Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
2.
FEBS Lett ; 596(22): 2914-2927, 2022 11.
Article in English | MEDLINE | ID: mdl-35971617

ABSTRACT

Previous studies have shown that amyloid-ß oligomers (AßO) bind with high affinity to cellular prion protein (PrPC ). The AßO-PrPC complex binds to cell-surface co-receptors, including the laminin receptor (67LR). Our current studies revealed that in Neuroscreen-1 cells, 67LR is the major co-receptor involved in the cellular uptake of AßO and AßΟ-induced cell death. Both pharmacological (dibutyryl-cAMP, forskolin and rolipram) and physiological (pituitary adenylate cyclase-activating polypeptide) cAMP-elevating agents decreased cell-surface PrPC and 67LR, thereby attenuating the uptake of AßO and the resultant neuronal cell death. These cAMP protective effects are dependent on protein kinase A, but not dependent on the exchange protein directly activated by cAMP. Conceivably, cAMP protects neuronal cells from AßO-induced cytotoxicity by decreasing cell-surface-associated PrPC and 67LR.


Subject(s)
Amyloid beta-Peptides , PrPC Proteins , Amyloid beta-Peptides/metabolism , Prion Proteins , PrPC Proteins/metabolism , Laminin/metabolism , Cell Death , Receptors, Laminin/genetics , Pituitary Adenylate Cyclase-Activating Polypeptide
3.
J Cereb Blood Flow Metab ; 40(12): 2387-2400, 2020 12.
Article in English | MEDLINE | ID: mdl-31987006

ABSTRACT

The receptor tyrosine kinase PDGFRß is essential for pericyte migration to the endothelium. In mice lacking one allele of PDGFRß (PDGFRß+/-), previous reports have described an age-dependent loss of pericytes in the brain, leading to cerebrovascular dysfunction and subsequent neurodegeneration reminiscent of that seen in Alzheimer's disease and vascular dementia. We examined 12-20-month-old PDGFRß+/- mice to better understand how pericyte loss affects brain microvascular structure and perfusion in vivo. We observed a mild reduction of cortical pericyte number in PDGFRß+/- mice (27% fewer cell bodies) compared to controls, but no decrease in pericyte coverage of the endothelium. This mild degree of pericyte loss caused no discernable change in cortical microvascular density, length, basal diameter or reactivity to hypercapnia. Yet, it was associated with an increase in basal blood cell velocity, primarily in pre-capillary arterioles. Taken together, our results suggest that mild pericyte loss can lead to aberrant cerebral blood flow despite a lack of apparent effect on microvascular structure and reactivity.


Subject(s)
Brain/blood supply , Endothelium/metabolism , Pericytes/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Age Factors , Alleles , Alzheimer Disease/metabolism , Animals , Arterioles/cytology , Arterioles/metabolism , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Brain/physiopathology , Capillaries/cytology , Capillaries/metabolism , Case-Control Studies , Cerebrovascular Circulation/physiology , Endothelium/cytology , Female , Hypercapnia/metabolism , Hypercapnia/physiopathology , Male , Mice
5.
J Cereb Blood Flow Metab ; 39(3): 411-425, 2019 03.
Article in English | MEDLINE | ID: mdl-28933255

ABSTRACT

Smooth muscle cells and pericytes, together called mural cells, coordinate many distinct vascular functions. Canonically, smooth muscle cells are ring-shaped and cover arterioles with circumferential processes, whereas pericytes extend thin processes that run longitudinally along capillaries. In between these canonical mural cell types are cells with features of both smooth muscle cells and pericytes. Recent studies suggest that these transitional cells are critical for controlling blood flow to the capillary bed during health and disease, but there remains confusion on how to identify them and where they are located in the brain microvasculature. To address this issue, we measured the morphology, vascular territory, and α-smooth muscle actin content of structurally diverse mural cells in adult mouse cortex. We first imaged intact 3D vascular networks to establish the locations of major gradations in mural cell appearance as arterioles branched into capillaries. We then imaged individual mural cells occupying the regions within these gradations. This revealed two transitional cells that were often similar in appearance, but with sharply contrasting levels of α-smooth muscle actin. Our findings highlight the diversity of mural cell morphologies in brain microvasculature, and provide guidance for identification and categorization of mural cell types.


