta-binding alcohol dehydrogenase (ABAD) and receptor for advanced glycation endproducts (RAGE), which have been reported to potentiate Abeta-mediated neuronal perturbation, in mouse hippocampus, correlating with estrogen and Abeta levels. Female ICR mice were randomly divided into ovariectomized or sham-operated groups, and biochemical analyses were carried out at 5 weeks after the operation. OVX for 5 weeks significantly decreased hippocampal 17beta-estradiol level, while it tended to reduce the hormone level in serum, compared with the sham-operated control. In contrast, OVX did not affect hippocampal Abeta(1-40) level, although it significantly increased serum Abeta(1-40) level. Furthermore, we demonstrated that OVX increased hippocampal ABAD level in neurons, but not astrocytes, while it did not affect RAGE level. These findings suggest that the expression of neuronal ABAD depends on estrogen level in the hippocampus and the increase in serum Abeta and hippocampal ABAD induced by ovarian hormone decline may be associated with pre-stage of memory deficit in postmenopausal women and Abeta-mediated AD pathology.


Subject(s)
Alcohol Dehydrogenase/metabolism , Amyloid beta-Peptides/metabolism , Hippocampus/metabolism , Neurons/metabolism , Ovariectomy , Animals , Astrocytes/metabolism , Blotting, Western , DNA-Binding Proteins , Estradiol/pharmacology , Female , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/cytology , Hippocampus/enzymology , Mice , Mice, Inbred ICR , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Receptor for Advanced Glycation End Products , Receptors, Immunologic/metabolism
10.
J Neurosci ; 28(13): 3521-30, 2008 Mar 26.
Article in English | MEDLINE | ID: mdl-18367618

ABSTRACT

Soluble amyloid-beta (Abeta) peptide is likely to play a key role during early stages of Alzheimer's disease (AD) by perturbing synaptic function and cognitive processes. Receptor for advanced glycation end products (RAGE) has been identified as a receptor involved in Abeta-induced neuronal dysfunction. We investigated the role of neuronal RAGE in Abeta-induced synaptic dysfunction in the entorhinal cortex, an area of the brain important in memory processes that is affected early in AD. We found that soluble oligomeric Abeta peptide (Abeta42) blocked long-term potentiation (LTP), but did not affect long-term depression, paired-pulse facilitation, or basal synaptic transmission. In contrast, Abeta did not inhibit LTP in slices from RAGE-null mutant mice or in slices from wild-type mice treated with anti-RAGE IgG. Similarly, transgenic mice expressing a dominant-negative form of RAGE targeted to neurons showed normal LTP in the presence of Abeta, suggesting that neuronal RAGE functions as a signal transducer for Abeta-mediated LTP impairment. To investigate intracellular pathway transducing RAGE activation by Abeta, we used inhibitors of stress activated kinases. We found that inhibiting p38 mitogen-activated protein kinase (p38 MAPK), but not blocking c-Jun N-terminal kinase activation, was capable of maintaining LTP in Abeta-treated slices. Moreover, Abeta-mediated enhancement of p38 MAPK phosphorylation in cortical neurons was reduced by blocking antibodies to RAGE. Together, our results indicate that Abeta impairs LTP in the entorhinal cortex through neuronal RAGE-mediated activation of p38 MAPK.


Subject(s)
Amyloid beta-Peptides/toxicity , Neurons/cytology , Neurons/drug effects , Peptide Fragments/toxicity , Receptors, Immunologic/metabolism , Synapses/physiology , p38 Mitogen-Activated Protein Kinases/physiology , Action Potentials/physiology , Action Potentials/radiation effects , Animals , Animals, Newborn , Antibodies/pharmacology , Cells, Cultured , Dose-Response Relationship, Radiation , Electric Stimulation/methods , Entorhinal Cortex/cytology , Enzyme Activation , Enzyme-Linked Immunosorbent Assay/methods , In Vitro Techniques , Long-Term Potentiation/drug effects , Long-Term Potentiation/radiation effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Inhibition/drug effects , Neural Inhibition/radiation effects , Receptor for Advanced Glycation End Products , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Synapses/drug effects
11.
Am J Physiol Heart Circ Physiol ; 294(4): H1862-70, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18245560

