Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 104
Filter
1.
Mol Neurobiol ; 55(8): 6863-6880, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29353455

ABSTRACT

Since its discovery as a genetic risk factor for Alzheimer's disease, the APOE4 allele has been linked to the majority of the pathological findings associated with the disease progression. These include abnormalities of the endocytic, autophagic, and lysosomal machineries, which begin at the most early stages of Alzheimer's disease development. Considering that these three vesicular systems share common features and, in fact, comprise an interconnected cargo-trafficking and degradation network, some of the effects of APOE4 are interrelated, while others are system-specific. In turn, APOE4-driven impairments of endocytosis, autophagy, and lysosomal activity influence various aspects of Alzheimer's disease pathology, ranging from Aß generation and clearance to neuronal loss and cognitive deficits. This review discusses the detrimental effects of APOE4 on the endocytic-autophagic-lysosomal axis in the context of Alzheimer's disease, as well as the various mechanisms underlying them.


Subject(s)
Apolipoprotein E4/metabolism , Autophagy , Endocytosis , Lysosomes/metabolism , Alzheimer Disease/pathology , Animals , Humans , Models, Biological
2.
Kidney Cancer ; 1(1): 31-40, 2017 Jul 26.
Article in English | MEDLINE | ID: mdl-30334002

ABSTRACT

In 2017, there is no adjuvant systemic therapy proven to increase overall survival in non-metastatic renal cell carcinoma (RCC). The anti-PD-1 antibody nivolumab improves overall survival in metastatic treatment refractory RCC and is generally tolerable. Mouse solid tumor models have revealed a benefit with a short course of neoadjuvant PD-1 blockade compared to adjuvant therapy. Two ongoing phase 2 studies of perioperative nivolumab in RCC patients have shown preliminary feasibility and safety with no surgical delays or complications. The recently opened PROSPER RCC trial (A Phase 3 RandOmized Study Comparing PERioperative Nivolumab vs. Observation in Patients with Localized Renal Cell Carcinoma Undergoing Nephrectomy; EA8143) will examine if the addition of perioperative nivolumab to radical or partial nephrectomy can improve clinical outcomes in patients with high risk localized and locally advanced RCC. With the goal of increasing cure and recurrence-free survival (RFS) rates in non-metastatic RCC, we are executing a three-pronged, multidisciplinary approach of presurgical priming with nivolumab followed by resection and adjuvant PD-1 blockade. We plan to enroll 766 patients with clinical stage ≥T2 or node positive M0 RCC of any histology in this global, randomized, unblinded, phase 3 National Clinical Trials Network study. The investigational arm will receive two doses of nivolumab 240 mg IV prior to surgery followed by adjuvant nivolumab for 9 months. The control arm will undergo the current standard of care: surgical resection followed by observation. Patients are stratified by clinical T stage, node positivity, and histology. The trial is powered to detect a 14.4% absolute benefit in the primary endpoint of RFS from the ASSURE historical control of 55.8% to 70.2% at 5 years (HR = 0.70). The study is also powered to detect a significant overall survival benefit (HR 0.67). Key safety, feasibility, and quality of life endpoints are incorporated. PROSPER RCC exemplifies team science with a host of planned correlative work to investigate the impact of the baseline immune milieu and changes after neoadjuvant priming on clinical outcomes.

3.
Transl Psychiatry ; 3: e304, 2013 Sep 10.
Article in English | MEDLINE | ID: mdl-24022509

ABSTRACT

Alzheimer's disease (AD) is the most common form of dementia in the elderly. Although there are no drugs that modify the disease process, exposure to an enriched environment (EE) can slow the disease progression. Here, we characterize the effects of AD and EE on the post-transcriptional regulators, microRNAs (miRNAs), which may contribute to the detrimental and beneficial effects of AD and EE, respectively, on synaptic plasticity-related proteins and AD pathology. We found for the first time miRNAs that were inversely regulated in AD and EE, and may affect synaptic proteins and modulators, molecular factors associated with AD pathology, and survival and neuroprotective factors. MiRNAs that were upregulated only in 3xTgAD mice model of AD compared with their control mice were localized to synapses, predicted to downregulate essential synaptic proteins and are highly associated with regulating apoptosis, AD-associated processes and axon guidance. Studying the progressive change in miRNAs modulation during aging of 3xTgAD mice, we identified miRNAs that were regulated in earlier stages of AD, suggesting them as potential AD biomarkers. Last, we characterized AD- and EE-related effects in the mouse hippocampus on tomosyn protein levels, an inhibitor of the synaptic transmission machinery. While EE reduced tomosyn levels, tomosyn levels were increased in old 3xTgAD mice, suggesting a role for tomosyn in the impairment of synaptic transmission in AD. Interestingly, we found that miR-325 regulates the expression levels of tomosyn as demonstrated by a luciferase reporter assay, and that miR-325 was downregulated in AD and upregulated following EE. These findings improve our understanding of the molecular and cellular processes in AD pathology, following EE, and the interplay between the two processes, and open new avenues for the studies of understanding and controlling AD.


