Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
J Neuropathol Exp Neurol ; 71(4): 289-97, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22437340

ABSTRACT

Defects in synaptic development and plasticity may lead to autism. Brain-derived neurotrophic factor (BDNF) plays a critical role in synaptogenesis and synaptic plasticity. BDNF is synthesized as a precursor, pro-BDNF, which can be processed into either a truncated form or into mature BDNF. Previous studies reported increased BDNF-immunoreactive protein in autism, but the mechanism of this increase has not been investigated. We examined BDNF mRNA by real-time reverse transcription-polymerase chain reaction and BDNF protein by Western blotting and enzyme-linked immunosorbent assay in postmortem fusiform gyrus tissue from 11 patients with autism and 14 controls. BDNF mRNA levels were not different in the autism versus control samples, but total BDNF-like immunoreactive protein, measured by enzyme-linked immunosorbent assay, was greater in autism than in controls. Western blotting revealed greater pro-BDNF and less truncated BDNF in autism compared with controls. These data demonstrate that increased levels of BDNF-immunoreactive protein in autism are not transcriptionally driven. Increased pro-BDNF and reduced truncated BDNF are consistent with defective processing of pro-BDNF to its truncated form. Distortion of the balance among the 3 BDNF isoforms, each of which may exhibit different biological activities, could lead to changes in connectivity and synaptic plasticity and, hence, behavior. Thus, imbalance in proteolytic isoforms is a possible new mechanism for altered synaptic plasticity leading to autism.


Subject(s)
Autistic Disorder/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Protein Precursors/metabolism , Adult , Autistic Disorder/diagnosis , Autistic Disorder/genetics , Brain-Derived Neurotrophic Factor/genetics , Child , Child, Preschool , Female , Humans , Male , Middle Aged , Neuronal Plasticity/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Precursors/genetics , Proteolysis , RNA, Messenger/metabolism , Synapses/genetics , Young Adult
2.
J Neurosci ; 29(29): 9321-9, 2009 Jul 22.
Article in English | MEDLINE | ID: mdl-19625522

ABSTRACT

Downregulation of brain-derived neurotrophic factor (BDNF) in the cortex occurs early in the progression of Alzheimer's disease (AD). Since BDNF plays a critical role in neuronal survival, synaptic plasticity, and memory, BDNF reduction may contribute to synaptic and cellular loss and memory deficits characteristic of AD. In vitro evidence suggests that amyloid-beta (A beta) contributes to BDNF downregulation in AD, but the specific A beta aggregation state responsible for this downregulation in vivo is unknown. In the present study, we examined cortical levels of BDNF mRNA in three different transgenic AD mouse models harboring mutations in APP resulting in A beta overproduction, and in a genetic mouse model of Down syndrome. Two of the three A beta transgenic strains (APP(NLh) and TgCRND8) exhibited significantly decreased cortical BDNF mRNA levels compared with wild-type mice, whereas neither the other strain (APP(swe)/PS-1) nor the Down syndrome mouse model (Ts65Dn) was affected. Only APP(NLh) and TgCRND8 mice expressed high A beta(42)/A beta(40) ratios and larger SDS-stable A beta oligomers (approximately 115 kDa). TgCRND8 mice exhibited downregulation of BDNF transcripts III and IV; transcript IV is also downregulated in AD. Furthermore, in all transgenic mouse strains, there was a correlation between levels of large oligomers, A beta(42)/A beta(40), and severity of BDNF decrease. These data show that the amount and species of A beta vary among transgenic mouse models of AD and are negatively correlated with BDNF levels. These findings also suggest that the effect of A beta on decreased BDNF expression is specific to the aggregation state of A beta and is dependent on large oligomers.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Brain/metabolism , Amyloid beta-Protein Precursor/genetics , Analysis of Variance , Animals , Blotting, Western , Disease Models, Animal , Down Syndrome/genetics , Down Syndrome/metabolism , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Humans , Mice , Mice, Transgenic , Polymerase Chain Reaction , Presenilin-1/genetics , Protease Nexins , RNA, Messenger/metabolism , Receptors, Cell Surface/genetics
3.
J Neurosci ; 27(10): 2628-35, 2007 Mar 07.
Article in English | MEDLINE | ID: mdl-17344400

ABSTRACT

Alzheimer's disease (AD) is a senile dementia characterized by amyloid plaques, neurofibrillary tangles, and synaptic and cell loss. The "amyloid cascade" hypothesis suggests that amyloid-beta (Abeta), the peptide deposited as amyloid plaques, is the primary insult in AD. However, debate continues over the mechanism of Abeta toxicity and whether fibrillar or oligomeric Abeta is the active species of the peptide that ultimately causes the synaptic loss and dementia associated with AD. Brain-derived neurotrophic factor (BDNF) is required for survival and function of cells compromised in AD. Decreased BDNF causes defects in long-term potentiation and memory and correlates with cognitive decline. We previously demonstrated that BDNF reduction occurs early in the course of AD, suggesting that decreased BDNF may promote neuronal dysfunction in AD. We also demonstrated that three of seven human BDNF transcripts are specifically downregulated in AD. What pathological feature(s) of AD leads to the decreased BDNF is unknown. In this study, we administered both fibrillar and oligomeric conformations of Abeta(1-42) to differentiated SH-SY5Y, a human neuroblastoma cell line, and measured both phosphorylated cAMP response element-binding protein (CREB), a regulator of BDNF transcription, and BDNF total mRNA. We found that oligomeric but not fibrillar preparations of Abeta(1-42) significantly decrease both phosphorylated CREB and total BDNF mRNA. Furthermore, oligomeric Abeta(1-42) decreases BDNF transcripts IV and V in these cells, demonstrating that Abeta(1-42) downregulates the major BDNF transcript decreased in vivo in the AD brain. Thus, oligomeric Abeta(1-42) could compromise neuronal function, causing memory loss and cognitive dysfunction by downregulation of BDNF in AD.


Subject(s)
Amyloid/chemistry , Amyloid/pharmacology , Brain-Derived Neurotrophic Factor/genetics , Neuroblastoma/metabolism , Neuroblastoma/pathology , RNA, Messenger/metabolism , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/pharmacology , Cell Differentiation/drug effects , Cell Line, Tumor , Cyclic AMP Response Element-Binding Protein/metabolism , Down-Regulation , Humans , Molecular Conformation , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Phosphorylation/drug effects , RNA, Messenger/antagonists & inhibitors , Tretinoin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...