Subject(s)
Brain/blood supply , Cerebral Cortex/cytology , Microvessels/cytology , Myocytes, Smooth Muscle/cytology , Pericytes/cytology , Actins/analysis , Animals , Arterioles/anatomy & histology , Capillaries/anatomy & histology , Cerebral Cortex/anatomy & histology , Cerebral Cortex/blood supply , Cerebral Cortex/diagnostic imaging , Mice , Microscopy, Confocal/methods , Microvessels/diagnostic imaging
6.
Front Aging Neurosci ; 10: 210, 2018.
Article in English | MEDLINE | ID: mdl-30065645

ABSTRACT

The biology of brain microvascular pericytes is an active area of research and discovery, as their interaction with the endothelium is critical for multiple aspects of cerebrovascular function. There is growing evidence that pericyte loss or dysfunction is involved in the pathogenesis of Alzheimer's disease, vascular dementia, ischemic stroke and brain injury. However, strategies to mitigate or compensate for this loss remain limited. In this review, we highlight a novel finding that pericytes in the adult brain are structurally dynamic in vivo, and actively compensate for loss of endothelial coverage by extending their far-reaching processes to maintain contact with regions of exposed endothelium. Structural remodeling of pericytes may present an opportunity to foster pericyte-endothelial communication in the adult brain and should be explored as a potential means to counteract pericyte loss in dementia and cerebrovascular disease. We discuss the pathophysiological consequences of pericyte loss on capillary function, and the biochemical pathways that may control pericyte remodeling. We also offer guidance for observing pericytes in vivo, such that pericyte structural remodeling can be more broadly studied in mouse models of cerebrovascular disease.

7.
Cell Rep ; 22(1): 8-16, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29298435

ABSTRACT

Direct contact and communication between pericytes and endothelial cells is critical for maintenance of cerebrovascular stability and blood-brain barrier function. Capillary pericytes have thin processes that reach hundreds of micrometers along the capillary bed. The processes of adjacent pericytes come in close proximity but do not overlap, yielding a cellular chain with discrete territories occupied by individual pericytes. Little is known about whether this pericyte chain is structurally dynamic in the adult brain. Using in vivo two-photon imaging in adult mouse cortex, we show that while pericyte somata were immobile, the tips of their processes underwent extensions and/or retractions over days. The selective ablation of single pericytes provoked exuberant extension of processes from neighboring pericytes to contact uncovered regions of the endothelium. Uncovered capillary regions had normal barrier function but were dilated until pericyte contact was regained. Pericyte structural plasticity may be critical for cerebrovascular health and warrants detailed investigation.


Subject(s)
Blood-Brain Barrier/metabolism , Capillaries/metabolism , Endothelial Cells/metabolism , Pericytes/metabolism , Animals , Blood-Brain Barrier/cytology , Capillaries/cytology , Endothelial Cells/cytology , Mice , Mice, Transgenic , Pericytes/cytology
8.
J Lipid Res ; 57(11): 2028-2039, 2016 11.
Article in English | MEDLINE | ID: mdl-27623848

ABSTRACT

Th1 pro-inflammatory cytokines, i.e., TNF-α and IFN-γ, in combination are known to induce cell death in several cell types, including oligodendrocytes, but the mechanism of their synergistic cytotoxicity is unclear. Although ceramide (Cer) has been implicated in cytokine- and stress-induced cell death, its intracellular levels alone cannot explain cytokine synergy. We considered the possibility that Cer released as part of extracellular vesicles may contribute to cytokine-induced synergistic cell death. Using a human oligodendroglioma (HOG) cell line as a model, here we show that exosomes derived from TNF-α-treated "donor" cells, while being mildly toxic to fresh cultures (similar to individual cytokines), induce enhanced cell death when added to IFN-γ-primed target cultures in a fashion resembling the effect of cytokine combination. Further, the sphingolipid profiles of secreted exosomes, as determined by HPLC-MS/MS, revealed that the treatment with the cytokines time-dependently induced the formation and exosomal release, in particular of C16-, C24-, and C24:1-Cer species; C16-, C24-, and C24:1-dihydroCer species; and C16-, C24-, and C24:1-SM species. Finally, exogenous C6-Cer or C16-Cer mimicked and enhanced the cytotoxic effects of the cytokines upon HOG cells, thereby supporting the cell death-signaling role of extracellular Cer.