Subject(s)
Aging/genetics , Alzheimer Disease/genetics , Environment , Gene Expression Regulation/physiology , Hippocampus/metabolism , MicroRNAs/genetics , Neuronal Plasticity/genetics , Aging/metabolism , Animals , Apoptosis/genetics , Disease Models, Animal , Gene Expression Profiling , Mice , MicroRNAs/metabolism , Nerve Tissue Proteins/metabolism , R-SNARE Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Synapses/genetics , Synapses/metabolism , Synaptic Transmission/genetics
4.
Neuroscience ; 121(2): 315-25, 2003.
Article in English | MEDLINE | ID: mdl-14521991

ABSTRACT

Apolipoprotein E (apoE4) and head trauma are important genetic and environmental risk factors for Alzheimer's disease. Furthermore, apoE4 increases both the acute and chronic consequences of head trauma. The latter are associated with the deposition of amyloid-beta, which is particularly elevated in apoE4 subjects. The short-term effects of head injury are associated with transiently increased metabolism of amyloid precursor protein (APP) and its secreted fragment, APPs. In the present study, we examined the possibility that the acute, short-term pathological effects of apoE4 following head trauma and the corresponding neuroprotective effects of apoE3 are related to isoform-specific effects of apoE on APP metabolism. Accordingly, male transgenic mice expressing human apoE3 or apoE4 on a null mouse apoE background and apoE-deficient and control mice were subjected to closed head injury (CHI). The resulting effects on brain APP, and on its secreted products, APPs and secreted product of the alpha-cleavage of APP (APPsalpha) were then determined 24 h following injury. Immunoblotting revealed no significant differences between the basal APP, APPs and APPsalpha levels of the hippocampus or the cortex of the control and the apoE3 and ApoE4 transgenic mice. The apoE-deficient mice also had similar cortical basal levels of APP and its metabolites, whereas their corresponding basal hippocampal APP and APPs levels were lower than those of the other groups. CHI lowered the hipppocampal APPs and APPsalpha levels of the apoE4 transgenic mice, whereas those of the apoE3 transgenic mice and of the control and apoE-deficient mice were not affected by this insult. In contrast, CHI raised the cortical APP and APPs levels of the apoE3 transgenic mice but had no significant effect on those of the other mice groups. These animal model findings suggest that the acute, short-term pathological effects of apoE4 following CHI and the corresponding neuroprotective effects of apoE3 may be mediated by their opposing effects on the expression and cleavage of cortical and hippocampal APP. Similar isoform-specific interactions between apoE and APP may play a role in the acute, short-term effects of head trauma in humans.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Apolipoproteins E/metabolism , Head Injuries, Closed/metabolism , Animals , Apolipoprotein E3 , Apolipoprotein E4 , Apolipoproteins E/deficiency , Brain Chemistry , Disease Models, Animal , Humans , Immunoblotting , Male , Mice , Mice, Transgenic , Subcellular Fractions/metabolism
5.
Mult Scler ; 9(5): 476-80, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14582773

ABSTRACT

Experimental autoimmune encephalomyelitis (EAE) was found to have a chronic and significantly worse course in apolipoprotein-E (apoE) deficient female mice when compared with matched controls. Disease measures compared included incidence of EAE (64% versus 31%, P < 0.05, chi2 test), maximal clinical score (average +/- SD 2.81 +/- 2.5 versus 0.75 +/- 1.1, P < 0.01, Mann-Whitney test) and mortality (27.3% versus 0%, P = 0.02, Mann-Whitney test and chi2 test). ApoE deficient mice had significantly increased lymphocyte proliferation responses to both myelin antigens and mitogens and significantly more infiltrating lesions in the central nervous system (CNS) in histopathology. Defective neuronal repair mechanisms and enhanced immune reactivity in apoE deficient mice may explain our findings. Clinical implications for MS are discussed.


