Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Aging Cell ; 20(8): e13433, 2021 08.
Article in English | MEDLINE | ID: mdl-34261192

ABSTRACT

Recent studies indicate a crucial role for neuronal glycogen storage and degradation in memory formation. We have previously identified alpha-amylase (α-amylase), a glycogen degradation enzyme, located within synaptic-like structures in CA1 pyramidal neurons and shown that individuals with a high copy number variation of α-amylase perform better on the episodic memory test. We reported that neuronal α-amylase was absent in patients with Alzheimer's disease (AD) and that this loss corresponded to increased AD pathology. In the current study, we verified these findings in a larger patient cohort and determined a similar reduction in α-amylase immunoreactivity in the molecular layer of hippocampus in AD patients. Next, we demonstrated reduced α-amylase concentrations in oligomer amyloid beta 42 (Aß42 ) stimulated SH-SY5Y cells and neurons derived from human-induced pluripotent stem cells (hiPSC) with PSEN1 mutation. Reduction of α-amylase production and activity, induced by siRNA and α-amylase inhibitor Tendamistat, respectively, was further shown to enhance glycogen load in SH-SY5Y cells. Both oligomer Aß42  stimulated SH-SY5Y cells and hiPSC neurons with PSEN1 mutation showed, however, reduced load of glycogen. Finally, we demonstrate the presence of α-amylase within synapses of isolated primary neurons and show that inhibition of α-amylase activity with Tendamistat alters neuronal activity measured by calcium imaging. In view of these findings, we hypothesize that α-amylase has a glycogen degrading function within synapses, potentially important in memory formation. Hence, a loss of α-amylase, which can be induced by Aß pathology, may in part underlie the disrupted memory formation seen in AD patients.


Subject(s)
Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Glycogenolysis/genetics , alpha-Amylases/metabolism , Animals , Humans , Male , Mice
2.
Alzheimers Res Ther ; 12(1): 158, 2020 11 21.
Article in English | MEDLINE | ID: mdl-33220711

ABSTRACT

BACKGROUND: Previous studies have shown that copy number variation (CNV) in the alpha (α)-amylase gene (AMY1A) is associated with body mass index, insulin resistance, and blood glucose levels, factors also shown to increase the risk of Alzheimer's dementia (AD). We have previously demonstrated the presence of α-amylase in healthy neuronal dendritic spines and a reduction of the same in AD patients. In the current study, we investigate the relationship between AMY1A copy number and AD, memory performance, and brain α-amylase activity. METHODS AND MATERIALS: The association between AMY1A copy number and development of AD was analyzed in 5422 individuals (mean age at baseline 57.5 ± 5.9, females 58.2%) from the Malmö diet and cancer study genotyped for AMY1A copy number, whereof 247 where diagnosed with AD during a mean follow-up of 20 years. Associations between AMY1A copy number and cognitive performance where analyzed in 791 individuals (mean age at baseline 54.7 ± 6.3, females 63%), who performed Montreal Cognitive Assessment (MoCA) test. Correlation analysis between α-amylase activity or α-amylase gene expression and AMY1A copy number in post-mortem hippocampal tissue from on demented controls (n = 8) and AD patients (n = 10) was also performed. RESULTS: Individuals with very high ( ≥10) AMY1A copy number had a significantly lower hazard ratio of AD (HR = 0.62, 95% CI 0.41-0.94) and performed significantly better on MoCA delayed word recall test, compared to the reference group with AMY1A copy number 6. A trend to lower hazard ratio of AD was also found among individuals with low AMY1A copy number (1-5) (HR = 0.74, 95% CI 0.53-1.02). A tendency towards a positive correlation between brain α-amylase activity and AMY1A copy number was found, and females showed higher brain α-amylase activity compared to males. CONCLUSION: Our study suggests that the degree of α-amylase activity in the brain is affected by AMY1A copy number and gender, in addition to AD pathology. The study further suggests that very high AMY1A copy number is associated with a decreased hazard ratio of AD and we speculate that this effect is mediated via a beneficial impact of AMY1A copy number on episodic memory performance.