ABSTRACT

Protein kinase C-betaII (PKCbetaII) is an important modulator of cellular stress responses. To test the hypothesis that PKCbetaII modulates the response to myocardial ischemia-reperfusion (I/R) injury, we subjected mice to occlusion and reperfusion of the left anterior descending coronary artery. Homozygous PKCbeta-null (PKCbeta(-/-)) and wild-type mice fed the PKCbeta inhibitor ruboxistaurin displayed significantly decreased infarct size and enhanced recovery of left ventricular (LV) function and reduced markers of cellular necrosis and serum creatine phosphokinase and lactate dehydrogenase levels compared with wild-type or vehicle-treated animals after 30 min of ischemia followed by 48 h of reperfusion. Our studies revealed that membrane translocation of PKCbetaII in LV tissue was sustained after I/R and that gene deletion or pharmacological blockade of PKCbeta protected ischemic myocardium. Homozygous deletion of PKCbeta significantly diminished phosphorylation of c-Jun NH(2)-terminal mitogen-activated protein kinase and expression of activated caspase-3 in LV tissue of mice subjected to I/R. These data implicate PKCbeta in I/R-mediated myocardial injury, at least in part via phosphorylation of JNK, and suggest that blockade of PKCbeta may represent a potent strategy to protect the vulnerable myocardium.


Subject(s)
Myocardial Reperfusion Injury/metabolism , Myocardium/enzymology , Protein Kinase C/metabolism , Signal Transduction , Ventricular Function, Left , Animals , Caspase 3/metabolism , Cell Membrane/metabolism , Coronary Vessels/surgery , Creatine Kinase/blood , Disease Models, Animal , Enzyme Activation , Indoles/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , L-Lactate Dehydrogenase/blood , Ligation , Male , Maleimides/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardial Reperfusion Injury/prevention & control , Myocardium/pathology , Necrosis , Phosphorylation , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/deficiency , Protein Kinase C/genetics , Protein Kinase C beta , Protein Kinase Inhibitors/pharmacology , Protein Transport , Recovery of Function , Signal Transduction/drug effects , Time Factors , Ventricular Function, Left/drug effects
12.
J Biol Chem ; 283(9): 5685-91, 2008 Feb 29.
Article in English | MEDLINE | ID: mdl-18167351

ABSTRACT

Alzheimer patients have increased levels of both the 42 amyloid-beta-peptide (Abeta) and the amyloid binding alcohol dehydrogenase (ABAD), which is an intracellular binding site for Abeta. The overexpression of Abeta and ABAD in transgenic mice has shown that the binding of Abeta to ABAD results in amplified neuronal stress and impairment of learning and memory. From a proteomic analysis of the brains from these animals, we have identified for the first time that the protein endophilin I increases in Alzheimer diseased brain. The increase in endophilin I levels in neurons is linked to an increase in the activation of the stress kinase c-Jun N-terminal kinase with the subsequent death of the neurons. We also demonstrate in living animals that the expression level of endophilin I is an indicator for the interaction of ABAD and Abeta as its expression levels return to normal if this interaction is perturbed. Therefore this identifies endophilin I as a new indicator of the progression of Alzheimer disease.


Subject(s)
Acyltransferases/biosynthesis , Alcohol Dehydrogenase/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Brain/metabolism , Neurons/metabolism , Acyltransferases/genetics , Alcohol Dehydrogenase/genetics , Alzheimer Disease/genetics , Amyloid beta-Peptides/genetics , Animals , Biomarkers/metabolism , Brain/pathology , Cell Death/genetics , Humans , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Memory , Mice , Mice, Transgenic , Neurons/pathology , Protein Binding/genetics
13.
Expert Rev Neurother ; 7(11): 1517-25, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17997700

ABSTRACT

Metabolic dysfunction is one of the early features in Alzheimer's disease (AD) affected brain. Amyloid-beta peptide (Abeta), a major peptide deposited in neuritic plaques, has been considered as an important initiating molecule in the pathogenesis of AD. However, the pathogenic role of Abeta remains to be determined. Here, we review current studies showing that progressive accumulation of Abeta occurs within the mitochondria of both transgenic mice overexpressing mutant Abeta peptide precursor protein and autopsied brains from AD patients. Interaction of Abeta with Abeta-binding alcohol dehydrogenase (ABAD), a short-chain alcohol dehydrogenase in the mitochondrial matrix, leads to mitochondrial dysfunction evidenced by increased reactive oxygen species generation, mitochondrial membrane permeability formation and caspase-3-like activity induction, and decreased activities of the Krebs cycle. These effects can be blocked by intracellular transduction of the ABAD decoy peptide. We hypothesize that Abeta-induced and mitochondria-dependent cytotoxic pathways might play an important role in AD pathogenesis and could be a potential therapeutic target.