Subject(s)
Apolipoproteins E/genetics , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Multiple Sclerosis/physiopathology , Alzheimer Disease/immunology , Alzheimer Disease/physiopathology , Animals , Cell Division/immunology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiple Sclerosis/immunology , Th1 Cells/cytology
6.
Neurology ; 57(8): 1482-5, 2001 Oct 23.
Article in English | MEDLINE | ID: mdl-11673593

ABSTRACT

APOE genotype influences the age at onset of some neurodegenerative diseases such as AD and the rate of progression in others such as MS. The authors hypothesize that APOE genotype ubiquitously determines the efficacy of neuronal maintenance and repair in these diseases and that the seemingly divergent clinical effects are due to the stage of disease at which the diagnosis is made. Early diagnosis facilitates the measurement of effects on disease progression rate, whereas late diagnosis results in a marked effect of APOE genotype on disease onset.


Subject(s)
Apolipoproteins E/genetics , Neurodegenerative Diseases/genetics , Age of Onset , Disease Progression , Genotype , Humans , Neurodegenerative Diseases/physiopathology
7.
Cell Growth Differ ; 12(3): 157-67, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11306516

ABSTRACT

The mammalian Rho family GTPases TC10 and Cdc42 share many properties. Activated forms of both proteins stimulate transcription mediated by nuclear factor kappaB, serum response factor, and the cyclin D1 promoter; activate c-Jun NH2-terminal kinase; cooperate with activated Raf to transform NIH-3T3 cells; and, by a mechanism independent of all of these effects, induce filopodia formation. In contrast, previously reported differences between TC10 and Cdc42 are not striking. We now present studies of TC10 and Cdc42 in cell culture that reveal clear functional differences: (a) wild-type TC10 localizes predominantly to the plasma membrane and less extensively to a perinuclear membranous compartment, whereas wild-type Cdc42 localizes predominantly to this compartment and less extensively to the plasma membrane; (b) expression of Rho guanine nucleotide dissociation inhibitor alpha results in a redistribution of wild-type Cdc42 to the cytosol but has no effect on the plasma membrane localization of wild-type TC10; (c) TC10 fails to rescue a Saccharomyces cerevisiae cdc42 mutation, unlike mammalian Cdc42; (d) dominant negative Cdc42, but not dominant negative TC10, inhibits neurite outgrowth in PC12 cells stimulated by nerve growth factor; and (e) activation of nuclear factor kappaB-dependent transcription by Cdc42, but not by TC10, is inhibited by sodium salicylate. These findings point to distinct pathways in which TC10 and Cdc42 may act and distinct modes of regulation of these proteins.


Subject(s)
Cell Compartmentation/physiology , Cell Membrane/enzymology , Cells, Cultured/enzymology , Signal Transduction/physiology , cdc42 GTP-Binding Protein/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , COS Cells , Cell Membrane/ultrastructure , Cells, Cultured/cytology , Green Fluorescent Proteins , Guanine Nucleotide Dissociation Inhibitors/metabolism , HeLa Cells , Humans , Luminescent Proteins/genetics , NF-kappa B/drug effects , NF-kappa B/metabolism , Nerve Growth Factor/pharmacology , Neurites/drug effects , Neurites/metabolism , PC12 Cells/cytology , PC12 Cells/drug effects , PC12 Cells/metabolism , Rats , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism , rho-Specific Guanine Nucleotide Dissociation Inhibitors
8.
Neurology ; 56(4): 529-30, 2001 Feb 27.
Article in English | MEDLINE | ID: mdl-11222800

ABSTRACT

Cerebral necrosis following stroke exposes brain antigens to the immune system, potentially initiating an antibody response. The authors measured levels of antibodies to specific neuronal antigens, neurofilaments (NF), and a ubiquitous antigen, cardiolipin (CL), in 45 patients following an acute first-ever stroke, within 48 hours, and 1, 3, and 6 months later. The mean levels of anti-NF antibodies were elevated compared with baseline at 1, 3, and 6, months (p = 0.012, 0.002, and 0.003 by paired t-test). Anti-CL levels did not change significantly.