Subject(s)
Ceramides/metabolism , Interferon-gamma/metabolism , Oligodendroglioma/metabolism , Tumor Necrosis Factor-alpha/metabolism , Cell Death/genetics , Cell Line, Tumor , Ceramides/chemistry , Ceramides/genetics , Chromatography, High Pressure Liquid , Exosomes , Extracellular Vesicles/metabolism , Humans , Interferon-gamma/administration & dosage , Interferon-gamma/genetics , Oligodendroglia/metabolism , Oligodendroglia/pathology , Oligodendroglioma/pathology , Sphingolipids/chemistry , Sphingolipids/metabolism , Tandem Mass Spectrometry , Tumor Necrosis Factor-alpha/administration & dosage , Tumor Necrosis Factor-alpha/genetics
9.
Eur J Neurosci ; 43(10): 1298-306, 2016 05.
Article in English | MEDLINE | ID: mdl-26369866

ABSTRACT

A distributed network of neurons regulates wake, non-rapid eye movement (NREM) sleep, and REM sleep. However, there are also glia in the brain, and there is growing evidence that neurons and astroglia communicate intimately to regulate behaviour. To identify the effect of optogenetic stimulation of astrocytes on sleep, the promoter for the astrocyte-specific cytoskeletal protein, glial fibrillary acidic protein (GFAP) was used to direct the expression of channelrhodopsin-2 (ChR2) and the linked reporter gene, enhanced yellow fluorescent protein (EYFP), in astrocytes. rAAV-GFAP-ChR2 (H134R)-EYFP or rAAV-GFAP-EYFP was microinjected (750 nL) into the posterior hypothalamus (bilateral) of mice. Three weeks later baseline sleep was recorded (0 Hz) and 24 h later optogenetic stimulation applied during the first 6 h of the lights-off period. Mice with ChR2 were given 5, 10 or 30 Hz stimulation for 6 h (10-ms pulses; 1 mW; 1 min on 4 min off). At least 36 h elapsed between the stimulation periods (5, 10, 30 Hz) and although 0 Hz was always first, the order of the other three stimulation rates was randomised. In mice with ChR2 (n = 7), 10 Hz, but not 5 or 30 Hz stimulation increased both NREM and REM sleep during the 6-h period of stimulation. Delta power did not increase. In control mice (no ChR2; n = 5), 10 Hz stimulation had no effect. This study demonstrates that direct stimulation of astrocytes powerfully induces sleep during the active phase of the sleep-wake cycle and underlines the inclusion of astrocytes in network models of sleep-wake regulation.


Subject(s)
Astrocytes/physiology , Hypothalamus, Posterior/physiology , Optogenetics , Sleep , Animals , Female , Male , Mice , Mice, Inbred C57BL , Sleep, REM
10.
J Alzheimers Dis ; 46(3): 581-91, 2015.
Article in English | MEDLINE | ID: mdl-26402511

ABSTRACT

Using a variety of animal models of Alzheimer's disease (AD), there have been a number of recent studies reporting varying degrees of success with anti-AD therapeutics. The efficacies are often discussed in terms of the modulatory effects of the compounds tested on identified or assumed targets among the known (or proposed) pathogenic and neuroprotective mechanisms, largely within the context of the dominant amyloid cascade hypothesis. However, it is clear that several of the relatively more efficacious treatments tend to be multifunctional and target multiple pathological processes associated with AD including most commonly, oxidative and metabolic stress and neuroinflammation. Increasing evidence suggests that vascular and neurodegenerative pathologies often co-exist and that neurovascular dysfunction plays a critical role in the development or progression of AD. In this review, we will discuss the significance of vasculoprotection or neurovascular unit integrity as a common, multi-targeted mechanism underlying the reported efficacy of a majority of anti-AD therapeutics--amyloid-targeted or otherwise--while providing a strong support for future neurovascular-based treatment strategies and interventions.