Subject(s)
Amyloid beta-Peptides/adverse effects , Amyloid beta-Peptides/physiology , Mitochondrial Proteins/adverse effects , Mitochondrial Proteins/physiology , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/toxicity , Animals , Humans , Mitochondrial Proteins/toxicity
14.
J Alzheimers Dis ; 12(2): 177-84, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17917162

ABSTRACT

As an important molecule in the pathogenesis of Alzheimer's disease (AD), amyloid-beta (Abeta) interferes with multiple aspects of mitochondrial function, including energy metabolism failure, production of reactive oxygen species (ROS) and permeability transition pore formation. Recent studies have demonstrated that Abeta progressively accumulates within mitochondrial matrix, providing a direct link to mitochondrial toxicity. Abeta-binding alcohol dehydrogenase (ABAD) is localized to the mitochondrial matrix and binds to mitochondrial Abeta. Interaction of ABAD with Abeta exaggerates Abeta-mediated mitochondrial and neuronal perturbation, leading to impaired synaptic function, and dysfunctional spatial learning/memory. Thus, blockade of ABAD/Abeta interaction may be a potential therapeutic strategy for AD.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Mitochondrial Diseases , Alzheimer Disease/epidemiology , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Energy Metabolism , Humans , Mitochondrial Diseases/epidemiology , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/physiopathology
15.
J Neurochem ; 103(3): 1132-44, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17683481

ABSTRACT

Collapsin response mediator protein 2 (CRMP2) is an abundant brain-enriched protein that can regulate microtubule assembly in neurons. This function of CRMP2 is regulated by phosphorylation by glycogen synthase kinase 3 (GSK3) and cyclin-dependent kinase 5 (Cdk5). Here, using novel phosphospecific antibodies, we demonstrate that phosphorylation of CRMP2 at Ser522 (Cdk5-mediated) is increased in Alzheimer's disease (AD) brain, while CRMP2 expression and phosphorylation of the closely related isoform CRMP4 are not altered. In addition, CRMP2 phosphorylation at the Cdk5 and GSK3 sites is increased in cortex and hippocampus of the triple transgenic mouse [presenilin-1 (PS1)(M146V)KI; Thy1.2-amyloid precursor protein (APP)(swe); Thy1.2tau(P301L)] that develops AD-like plaques and tangles, as well as the double (PS1(M146V)KI; Thy1.2-APP(swe)) transgenic mouse. The hyperphosphorylation is similar in magnitude to that in human AD and is evident by 2 months of age, ahead of plaque or tangle formation. Meanwhile, there is no change in CRMP2 phosphorylation in two other transgenic mouse lines that display elevated amyloid beta peptide levels (Tg2576 and APP/amyloid beta-binding alcohol dehydrogenase). Similarly, CRMP2 phosphorylation is normal in hippocampus and cortex of Tau(P301L) mice that develop tangles but not plaques. These observations implicate hyperphosphorylation of CRMP2 as an early event in the development of AD and suggest that it can be induced by a severe APP over-expression and/or processing defect.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Brain/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Plaque, Amyloid/metabolism , Aged , Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Binding Sites , Brain/physiopathology , Cyclin-Dependent Kinase 5/metabolism , Disease Models, Animal , Disease Progression , Gene Expression Regulation/physiology , Glycogen Synthase Kinase 3/metabolism , Humans , Mice , Mice, Transgenic , Microtubules/metabolism , Neurofibrillary Tangles/metabolism , Phosphorylation , Time Factors
16.
Mol Cell Neurosci ; 35(2): 377-82, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17490890

ABSTRACT

Alzheimer's patients have increased levels of both the 42 beta amyloid-beta-peptide (Abeta) and amyloid binding alcohol dehydrogenase (ABAD) which is an intracellular binding site for Abeta. The over-expression of Abeta and ABAD in transgenic mice has shown that the binding of Abeta to ABAD results in exaggerating neuronal stress and impairment of learning and memory. From a proteomic analysis of the brains from these animals we identified that peroxiredoxin II levels increase in Alzheimer's diseased brain. This increase in peroxiredoxin II levels protects neurons against Abeta induced toxicity. We also demonstrate, for the first time in living animals, that the expression level of peroxiredoxin II is an indicator for the interaction of ABAD and Abeta as its expression levels return to normal if this interaction is perturbed. Therefore this indicates the possibility of reversing changes observed in Alzheimer's disease and that the Abeta-ABAD interaction is a suitable drug target.