Subject(s)
Antibodies/immunology , Brain/immunology , Cardiolipins/blood , Neurofilament Proteins/blood , Stroke/blood , Stroke/immunology , Acute Disease , Aged , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male
9.
Neurology ; 56(3): 312-6, 2001 Feb 13.
Article in English | MEDLINE | ID: mdl-11171894

ABSTRACT

BACKGROUND AND OBJECTIVE: The authors recently reported that the APOE epsilon4 allele is associated with significantly greater progression of disability in a 2-year follow-up of patients with MS. In this study, these findings are substantiated and extended in a much larger group of patients followed for up to 40 years. METHODS: Two hundred five patients with clinically definite MS who were genotyped for the APOE epsilon4 carrier state were included. Groups of patients with (n = 41) and without (n = 164) APOE epsilon4 alleles were compared for latency to expanded disability status scale (EDSS) scores of 4.0 and 6.0 by Kaplan-Meier analysis with the log rank test. The results were adjusted for age at onset and sex by Cox regression analysis. RESULTS: The APOE epsilon4 allele frequency in patients with MS (0.10) was similar to that in the general Israeli population. There was a significant effect of APOE genotype on the latency to reach EDSS 4.0 and 6.0 (p = 0.0002 and p = 0.0006 by two-tailed log rank test). Median latencies were shorter by 12 and 11 years in the APOE epsilon4 group for these outcomes. These results were significant after adjustment for age at onset and sex. CONCLUSIONS: The APOE epsilon4 allele is associated with significantly faster progression of disability in MS. This is the first genetic factor to be identified with a major impact on the progression of disability in this disease.


Subject(s)
Apolipoproteins E/genetics , Multiple Sclerosis/genetics , Multiple Sclerosis/physiopathology , Adult , Disease Progression , Female , Follow-Up Studies , Genotype , Humans , Male , Time Factors
10.
J Cell Biol ; 152(1): 111-26, 2001 Jan 08.
Article in English | MEDLINE | ID: mdl-11149925

ABSTRACT

Determinants of membrane targeting of Rho proteins were investigated in live cells with green fluorescent fusion proteins expressed with or without Rho-guanine nucleotide dissociation inhibitor (GDI)alpha. The hypervariable region determined to which membrane compartment each protein was targeted. Targeting was regulated by binding to RhoGDI alpha in the case of RhoA, Rac1, Rac2, and Cdc42hs but not RhoB or TC10. Although RhoB localized to the plasma membrane (PM), Golgi, and motile peri-Golgi vesicles, TC10 localized to PMs and endosomes. Inhibition of palmitoylation mislocalized H-Ras, RhoB, and TC10 to the endoplasmic reticulum. Although overexpressed Cdc42hs and Rac2 were observed predominantly on endomembrane, Rac1 was predominantly at the PM. RhoA was cytosolic even when expressed at levels in vast excess of RhoGDI alpha. Oncogenic Dbl stimulated translocation of green fluorescent protein (GFP)-Rac1, GFP-Cdc42hs, and GFP-RhoA to lamellipodia. RhoGDI binding to GFP-Cdc42hs was not affected by substituting farnesylation for geranylgeranylation. A palmitoylation site inserted into RhoA blocked RhoGDI alpha binding. Mutations that render RhoA, Cdc42hs, or Rac1, either constitutively active or dominant negative abrogated binding to RhoGDI alpha and redirected expression to both PMs and internal membranes. Thus, despite the common essential feature of the CAAX (prenylation, AAX tripeptide proteolysis, and carboxyl methylation) motif, the subcellular localizations of Rho GTPases, like their functions, are diverse and dynamic.


Subject(s)
Guanine Nucleotide Dissociation Inhibitors/metabolism , rho GTP-Binding Proteins/metabolism , 3T3 Cells , Amino Acid Sequence , Animals , Biological Transport , CHO Cells , COS Cells , Cell Line , Cell Membrane/metabolism , Chlorocebus aethiops , Cricetinae , Dogs , Green Fluorescent Proteins , Guanine Nucleotide Dissociation Inhibitors/genetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Molecular Sequence Data , Palmitic Acid/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , rho GTP-Binding Proteins/genetics , rho-Specific Guanine Nucleotide Dissociation Inhibitors
11.
Neuroscience ; 101(4): 879-84, 2000.
Article in English | MEDLINE | ID: mdl-11113336