Subject(s)
Alzheimer Disease/therapy , Amyloid beta-Peptides/metabolism , Antipsychotic Agents/therapeutic use , Cerebrovascular Disorders/prevention & control , Neuroprotective Agents/therapeutic use , Amyloid beta-Peptides/drug effects , Animals , Humans , Oxidative Stress/drug effects
11.
J Neurochem ; 128(6): 919-26, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24188160

ABSTRACT

Tumor progression locus 2 (Tpl2)/cancer Osaka thyroid kinase is a newer member of MAP3K family that is now known for its essential role in tumor necrosis factor-aplha (TNFα) expression in macrophages, but its pro-inflammatory signaling, if any, in glia is unknown. When cultures of murine microglia and astrocytes were exposed to lipopolysaccharide, there was a rapid activation (i.e., phosphorylation) of Tpl2 in parallel to the activation of down-stream effector MAPKs, that is, extracellular signal regulated kinase (ERK), p38 MAPK and C-Jun N-terminal kinase (JNK). Pre-incubation of the cultures with a Tpl2 inhibitor selectively suppressed the activation of the primary down-stream target, that is, ERK relative to p38 MAPK and JNK. That Tpl2 activation was functionally involved in glial inflammatory response was indicated by a reduced release of the cytokines, i.e. TNFα and the expression of inducible nitric oxide synthase in the presence of the kinase inhibitor. Furthermore, over-expression of a wild-type Tpl2 construct in C-6 glia resulted in an enhanced transcriptional activation of inducible nitric oxide synthase, while transfection with a dominant negative form of Tpl-2 had the opposite effect. The findings assign an important pro-inflammatory signaling function for Tpl2 pathway in glial cells.


Subject(s)
Astrocytes/enzymology , MAP Kinase Kinase Kinases/immunology , MAP Kinase Kinase Kinases/metabolism , Microglia/enzymology , Neuritis/metabolism , Proto-Oncogene Proteins/immunology , Proto-Oncogene Proteins/metabolism , Animals , Astrocytes/cytology , Astrocytes/immunology , Cells, Cultured , Female , Gene Expression/immunology , Lipopolysaccharides/pharmacology , MAP Kinase Kinase Kinases/genetics , MAP Kinase Signaling System/immunology , Male , Mice , Mice, Inbred C57BL , Microglia/cytology , Microglia/immunology , Neuritis/chemically induced , Neuritis/immunology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Pregnancy , Proto-Oncogene Proteins/genetics , Tumor Necrosis Factor-alpha/metabolism
12.
J Alzheimers Dis ; 36(4): 781-9, 2013.
Article in English | MEDLINE | ID: mdl-23703152

ABSTRACT

Previous studies demonstrated that a high fat/high cholesterol (HFC) diet results in a loss of working memory in mice correlated with neuroinflammatory changes and increased AßPP processing (Thirumangalakudi et al. (2008) J Neurochem 106, 475-485). To further explore the nature of the molecular correlates of cognitive impairment, in this study, we examined changes in tau phosphorylation, insulin/IGF-1 signaling (IIS) including GSK3, and levels of specific synaptic proteins. Immunoblot analysis of hippocampal tissue from C57BL/6 mice fed HFC for 2 months with anti-phospho-tau (i.e., PHF1 and phospho-Thr-231 tau) antibodies demonstrated the presence of hyperphosphorylated tau. The tau phosphorylation correlated with activated GSK3, a prominent tau kinase normally kept inactive under the control of IIS. That IIS itself was impaired due to the hyperlipidemic diet was confirmed by a down-regulation of insulin receptor substrate-1 and phospho-Akt levels. Although no significant changes in the levels of the pre-synaptic protein (i.e., synaptophysin) in response to HFC were apparent in immunoblot analysis, there was a clear down-regulation of the post-synaptic protein, PSD95, and drebrin, a dendritic spine-specific protein, indicative of altered synaptic plasticity. The results, in concert with previous findings with the same model, suggest that high dietary fat/cholesterol elicits brain insulin resistance and altered IIS leading to Alzheimer's disease-like cognitive impairment in 'normal' mice.


