Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
Biochem Biophys Res Commun ; 411(3): 620-6, 2011 Aug 05.
Article in English | MEDLINE | ID: mdl-21771586

ABSTRACT

Vascular endothelial growth factor (VEGF) was investigated in the present study to see whether it could provide a therapeutic opportunity for the treatment of Alzheimer's disease (AD). PDGF-hAPP(V717I) transgenic mice were treated with VEGF or PBS by intraperitoneal injection for three consecutive days. The results showed that VEGF ameliorated the memory impairment of mice, accompanied by CD34(+) cells increasing in peripheral blood, vWF(+) vessels increasing in hippocampus, and CD34(+)/VEGFR2(+), vWF(+)/VEGFR2(+) and BrdU(+)/vWF(+) cells expressing in hippocampus. Furthermore, the level of choline acetyltransferase (ChAT) was considerably enhanced and Aß deposition was decreased in the brains of mice upon VEGF treatment. These observations suggest that VEGF should be pursued as a novel therapeutic agent for treatment of AD.


Subject(s)
Alzheimer Disease/complications , Brain/blood supply , Memory Disorders/drug therapy , Neovascularization, Physiologic/drug effects , Vascular Endothelial Growth Factor A/therapeutic use , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Disease Models, Animal , Memory Disorders/etiology , Mice , Mice, Transgenic , Platelet-Derived Growth Factor/genetics
2.
Sci Rep ; 7(1): 3446, 2017 06 13.
Article in English | MEDLINE | ID: mdl-28611418

ABSTRACT

Amyloid ß-protein (Aß) is believed to contribute to the development of Alzheimer's disease (AD). Here we showed that Aß25-35 rapidly caused activation of autophagy, subsequently leading to reduction of autophagy associated with cellular apoptosis. Further investigation revealed that the accumulation of ß-arrestin 1 (ARRB1) caused by Aß25-35 contributed to the induction of autophagic flux. The depletion of ARRB1 led to decreases in the expression of LC3B, Atg7, and Beclin-1, which are essential for the initiation of autophagy. ARRB1 depletion also reduced downstream ERK activity and promoted Aß25-35-induced cell death. As with ARRB1, transient upregulation of ARRB2 by Aß25-35 was observed after short treatment durations, whereas genetic reduction of ARRB2 caused a marked increase in the expression of the α7nAch receptor at the cell surface, which resulted in partial reversal of Aß25-35-induced cell death. Although expression of both ARRB1 and ARRB2 was reduced in serum from patients with AD, the levels of ARRB1 were much lower than those of ARRB2 in AD. Thus, our findings indicate that ARRB1/2 play different roles in Aß25-35 cytotoxicity, which may provide additional support for exploring the underlying molecular mechanism of AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/toxicity , Autophagy , Peptide Fragments/toxicity , beta-Arrestin 1/metabolism , beta-Arrestin 2/metabolism , Cell Death , Cell Line, Tumor , Humans , Neurons/drug effects , Neurons/metabolism , alpha7 Nicotinic Acetylcholine Receptor/genetics , alpha7 Nicotinic Acetylcholine Receptor/metabolism , beta-Arrestin 1/genetics , beta-Arrestin 2/genetics
3.
Neuroreport ; 28(4): 222-228, 2017 Mar 01.
Article in English | MEDLINE | ID: mdl-28118288

ABSTRACT

Increasing research suggests that mitochondrial defects play a major role in Alzheimer's disease (AD) pathogenesis. We aimed to better understand changes in mitochondria with the development and progression of AD. We compared APPsw/PS1dE9 transgenic mice at 3, 6, 9, and 12 months old as an animal model of AD and age-matched C57BL/6 mice as controls. The learning ability and spatial memory ability of APPsw/PS1dE9 mice showed significant differences compared with controls until 9 and 12 months. Mitochondrial morphology was altered in hippocampus tissue of APPsw/PS1dE9 mice beginning from the third month. 'Medullary corpuscle', which is formed by the accumulation of a large amount of degenerative and fragmented mitochondria in neuropils, may be the characteristic change observed on electron microscopy at a late stage of AD. Moreover, levels of mitochondrial fusion proteins (optic atrophy 1 and mitofusin 2) and fission proteins (dynamin-related protein 1 and fission 1) were altered in transgenic mice compared with controls with progression of AD. We found increased levels of fission and fusion proteins in APP/PS1 mice at 3 months, indicating that the presence of abnormal mitochondrial dynamics may be events in early AD progression. Changes in mitochondrial preceded the onset of memory decline as measured by the modified Morris water maze test. Abnormal mitochondrial dynamics could be a marker for early diagnosis of AD and monitoring disease progression. Further research is needed to study the signaling pathways that govern mitochondrial fission/fusion in AD.


Subject(s)
Aging , Alzheimer Disease/physiopathology , Mitochondrial Dynamics , Alzheimer Disease/metabolism , Animals , Disease Models, Animal , Hippocampus/metabolism , Hippocampus/pathology , Male , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/ultrastructure , Mitochondrial Proteins/metabolism , Spatial Learning , Spatial Memory
4.
Clin Exp Med ; 16(1): 89-98, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26188488

ABSTRACT

Alzheimer's disease (AD) is the leading cause of dementia in the elderly and is characterized by amyloid plaques, neurofibrillary tangles, and neuronal loss. Cumulative evidence supports that neuroinflammation is an important factor for the pathogenesis of AD and contributes to amyloid beta (Aß) generation. However, there has been no effective treatment for AD. Wharton's Jelly-derived mesenchymal stem cells (WJ-MSCs) have a potential therapeutic effect in the treatment for neurological diseases. In the present study, we evaluated the therapeutic effect of WJ-MSC transplantation on the neuropathology and memory deficits in amyloid precursor protein (APP) and presenilin-1 (PS1) double-transgenic mice and discussed the mechanism. WJ-MSCs were intravenously transplanted into the APP/PS1 mice. Four weeks after treatment, WJ-MSCs significantly improved the spatial learning and alleviated the memory decline in the APP/PS1 mice. Aß deposition and soluble Aß levels were significantly reduced after WJ-MSC treatment. Furthermore, WJ-MSCs significantly increased the expression of the anti-inflammatory cytokine, IL-10. Meanwhile, pro-inflammatory microglial activation and the expressions of pro-inflammatory cytokines, IL-1ß and TNFα, were significantly down-regulated by WJ-MSC treatment. Thus, our findings suggest that WJ-MSCs might produce beneficial effects on the prevention and treatment for AD through modulation of neuroinflammation.


Subject(s)
Amyloid beta-Protein Precursor/genetics , Memory Disorders/therapy , Presenilin-1/genetics , Stem Cell Transplantation/methods , Wharton Jelly/cytology , Administration, Intravenous , Amyloid beta-Peptides/metabolism , Animals , Cytokines/metabolism , Disease Models, Animal , Gene Expression Regulation , Humans , Memory Disorders/genetics , Memory Disorders/immunology , Mice , Mice, Transgenic , Spatial Learning/physiology
SELECTION OF CITATIONS
SEARCH DETAIL