ABSTRACT

Apolipoprotein E, the major brain lipid-binding protein, is expressed in humans as three common isoforms (E2, E3 and E4). Previous studies revealed that the allele apolipoprotein E4 is a major genetic risk factor of Alzheimer's disease and that traumatic brain injury is associated with increased risk for developing this disease. Furthermore, it has been suggested that the effects of traumatic head injury and apolipoprotein E4 in Alzheimer's disease are synergistic. To test the hypothesis that the apolipoprotein E genotype affects susceptibility to brain injury, we subjected transgenic mice, expressing either human apolipoprotein E3 or human apolipoprotein E4 on a null mouse apolipoprotein E background and apolipoprotein E-deficient knockouts, to closed head injury and compared mortality, neurological recovery and the extent of brain damage of the survivors. More than 50% of the transgenic mice expressing human apolipoprotein E4 died following closed head injury, whereas only half as many of the transgenic mice expressing human apolipoprotein E3, and of the control and apolipoprotein E-deficient mice died during this period (P<0.02). A neurological severity score used for clinical assessment of the surviving mice up to 11 days after closed head injury revealed that the four mouse groups displayed similar severity of damage at 1h following injury. At three and 11 days post-injury, however, the neurological severity scores of the transgenic mice expressing human apolipoprotein E3 were significantly lower than those of the other three groups whose scores were similar, indicating better recovery of the transgenic mice expressing human apolipoprotein E3. Histopathological examination of the mice performed 11 days post-injury revealed, consistent with the above neurological results, that the size of the damaged brain area of the transgenic mice expressing human apolipoprotein E3 was smaller than that of the other head-injured groups. These findings show that transgenic mice expressing human apolipoprotein E4 are more susceptible than those expressing apolipoprotein E3 to closed head injury. We suggest that this effect is due to both a protective effect of apolipoprotein E3 and an apolipoprotein E4-related pathological function.


Subject(s)
Apolipoproteins E/genetics , Genetic Predisposition to Disease , Head Injuries, Closed/genetics , Alleles , Animals , Apolipoprotein E4 , Brain/pathology , Head Injuries, Closed/mortality , Head Injuries, Closed/pathology , Head Injuries, Closed/physiopathology , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic/genetics , Nervous System/physiopathology , Neuroprotective Agents/metabolism , Reference Values
12.
Cell Death Differ ; 7(9): 825-33, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11042677

ABSTRACT

When PC12 cells are deprived of trophic support they undergo apoptosis. We have previously shown that survival of trophic factor-deprived PC12M1 cells can be promoted by activation of the G protein-coupled muscarinic receptors. The mechanism whereby muscarinic receptors inhibit apoptosis is poorly understood. In the present study we investigated this mechanism by examining the effect of muscarinic receptor activation on the serum deprivation-induced activity of key players in apoptosis, the caspases, in PC12M1 cells. The results showed that m1 muscarinic activation inhibits caspase activity induced by serum deprivation. This effect appeared to be caused by the prevention of activation of caspases such as caspase-2 and caspase-3, and not by the inhibition of existing activity. Muscarinic receptor activation also stimulated the mitogen-activated protein kinase/extracellular signaling-regulated kinase (MAPK/ERK) and phosphoinositide (PI) 3-kinase signaling pathways. The PI 3-kinase pathway inhibitors wortmannin and LY294002, as well as the MAPK/ERK pathway PD98059 inhibitor, did not however suppress the inhibitory effect of the muscarinic receptors on caspase activity. The results therefore suggested that the muscarinic survival effect is mediated by a pathway that leads to caspase inhibition by MAPK/ERK- and PI 3-kinase-independent signaling cascades.


Subject(s)
Apoptosis , Caspases/metabolism , MAP Kinase Signaling System , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases , Receptors, Muscarinic/metabolism , Signal Transduction , Animals , Atropine/pharmacology , Caspase Inhibitors , Caspases/biosynthesis , Cell Extracts , Culture Media, Serum-Free , Dose-Response Relationship, Drug , Enzyme Activation , Enzyme Inhibitors/pharmacology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Muscarinic Agonists/pharmacology , Muscarinic Antagonists/pharmacology , Oxotremorine/pharmacology , PC12 Cells , Peptide Hydrolases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Receptor, Muscarinic M1
13.
Exp Toxicol Pathol ; 52(2): 177-83, 2000 May.
Article in English | MEDLINE | ID: mdl-10965994

ABSTRACT

Previous studies have revealed that apolipoprotein E (apoE)-deficient mice have distinct memory deficits and neurochemical derangements and are oxidatively stressed prior to and following closed head injury. The objective of this study was to evaluate the possibility that the enhanced susceptibility of apoE-deficient mice to closed head injury is related to impairments in their antioxidative iron-chelating mechanisms. ApoE-deficient and control mice were subjected to closed had injury, after which the extent of brain-damage and the level of iron-containing cells were assessed. Examination of the brain-damaged areas in the injured mice revealed that, by Day 3 post injury, animals of both groups were maximally and similarly affected. While the size of the damaged area of the injured control mice diminished significantly by Day 7, however recovery was not observed in injured apoE-deficient mice up to at least 14 days post-injury. Histopathologically, the decrease in the damaged areas in the control mice was interpreted as related to decreased edema. Numbers of iron-containing cells at Days 3 and 7 after injury were greater in the brains of control mice than in the apoE-deficient mice. Whereas the number of iron-containing cells in injured control mice decreased at days 9 and 14-post injury, that of the injured apoE-deficient mice plateaued by Day 9 at a level more than two-fold higher than the maximal level seen for controls. The size of the damaged areas and the number of iron-containing cells were correlated (P < 0.03) for both mouse groups at days 9 and 14 after injury. The data suggest that the increased susceptibility of apoE-deficient mice to closed head injury may be due, at least in part, to impaired iron scavenging and sustained oxidative stress.