Subject(s)
Cholesterol, Dietary/metabolism , Diet, High-Fat , Hippocampus/metabolism , Insulin-Like Growth Factor I/metabolism , Insulin/metabolism , Signal Transduction/physiology , tau Proteins/metabolism , Animals , Brain/metabolism , Cholesterol, Dietary/adverse effects , Diet, High-Fat/adverse effects , Hippocampus/physiology , Insulin Resistance/physiology , Mice , Mice, Inbred C57BL , Phosphorylation/physiology
13.
Cell Metab ; 17(5): 685-94, 2013 May 07.
Article in English | MEDLINE | ID: mdl-23663737

ABSTRACT

Classic cardio-metabolic risk factors such as hypertension, stroke, diabetes, and hypercholesterolemia all increase the risk of Alzheimer's disease. We found increased transcription of ß-secretase/BACE1, the rate-limiting enzyme for Aß generation, in eNOS-deficient mouse brains and after feeding mice a high-fat, high-cholesterol diet. Up- or downregulation of PGC-1α reciprocally regulated BACE1 in vitro and in vivo. Modest fasting in mice reduced BACE1 transcription in the brains, which was accompanied by elevated PGC-1 expression and activity. Moreover, the suppressive effect of PGC-1 was dependent on activated PPARγ, likely via SIRT1-mediated deacetylation in a ligand-independent manner. The BACE1 promoter contains multiple PPAR-RXR sites, and direct interactions among SIRT1-PPARγ-PGC-1 at these sites were enhanced with fasting. The interference on the BACE1 gene identified here represents a unique noncanonical mechanism of PPARγ-PGC-1 in transcriptional repression in neurons in response to metabolic signals that may involve recruitment of corepressor NCoR.


Subject(s)
Alzheimer Disease/genetics , Amyloid Precursor Protein Secretases/genetics , Neurons/metabolism , PPAR gamma/genetics , Sirtuin 1/genetics , Stress, Physiological/physiology , Transcription Factors/genetics , Acetylation , Alzheimer Disease/enzymology , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amyloid Precursor Protein Secretases/biosynthesis , Amyloid Precursor Protein Secretases/metabolism , Animals , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Down-Regulation , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type III/deficiency , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , PPAR gamma/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Promoter Regions, Genetic , Rats , Sirtuin 1/metabolism , Stress, Physiological/genetics , Transcription Factors/metabolism , Transcription, Genetic , Up-Regulation
14.
FASEB J ; 27(3): 1127-36, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23239823

ABSTRACT

Remyelination has to occur to fully regenerate injured spinal cords or brain tissues. A growing body of evidence has suggested that exogenous cell transplantation is one promising strategy to promote remyelination. However, direct injection of neural stem cells or oligodendrocyte progenitor cells (OPCs) to the lesion site may not be an optimal therapeutic strategy due to poor viability and functionality of transplanted cells resulted from the local hostile tissue environment. The overall objective of this study was to engineer an injectable biocompatible hydrogel system as a supportive niche to provide a regeneration permissive microenvironment for transplanted OPCs to survive, functionally differentiate, and remyelinate central nervous system (CNS) lesions. A highly biocompatible hydrogel, based on thiol-functionalized hyaluronic acid and thiol-functionalized gelatin, which can be crosslinked by poly-(ethylene glycol) diacrylate (PEGDA), was used. These hydrogels were optimized first regarding cell adhesive properties and mechanical properties to best support the growth properties of OPCs in culture. Transplanted OPCs with the hydrogels optimized in vitro exhibited enhanced survival and oligodendrogenic differentiation and were able to remyelinate demyelinated axons inside ethidium bromide (EB) demyelination lesion in adult spinal cord. This study provides a new possible therapeutic approach to treat CNS injuries in which cell therapies may be essential.