Subject(s)
Alcohol Dehydrogenase/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid/metabolism , Brain/metabolism , Heat-Shock Proteins/metabolism , Peroxidases/metabolism , Up-Regulation/physiology , Amyloid beta-Protein Precursor/genetics , Animals , Brain/pathology , Cells, Cultured , Disease Models, Animal , Electrophoresis, Gel, Two-Dimensional/methods , Gene Expression Regulation , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Peroxiredoxins , Protein Binding , Proteomics/methods
17.
Curr Mol Med ; 7(8): 735-42, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18331231

ABSTRACT

This review focuses on the current findings regarding interaction between amyloid beta peptide (Abeta) and receptor for advanced glycation endproducts (RAGE) and its roles in the pathogenesis of Alzheimer's disease (AD). As a ubiquitously expressed cell surface receptor, RAGE mediates the effects of Abeta on microglia, blood-brain barrier (BBB) and neurons through activating different signaling pathways. Data from autopsy brain tissues, in vitro cell cultures and transgenic mouse models suggest that Abeta-RAGE interaction exaggerates neuronal stress, accumulation of Abeta, impaired learning memory, and neuroinflammation. Blockade of RAGE protects against Abeta-mediated cellular perturbation. These findings may have an important therapeutic implication for neurodegenerative disorders relevant to AD.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Receptors, Immunologic/metabolism , Alzheimer Disease/physiopathology , Animals , Brain/metabolism , Brain/pathology , Humans , Protein Binding , Receptor for Advanced Glycation End Products , Synapses/pathology
18.
Curr Alzheimer Res ; 3(5): 515-20, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17168650

ABSTRACT

Mitochondrial dysfunction has been implicated in causing metabolic abnormalities in Alzheimer's disease (AD). The searches for mitochondrial DNA variants associated with AD susceptibility have generated conflicting results. The age-related accumulation of somatic mitochondrial DNA deletion has been suggested to play a pathogenic role in the development of AD. Recent studies have demonstrated that amyloid-beta peptide (Abeta) progressively accumulates in mitochndrial matrix, as demonstrated in both transgenic mice over-expressing mutant amyloid precursor protein (APP) and autopsy brain from AD patients. Abeta-mediated mitochondrial stress was evidenced by impaired oxygen consumption and decreased respiratory chain complexes III and IV activities in brains from AD patients and AD-type transgenic mouse model. Furthermore, our studies indicated that interaction of intramitochondrial Abeta with a mitochondrial enzyme, amyloid binding alcohol dehydrogenase (ABAD), inhibits its enzyme activity, enhances generation of reactive oxygen species (ROS), impairs energy metabolism, and exaggerates Abeta-induced spatial learning/memory deficits and neuropathological changes in transgenic AD-type mouse model. Interception of ABAD-Abeta interaction may be a potential therapeutic strategy for Alzheimer's disease.


Subject(s)
Alzheimer Disease/etiology , Mitochondrial Diseases/complications , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , DNA, Mitochondrial/metabolism , Humans , Mice
19.
Endocrinol Metab Clin North Am ; 35(3): 511-24, viii, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16959583

ABSTRACT

The presence of elevated blood glucose levels characterizes the diabetic state. Hyperglycemia may be caused by a number of underlying factors; however, the consequences of chronically elevated glucose are similar. Both the macrovasculature and microvasculature are exquisitely sensitive to the long-term effects of elevated blood glucose. Cardiovascular disease remains the leading cause of morbidity and mortality in diabetes, regardless of the underlying cause of hyperglycemia. Although other substrates, such as DNA, are susceptible to glycation, this article addresses the impact of nonenzymatic glycation on the proteome. The impact of Advanced Glycation End products (AGEs) on alteration of protein function and signal transduction mechanisms contributes to the pathogenesis of diabetes complications. This suggests that blocking the generation or molecular impact of AGEs may modulate the complications of diabetes.


Subject(s)
Cardiovascular Diseases/etiology , Diabetes Complications , Glycation End Products, Advanced , Receptors, Immunologic , Animals , Atherosclerosis , Coronary Restenosis , Diabetes Mellitus/etiology , Diabetes Mellitus/therapy , Disease Models, Animal , Humans , Inflammation/complications , Receptor for Advanced Glycation End Products
20.
J Alzheimers Dis ; 9(2): 127-37, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16873960

ABSTRACT

Mitochondrial and metabolic dysfunction have been linked to Alzheimer's disease for some time. Key questions regarding this association concern the nature and mechanisms of mitochondrial dysfunction, and whether such changes in metabolic properties are pathogenic or secondary, with respect to neuronal degeneration. In terms of mitochondria and Alzheimer's, altered function could reflect intrinsic properties of this organelle, potentially due to mutations in mitochondrial DNA, or extrinsic changes secondary to signal transduction mechanisms activated in the cytosol. This review presents data relevant to these questions, and considers the implication of recent findings demonstrating the presence of amyloid-beta peptide in mitochondria, as well as intra-mitochondrial molecular targets with which it can interact. Regardless of the underlying mechanism(s), it is likely that mitochondrial dysfunction contributes to oxidant stress which is commonly observed in brains of patients with Alzheimer's and transgenic models of Alzheimer's-like pathology.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Mitochondria/metabolism , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Humans , Mitochondria/pathology , Oxidative Stress/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...