Subject(s)
Apolipoproteins E/deficiency , Brain/metabolism , Head Injuries, Closed/metabolism , Iron/metabolism , Animals , Antioxidants , Brain/pathology , Disease Susceptibility , Free Radical Scavengers , Head Injuries, Closed/pathology , Iron Chelating Agents , Kinetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , Necrosis , Oxidative Stress , Reactive Oxygen Species
14.
J Neural Transm Suppl ; 59: 163-9, 2000.
Article in English | MEDLINE | ID: mdl-10961428

ABSTRACT

Genetic studies suggest that the neuropathology and etiology of Alzheimer's disease (AD) are associated with several genotypes including mutations in the amyloid precursor protein (APP) gene and the allele E4 of apolipoprotein E (apoE). The present study investigated the possibility that cross talk interactions exist between APP and apoE and the extent to which they are affected by the apoE genotype. This was pursued by cell culture and immunoblot experiments utilizing neuroblastoma N2a cells in which the effects of distinct apoE isoforms on the levels of intracellular APP and of secreted APPs were determined. This revealed that treatment of the cells with apoE4, the AD risk factor, resulted in a marked increase in the levels of secreted APPs. This effect was dose dependent (ED50 approximately/= 2.5 microg/ml) and isoform specific in that apoE3 had virtually no effect on the secretion of APPs.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Apolipoproteins E/pharmacology , Apolipoprotein E4 , Dose-Response Relationship, Drug , Humans , Immunoblotting , Intracellular Membranes/metabolism , Protein Isoforms/metabolism , Tumor Cells, Cultured
15.
Neuroscience ; 97(3): 419-24, 2000.
Article in English | MEDLINE | ID: mdl-10828524

ABSTRACT

Apolipoprotein E genotype is an important risk factor of Alzheimer's disease, which is associated with the degeneration of distinct brain neuronal systems. In the present study we employed apolipoprotein E-deficient mice and human apolipoprotein E3 and apolipoprotein E4 transgenic mice on a null mouse apolipoprotein E background, to examine the extent to which distinct brain neuronal systems are affected by apolipoprotein E and the isoform specificity of this effect. This was pursued by histological and autoradiographic measurements utilizing neuron specific presynaptic markers. The results thus obtained revealed significant reductions in the levels of brain cholinergic and noradrenergic nerve terminals in young apolipoprotein E-deficient mice and no changes in brain dopaminergic nerve terminals. These cholinergic and noradrenergic presynaptic derangements were ameliorated similarly in human apolipoprotein E3 and apolipoprotein E4 transgenic mice. In the case of the cholinergic system, this resulted in complete reversal of the presynaptic deficits, whereas in the case of the noradrenergic neurons the amelioration was partial. These findings suggest that brain cholinergic and noradrenergic neurons are markedly more dependent on brain apolipoprotein E than brain dopaminergic neurons and that the isoform specificity of these effects is not apparent at a young age under non-challenged conditions.


Subject(s)
Alzheimer Disease/metabolism , Apolipoproteins E/deficiency , Cholinergic Fibers/pathology , Presynaptic Terminals/metabolism , Acetylcholine/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Animals , Apolipoprotein E3 , Apolipoprotein E4 , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Cholinergic Fibers/metabolism , Dopamine/metabolism , Humans , Male , Mice , Mice, Knockout , Norepinephrine/metabolism , Presynaptic Terminals/pathology , Presynaptic Terminals/ultrastructure
16.
J Neurosci Res ; 60(4): 559-64, 2000 May 15.
Article in English | MEDLINE | ID: mdl-10797559