Subject(s)
Demyelinating Diseases/therapy , Hydrogels , Myelin Sheath/metabolism , Neural Stem Cells/transplantation , Oligodendroglia/transplantation , Spinal Cord Injuries/therapy , Stem Cell Transplantation , Animals , Axons/metabolism , Axons/pathology , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Female , Gelatin/chemistry , Gelatin/pharmacology , Humans , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Hydrogels/chemistry , Hydrogels/pharmacology , Myelin Sheath/pathology , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Oligodendroglia/metabolism , Oligodendroglia/pathology , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , Rats , Rats, Nude , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , Transplantation, Homologous
15.
J Alzheimers Dis ; 25(2): 295-307, 2011.
Article in English | MEDLINE | ID: mdl-21422528

ABSTRACT

In addition to its function in calcium and bone metabolism, vitamin D is neuroprotective and important for mitigating inflammation. Alzheimer's disease (AD) is a progressive neurodegenerative disorder of the central nervous system, characterized by neuronal loss in many areas of the brain, and the formation of senile (neuritic) plaques, which increase in number and size over time. The goal of this project was to investigate whether vitamin D3 supplementation would affect amyloid plaque formation in amyloid-ß protein precursor (AßPP) transgenic mice that spontaneously develop amyloid plaques within 3-4 months of birth. AßPP mice were fed control, vitamin D3-deficient or vitamin D3-enriched diets for five months, starting immediately after weaning. At the end of the study, the animals were subjected to behavioral studies, sacrificed, and examined for bone changes and brain amyloid load, amyloid-ß (Aß) peptide levels, inflammatory changes, and nerve growth factor (NGF) content. The results obtained indicate that a vitamin D3-enriched diet correlates with a decrease in the number of amyloid plaques, a decrease in Aß peptides, a decrease in inflammation, and an increase in NGF in the brains of AßPP mice. These observations suggest that a vitamin D3-enriched diet may benefit AD patients.


Subject(s)
Brain/drug effects , Brain/metabolism , Cholecalciferol/administration & dosage , Plaque, Amyloid/diet therapy , Plaque, Amyloid/pathology , Amyloid beta-Protein Precursor/genetics , Animals , Bone and Bones/metabolism , Bone and Bones/pathology , Cholecalciferol/blood , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay/methods , Humans , Learning Disabilities/diet therapy , Learning Disabilities/etiology , Male , Maze Learning/drug effects , Maze Learning/physiology , Memory Disorders/diet therapy , Memory Disorders/etiology , Mice , Mice, Transgenic , NFI Transcription Factors/metabolism , Nerve Growth Factor/metabolism , Plaque, Amyloid/genetics , Presenilin-1/genetics , Tumor Necrosis Factor-alpha/metabolism
16.
J Neurochem ; 117(3): 359-74, 2011 May.
Article in English | MEDLINE | ID: mdl-21320126

ABSTRACT

Current evidence suggests that Alzheimer's disease (AD) is a multi-factorial disease that starts with accumulation of multiple proteins. We have previously proposed that inhibition of γ-secretase may impair membrane recycling causing neurodegeneration starting at synapses (Sambamurti K., Suram A., Venugopal C., Prakasam A., Zhou Y., Lahiri D. K. and Greig N. H. A partial failure of membrane protein turnover may cause Alzheimer's disease: a new hypothesis. Curr. Alzheimer Res., 3, 2006, 81). We also proposed familal AD mutations increase Aß42 by inhibiting γ-secretase. Herein, we discuss the failure of Eli Lilly's γ-secretase inhibitor, semagacestat, in clinical trials in the light of our hypothesis, which extends the problem beyond toxicity of Aß aggregates. We elaborate that γ-secretase inhibitors lead to accumulation of amyloid precursor protein C-terminal fragments that can later be processed by γ-secretase to yields bursts of Aß to facilitate aggregation. Although we do not exclude a role for toxic Aß aggregates, inhibition of γ-secretase can affect numerous substrates other than amyloid precursor protein to affect multiple pathways and the combined accumulation of multiple peptides in the membrane may impair its function and turnover. Taken together, protein processing and turnover pathways play an important role in maintaining cellular homeostasis and unless we clearly see consistent disease-related increase in their levels or activity, we need to focus on preserving their function rather than inhibiting them for treatment of AD and similar diseases.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Enzyme Inhibitors/therapeutic use , Animals , Humans
17.
Brain Pathol ; 21(3): 330-48, 2011 May.
Article in English | MEDLINE | ID: mdl-20969653

