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 Alzheimers Dis ; 63(4): 1433-1443, 2018.
Article in English | MEDLINE | ID: mdl-29843236

ABSTRACT

The pathophysiology of Alzheimer's disease (AD) includes signaling defects mediated by the transforming growth factor ß-bone morphogenetic protein-growth and differentiation factor (TGFß-BMP-GDF) family of proteins. In animal models of AD, administration of BMP9/GDF2 improves memory and reduces amyloidosis. The best characterized type I receptor of BMP9 is ALK1. We characterized ALK1 expression in the hippocampus using immunohistochemistry. In the rat, ALK1 immunoreactivity was found in CA pyramidal neurons, most frequently and robustly in the CA2 and CA3 fields. In addition, there were sporadic ALK1-immunoreactive cells in the stratum oriens, mainly in CA1. The ALK1 expression pattern in human hippocampus was similar to that of rat. Pyramidal neurons within the CA2, CA3, and CA4 were strongly ALK1-immunoreactive in hippocampi of cognitively intact subjects with no neurofibrillary tangles. ALK1 signal was found in the axons of alveus and fimbria, and in the neuropil across CA fields. Relatively strongest ALK1 neuropil signal was observed in CA1 where pyramidal neurons were occasionally ALK1-immunoractive. As in the rat, horizontally oriented neurons in the stratum oriens of CA1 were both ALK1- and GAD67-immunoreactive. Analysis of ALK1 immunoreactivity across stages of AD pathology revealed that disease progression was characterized by overall reduction of the ALK1 signal in CA3 in advanced, but not early, stages of AD. These data suggest that the CA3 pyramidal neurons may remain responsive to the ALK1 ligands, e.g., BMP9, during initial stages of AD and that ALK1 may constitute a therapeutic target in early and moderate AD.


Subject(s)
Activin Receptors, Type II/metabolism , Activin Receptors/metabolism , Alzheimer Disease/pathology , CA3 Region, Hippocampal/metabolism , Disease Progression , Aged , Alzheimer Disease/metabolism , Animals , Female , Glutamate Decarboxylase/metabolism , Growth Differentiation Factor 2 , Growth Differentiation Factors/metabolism , Humans , Male , Mice, Inbred C57BL , Middle Aged , Rats , Rats, Wistar
2.
J Alzheimers Dis ; 60(1): 43-56, 2017.
Article in English | MEDLINE | ID: mdl-28777754

ABSTRACT

Genome-wide association studies (GWAS) identified susceptibility loci associated with decreased hippocampal volume, and found hippocampal subfield-specific effects at MSRB3 (methionine sulfoxide reductase-B3). The MSRB3 locus was also linked to increased risk for late onset Alzheimer's disease (AD). In this study, we uncovered novel sites of MsrB3 expression in CA pyramidal layer and arteriolar walls by using automated immunohistochemistry on hippocampal sections from 23 individuals accompanied by neuropathology reports and clinical dementia rating scores. Controls, cognitively intact subjects with no hippocampal neurofibrillary tangles, exhibited MsrB3 signal as distinct but rare puncta in CA1 pyramidal neuronal somata. In CA3, however, MsrB3-immunoreactivity was strongest in the neuropil of the pyramidal layer. These patterns were replicated in rodent hippocampi where ultrastructural and immunohistofluorescence analysis revealed MsrB3 signal associated with synaptic vesicles and colocalized with mossy fiber terminals. In AD subjects, the number of CA1 pyramidal neurons with frequent, rather than rare, MsrB3-immunoreactive somatic puncta increased in comparison to controls. This change in CA1 phenotype correlated with the occurrence of AD pathological hallmarks. Moreover, the intensity of MsrB3 signal in the neuropil of CA3 pyramidal layer correlated with the signal pattern in neurons of CA1 pyramidal layer that was characteristic of cognitively intact individuals. Finally, MsrB3 signal in the arteriolar walls in the hippocampal white matter decreased in AD patients. This characterization of GWAS-implicated MSRB3 protein expression in human hippocampus suggests that patterns of neuronal and vascular MsrB3 protein expression reflect or underlie pathology associated with AD.


Subject(s)
Alzheimer Disease/pathology , Hippocampus/metabolism , Hippocampus/pathology , Methionine Sulfoxide Reductases/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Animals , Choroid Plexus/metabolism , Choroid Plexus/pathology , Choroid Plexus/ultrastructure , Ependyma/metabolism , Ependyma/pathology , Ependyma/ultrastructure , Female , Gene Expression Regulation/physiology , Genome-Wide Association Study , Hippocampus/ultrastructure , Humans , Male , Methionine Sulfoxide Reductases/ultrastructure , Mice , Mice, Inbred C57BL , Microscopy, Electron , Middle Aged , Pyramidal Cells/metabolism , Pyramidal Cells/pathology , Pyramidal Cells/ultrastructure , Rats , Rats, Wistar , Vesicle-Associated Membrane Protein 2/metabolism , Vesicular Glutamate Transport Protein 1/metabolism
3.
J Neuropathol Exp Neurol ; 75(8): 779-790, 2016 08.
Article in English | MEDLINE | ID: mdl-27346750

ABSTRACT

Genome-wide association studies have established BIN1 (Bridging Integrator 1) as the most significant late-onset Alzheimer disease (AD) susceptibility locus after APOE We analyzed BIN1 protein expression using automated immunohistochemistry on the hippocampal CA1 region in 19 patients with either no, mild, or moderate-to-marked AD pathology, who had been assessed by Clinical Dementia Rating and CERAD scores. We also examined the amygdala, prefrontal, temporal, and occipital regions in a subset of these patients. In non-demented controls without AD pathology, BIN1 protein was expressed in white matter, glia, particularly oligodendrocytes, and in the neuropil in which the BIN1 signal decorated axons. With increasing severity of AD, BIN1 in the CA1 region showed: 1) sustained expression in glial cells, 2) decreased areas of neuropil expression, and 3) increased cytoplasmic neuronal expression that did not correlate with neurofibrillary tangle load. In patients with AD, both the prefrontal cortex and CA1 showed a decrease in BIN1-immunoreactive (BIN1-ir) neuropil areas and increases in numbers of BIN1-ir neurons. The numbers of CA1 BIN1-ir pyramidal neurons correlated with hippocampal CERAD neuritic plaque scores; BIN1 neuropil signal was absent in neuritic plaques. Our data provide novel insight into the relationship between BIN1 protein expression and the progression of AD-associated pathology and its diagnostic hallmarks.


Subject(s)
Adaptor Proteins, Signal Transducing/biosynthesis , Alzheimer Disease/metabolism , Cerebral Cortex/metabolism , Disease Progression , Nuclear Proteins/biosynthesis , Tumor Suppressor Proteins/biosynthesis , Adaptor Proteins, Signal Transducing/genetics , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Cerebral Cortex/pathology , Female , Gene Expression , Humans , Intracellular Space/genetics , Intracellular Space/metabolism , Male , Middle Aged , Nuclear Proteins/genetics , Tumor Suppressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...