ABSTRACT

Apolipoprotein E (apoE)-deficient mice have learning and memory impairments that are associated with specific neurochemical changes and hyperphosphorylation of distinct epitopes of the cytoskeletal protein tau. Furthermore, such mice are highly susceptible to the sequelae of brain trauma and their ability to recover from head injury is impaired. In the present study we investigated the extent that the neuronal maintenance and repair impairments of apoE-deficient mice are related to aberrations at the tau phosphorylation level. This was pursued by subjecting control and apoE-deficient mice to closed head injury (CHI) and examination, utilizing immunoblot assays, of the resulting effects on tau phosphorylation. The results thus obtained revealed that tau of apoE-deficient mice is hyperphosphorylated before CHI and that this insult results in transient tau hyperphosphorylation, whose extent and time course in the two mouse groups varied markedly. Tau hyperphosphorylation in the injured controls was maximal by about 4 hr after injury and reverted to basal levels by 24 hr. In contrast, almost no head injury-induced tau hyperphosphorylation was observed in the apoE-deficient mice at 4 hr after injury. Some tau hyper-phosphorylation was detected in the head-injured apoE-deficient mice after longer time intervals, but its extent was markedly lower than the maximal values obtained in the head injured controls. These findings show that the chronic neuronal impairments brought about by apoE deficiency and the acute response to head injury are both associated with hyperphosphorylation of the same tau domain and that the ability of apoE-deficient mice to mount the acute tau hyperphosphorylation response to head injury is impaired.


Subject(s)
Apolipoproteins E/deficiency , Brain Injuries/metabolism , Head Injuries, Closed/metabolism , tau Proteins/metabolism , Animals , Apolipoproteins E/genetics , Brain Chemistry , Cytoskeletal Proteins/metabolism , Epitopes/metabolism , Immunoblotting , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Prosencephalon/metabolism , Time Factors
17.
Ann N Y Acad Sci ; 920: 315-20, 2000.
Article in English | MEDLINE | ID: mdl-11193170

ABSTRACT

A cholinergic hypofunction in Alzheimer's disease (AD) may lead to formation of beta-amyloids that might impair the coupling of M1 muscarinic ACh receptors (mAChRs) with G proteins. This disruption in coupling can lead to decreased signal transduction, to a reduction in levels of trophic amyloid precursor proteins (APPs), and to generation of more beta-amyloids that can also suppress ACh synthesis and release, aggravating further the cholinergic deficiency. These "vicious cycles," a presynaptic and a postsynaptic one, may be inhibited, in principle, by M1 selective agonists. Such properties can be detected in the functionally selective M1 agonists from the AF series [e.g., project drugs, AF102B, AF150(S)]. These M1 agonists promote the nonamyloidogenic APP processing pathways and decrease tau protein phosphorylation. The effects on tau proteins suggest a link between M1 mAChR-mediated signal transduction system(s) and the neuronal cytoskeleton via regulation of phosphorylation of tau microtubule-associated protein. This may indicate a dual role for M1 agonists: as inhibitors of two "vicious cycles," one induced by beta-amyloids, and the other due to overactivation of certain kinases (e.g., glycogen synthase kinase-3, GSK-3) or downregulation of phosphatases, respectively. Prolonged administration of AF150(S) in apolipoprotein E-knockout mice restored cognitive impairments, cholinergic hypofunction, and tau hyperphosphorylation, and unveiled a high-affinity binding site to M1 mAChRs. Except M1 agonists, there are no reports of compounds having such combined effects, for example, amelioration of cognition dysfunction and beneficial modulation of APPs together with tau phosphorylation. This unique property of M1 agonists to alter different aspects of AD pathogenesis could represent the most remarkable, yet unexplored, clinical value of such compounds.


Subject(s)
Alzheimer Disease/drug therapy , Muscarinic Agonists/therapeutic use , Receptors, Muscarinic/physiology , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Disease Models, Animal , Humans , Protein Processing, Post-Translational , Receptor, Muscarinic M1
18.
J Neurosci Res ; 58(4): 586-92, 1999 Nov 15.
Article in English | MEDLINE | ID: mdl-10533050