ABSTRACT

Prenatal systemic inflammation has been implicated in neurological diseases, but optimal animal models have not been developed. We investigated whether a partial genetic deletion of glial cell line-derived neurotrophic factor (Gdnf(+/-)) increased vulnerability of dopamine (DA) neurons to prenatal lipopolysaccharide (LPS). LPS [0.01 mg/kg intraperitoneal (i.p.)] or saline was administered to wild-type (WT) or Gdnf(+/-) pregnant mice on gestational day 9.5. Male offspring were examined at 3 weeks, 3 and 12 months of age. There was a progressive degeneration of tyrosine hydroxylase (TH)-positive neurons in the substantia nigra (SN) with age in Gdnf(+/-) but not in WT mice, with no observed effects on locus coeruleus (LC) noradrenergic neurons or DA neurons of the ventral tegmental area. Inflammatory markers were elevated in SN of LPS treated offspring, with exacerbation in Gdnf(+/-) mice. Intracellular accumulation of α-synuclein (α-syn) immunoreactivity in DA neurons of SN was observed in all groups of Gdnf(+/-) and in WT mice with prenatal LPS, with altered distribution between pars reticulata (pr) and pars compacta (pc). The findings suggest that prenatal LPS leads to accelerated neuropathology in the SN with age, and that a partial loss of GDNF exacerbates these effects, providing a novel model for age-related neuropathology of the nigrostriatal DA system.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor/metabolism , Neurodegenerative Diseases/pathology , Prenatal Exposure Delayed Effects/immunology , Substantia Nigra/immunology , Tyrosine 3-Monooxygenase/metabolism , Age Factors , Animals , Disease Models, Animal , Dopamine/metabolism , Female , Glial Cell Line-Derived Neurotrophic Factor/genetics , Glial Cell Line-Derived Neurotrophic Factor/immunology , Heterozygote , Lipopolysaccharides/immunology , Male , Mice , Mice, Knockout , Neostriatum/immunology , Neostriatum/metabolism , Neostriatum/pathology , Neurodegenerative Diseases/immunology , Neurodegenerative Diseases/metabolism , Neurons/immunology , Neurons/metabolism , Neurons/pathology , Pregnancy , Substantia Nigra/metabolism , Tumor Necrosis Factor-alpha/metabolism
18.
J Neurochem ; 115(3): 551-62, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20807313

ABSTRACT

There is increasing evidence that the incidence of Alzheimer's disease (AD) is significantly influenced by cardiovascular risk factors in association with a cluster of metabolic diseases including diabetes and atherosclerosis. The shared risk is also reflected in the dietary and lifestyle links to both metabolic disorders and AD-type cognitive dysfunction. Recent studies with genetic and diet-induced animal models have begun to illuminate convergent mechanisms and mediators between these two categories of disease conditions with distinct tissue-specific pathologies. Although it is clear that peripheral inflammation and insulin resistance are central to the pathogenesis of the disorders of metabolic syndrome, it seems that the same mechanisms are also in play across the blood-brain barrier that lead to AD-like molecular and cognitive changes. This review highlights these convergent mechanisms and discusses the role of cerebrovascular dysfunction as a conduit to brain emergence of these pathogenic processes that might also represent future therapeutic targets in AD in common with metabolic disorders.


Subject(s)
Alzheimer Disease/complications , Alzheimer Disease/physiopathology , Heart Diseases/complications , Heart Diseases/physiopathology , Metabolic Diseases/complications , Metabolic Diseases/physiopathology , Alzheimer Disease/epidemiology , Atherosclerosis/complications , Cerebrovascular Disorders/physiopathology , Diabetes Complications/physiopathology , Diet , Humans , Insulin Resistance , Life Style , Metabolic Syndrome/complications , Metabolic Syndrome/physiopathology , Risk
19.
J Neurochem ; 106(1): 475-85, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18410513

ABSTRACT

Recent findings suggest that hypercholesterolemia may contribute to the onset of Alzheimer's disease-like dementia but the underlying mechanisms remain unknown. In this study, we evaluated the cognitive performance in rodent models of hypercholesterolemia in relation to neuroinflammatory changes and amyloid precursor protein (APP) processing, the two key parameters of Alzheimer's disease pathogenesis. Groups of normal C57BL/6 and low density lipoprotein receptor (LDLR)-deficient mice were fed a high fat/cholesterol diet for an 8-week period and tested for memory in a radial arm maze. It was found that the C57BL/6 mice receiving a high fat diet were deficient in handling an increasing working memory load compared with counterparts receiving a control diet while the hypercholesterolemic LDLR-/- mice showed impaired working memory regardless of diet. Immunohistochemical analysis revealed the presence of activated microglia and astrocytes in the hippocampi from high fat-fed C57BL/6 mice and LDLR-/- mice. Consistent with a neuroinflammatory response, the hyperlipidemic mice showed increased expression of cytokines/mediators including tumor necrosis factor-alpha, interleukin-1beta and -6, nitric oxide synthase 2, and cycloxygenase 2. There was also an induced expression of the key APP processing enzyme i.e. beta-site APP cleaving enzyme 1 in both high fat/cholesterol-fed C57BL/6 and LDLR-/- mice accompanied by an increased generation of C-terminal fragments of APP. Although ELISA for beta-amyloid failed to record significant changes in the non-transgenic mice, a threefold increase in beta-amyloid 40 accumulation was apparent in a strain of transgenic mice expressing wild-type human APP on high fat/cholesterol diet. The findings link hypercholesterolemia with cognitive dysfunction potentially mediated by increased neuroinflammation and APP processing in a non-transgenic mouse model.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Cholesterol/metabolism , Encephalitis/metabolism , Hypercholesterolemia/metabolism , Memory Disorders/metabolism , Memory, Short-Term , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/biosynthesis , Animals , Aspartic Acid Endopeptidases/metabolism , Brain/metabolism , Brain/physiopathology , Cytokines/metabolism , Dietary Fats/adverse effects , Dietary Fats/metabolism , Disease Models, Animal , Encephalitis/etiology , Encephalitis/physiopathology , Hypercholesterolemia/complications , Hypercholesterolemia/physiopathology , Maze Learning , Memory Disorders/etiology , Memory Disorders/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Neurons/pathology , Peptide Fragments/biosynthesis , Receptors, LDL/genetics , Up-Regulation/physiology
20.
J Neurosci Res ; 85(13): 2856-62, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17638300

ABSTRACT

The tissue- and cell-specific expression of three neutral glycosphingolipids, gangliotetraosylceramide (GA1), gangliopentaosylceramide (GalNAc-GA1), and the novel 3-O-acetyl-sphingosine-series glycolipid (FMC-5), were examined with monospecific polyclonal antibodies. Immunohistochemical studies of rodent brain cross-sections indicated that both GA1 and FMC-5 antibodies stained myelin. In contrast, GalNAc-GA1 antibody distinctly stained neurons in cerebral cortex, but only partially delineated Purkinje cells and other neurons in cerebellum. Preliminary studies of mixed glial cultures suggested the following: 1) both FMC-5 and GA1 antibodies stained oligodendrocytes and oligo progenitors, and 2) GalNAc-GA1 antibody did not stain any cells in the culture. Because the GalNAc-GA1 was associated with neurons, we examined the immunoreactivity of GalNAc-GA1 antibody in primary neuronal cultures. Further studies using primary cultures of rat brain oligodendrocytes, and dissociated cerebellar neuronal cultures indicated that both GA1 and FMC-5 are specifically expressed by oligodendrocytes, whereas GalNAc-GA1 is primarily localized in interneurons and to some extent in Purkinje neurons.


Subject(s)
Brain/cytology , Brain/metabolism , Glycosphingolipids/metabolism , Myelin Sheath/enzymology , Oligodendroglia/enzymology , Sphingosine/metabolism , Animals , Cells, Cultured , Enzyme-Linked Immunosorbent Assay/methods , Female , Gerbillinae , Male , Microscopy, Electron, Transmission/methods , Neuroglia/metabolism , Neuroglia/ultrastructure , Neurons/metabolism , Neurons/ultrastructure , Rabbits
SELECTION OF CITATIONS
SEARCH DETAIL
...