ABSTRACT

Apolipoprotein E (ApoE) is the major brain lipoprotein and plays an important role in lipid transport. ApoE-deficient mice whose apoE gene has been knocked out have distinct cognitive and neurochemical deficits, and their recovery from brain injury is impaired. In the present study we examined the possibility that the neuronal derangements of apoE-deficient mice are related to impairments in their phospholipid metabolism. This was performed by comparison of the phospholipid, fatty acid, and cholesterol compositions of distinct membranal brain fractions of apoE-deficient and control mice. Analysis of the microsomal membrane fraction P(3) revealed that, in apoE-deficient mice, these membranes contain significantly lower levels of phosphatidylcholine (PC) than those of control mice. This effect was specific to PC and thus resulted in a twofold decrease of the PC to phosphatidylethanolamine (PE) ratio in apoE-deficient mice compared to the corresponding control ratio. In contrast, the cholesterol levels of the microsomal membranes of the two mice were similar, and the fatty acid composition of their PC was unchanged. There were, however, changes in the fatty acid composition of PE and phosphatidylserine (PS), which resulted in a lower ratio of polyunsaturated to saturated fatty acids in PE and in a higher ratio in apoE-deficient mice compared to the corresponding control values. These effects were specific to the microsomal fraction P(3) and were not observed with the brain subcellular membrane fraction P(2), which is composed mainly of plasma and mitochondrial membranes and whose phospholipid, fatty acid, and cholesterol levels were similar in apoE-deficient and control mice. These findings show that apoE deficiency results in specific and intracellular compartment-dependent changes in phospholipid metabolism, which may play an important role in mediating the neuronal effects of apoE.


Subject(s)
Apolipoproteins E/deficiency , Brain Chemistry/physiology , Phospholipids/metabolism , Animals , Cholesterol/metabolism , Fatty Acids/chemistry , Fatty Acids/metabolism , In Vitro Techniques , Male , Membranes/chemistry , Membranes/metabolism , Mice , Microsomes/chemistry , Microsomes/metabolism , Mitochondria/chemistry , Mitochondria/metabolism , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/metabolism , Subcellular Fractions/metabolism
19.
Cell ; 98(1): 69-80, 1999 Jul 09.
Article in English | MEDLINE | ID: mdl-10412982

ABSTRACT

We show that Nras is transiently localized in the Golgi prior to the plasma membrane (PM). Moreover, green fluorescent protein (GFP)-tagged Nras illuminated motile, peri-Golgi vesicles, and prolonged BFA treatment blocked PM expression. GFP-Hras colocalized with GFP-Nras, but GFP-Kras4B revealed less Golgi and no vesicular fluorescence. Whereas a secondary membrane targeting signal was required for PM expression, the CAAX motif alone was necessary and sufficient to target proteins to the endomembrane where they were methylated, a modification required for efficient membrane association. Thus, prenylated CAAX proteins do not associate directly with the PM but instead associate with the endomembrane and are subsequently transported to the PM, a process that requires a secondary targeting motif.


Subject(s)
Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , ras Proteins/metabolism , Amino Acid Sequence , Animals , CHO Cells , COS Cells , Cell Line , Cricetinae , Dogs , Green Fluorescent Proteins , Kidney , Kinetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Recombinant Fusion Proteins/metabolism , Transfection , ras Proteins/chemistry , ras Proteins/genetics
20.
Biochim Biophys Acta ; 1453(3): 359-68, 1999 Mar 30.
Article in English | MEDLINE | ID: mdl-10101254

ABSTRACT

Apolipoprotein E deficient mice have distinct memory deficits and neurochemical derangements and their recovery from closed head injury is impaired. In the present study, we examined the possibility that the neuronal derangements of apolipoprotein E deficient mice are associated with oxidative stress, which in turn affects their ability to recover from close head injury. It was found that brain phospholipid levels in apolipoprotein E deficient mice are lower than those of the controls (55+/-15% of control, P<0. 01), that the cholesterol levels of the two mice groups are similar and that the levels of conjugated dienes of the apolipoprotein E deficient mice are higher than those of control mice (132+/-15% of P<0.01). Brains of apolipoprotein E deficient mice had higher Mn-superoxide dismutase (134+/-7%), catalase (122+/-8%) and glutathione reductase (167+/-7%) activities than control (P<0.01), whereas glutathione peroxidase activity and the levels of reduced glutathione and ascorbic acid were similar in the two mouse groups. Closed head injury increased catalase and glutathione peroxidase activities in both mouse groups, whereas glutathione reductase increased only in control mice. The superoxide dismutase activity was unaffected in both groups. These findings suggest that the antioxidative metabolism of apolipoprotein E deficient mice is altered both prior to and following head injury and that antioxidative mechanisms may play a role in mediating the neuronal maintenance and repair derangements of the apolipoprotein E deficient mice.


Subject(s)
Apolipoproteins E/deficiency , Brain Chemistry , Head Injuries, Closed/metabolism , Oxidative Stress , Animals , Antioxidants/analysis , Ascorbic Acid/analysis , Catalase/analysis , Glutathione/analysis , Glutathione Peroxidase/analysis , Glutathione Reductase/analysis , Glutathione Transferase/analysis , Male , Mice , Superoxide Dismutase/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...