Subject(s)
Alzheimer Disease , Salivary alpha-Amylases , Alzheimer Disease/genetics , Amylases/genetics , Cognition , DNA Copy Number Variations/genetics , Female , Humans , Male , Salivary alpha-Amylases/genetics
3.
J Alzheimers Dis ; 73(3): 1201-1209, 2020.
Article in English | MEDLINE | ID: mdl-31884473

ABSTRACT

BACKGROUND: Previous studies have used immunohistology to demonstrate Alzheimer's disease (AD) characteristic accumulation of amyloid-ß (Aß) in the retina of AD patients, a finding indicating retina examination as a potential diagnostic tool for AD pathology. OBJECTIVE: To further explore this idea by investigating whether levels of Aß42 and Aß40 in retina are associated with corresponding levels in hippocampus, neuropathological assessments, apolipoprotein E (APOE) genotype, and levels of islet amyloid polypeptide (IAPP). METHODS: Levels of high molecular weight (HMW) Aß42, Aß40, and IAPP in ultra-centrifuged homogenates of retina and hippocampus from patients with AD, multiple sclerosis, AD with Lewy bodies, and non-demented controls were analyzed using Mesoscale Discovery electrochemiluminescence technology employing immunoassay and enzyme-linked immunosorbent assay. RESULTS: Higher levels of retinal and hippocampal Aß42-HMW, Aß40-HMW, and IAPP-HMW were found in individuals with high neuropathological scores of Aß plaques and in individuals carrying the APOEɛ4 allele. The retinal levels of Aß42-HMW and Aß40-HMW correlated with corresponding levels in hippocampus as well as with neurofibrillary tangles (NFT) and Aß scores. Retinal IAPP-HMW correlated with retinal levels of Aß42-HMW and with NFT and Aß scores. CONCLUSION: These results show that different isoforms of Aß can be detected in the human retina and moreover support the growing number of studies indicating that AD-related pathological changes occurring in the brain could be reflected in the retina.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Hippocampus/metabolism , Islet Amyloid Polypeptide/metabolism , Retina/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Apolipoproteins E/genetics , Female , Hippocampus/pathology , Humans , Male , Middle Aged , Neurofibrillary Tangles/genetics , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Retina/pathology
4.
PLoS One ; 14(6): e0218561, 2019.
Article in English | MEDLINE | ID: mdl-31206565

ABSTRACT

The biologically active pancreatic hormone peptide islet amyloid polypeptide (IAPP) regulates brain functions such as appetite and cognition. It also plays a role in clearance of amyloid beta (Aß), a peptide implicated in the dementia disorder Alzheimer's disease (AD). If IAPP becomes modified, it loses its biological activity and starts to aggregate. Such aggregations have been found in the AD brain and decreased plasma levels of the unmodified IAPP (uIAPP) have been shown in the same patients. In the current study, we analyze levels of uIAPP and total IAPP (unmodified and modified) in cerebrospinal fluid (CSF) to investigate its potential as a biomarker for AD. We found no differences in neither CSF nor plasma levels of uIAPP or total IAPP in AD patients compared to cognitive healthy individuals (NC). The levels of uIAPP in CSF of NC were positively correlated with uIAPP in plasma, Q-albumin and albumin levels in CSF, but negatively correlated with CSF levels of t-tau and p-tau. These findings were not seen in AD patients. Levels of total CSF IAPP correlated positively with total Q-albumin and albumin levels in CSF in both AD and NC. In addition, total plasma IAPP correlated positively with CSF t-tau and p-tau in NC and negatively with CSF Aß42 in AD patients. To conclude, our studies did not find evidence supporting the use of CSF IAPP as an AD biomarker. However, our findings, indicating a compromised translocation of uIAPP in and out of the brain in AD patients as well as the correlations between total plasma IAPP and AD biomarkers, encourage further research on the role for IAPP in AD.


Subject(s)
Alzheimer Disease/blood , Alzheimer Disease/cerebrospinal fluid , Biomarkers , Islet Amyloid Polypeptide/blood , Islet Amyloid Polypeptide/cerebrospinal fluid , Aged , Aged, 80 and over , Alzheimer Disease/diagnosis , Amyloid beta-Peptides/cerebrospinal fluid , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Middle Aged , Prognosis , tau Proteins/cerebrospinal fluid
5.
J Alzheimers Dis ; 68(1): 205-217, 2019.
Article in English | MEDLINE | ID: mdl-30775997

ABSTRACT

BACKGROUND: Astrocytes produce and store the energy reserve glycogen. However, abnormal large glycogen units accumulate if the production or degradation of glycogen is disturbed, a finding often seen in patients with Alzheimer's disease (AD). We have shown increased activity of glycogen degrading α-amylase in AD patients and α-amylase positive glial cells adjacent to AD characteristic amyloid-ß (Aß) plaques. OBJECTIVES: Investigate the role of α-amylase in astrocytic glycogenolysis in presence of Aß. METHODS: Presence of α-amylase and large glycogen units in postmortem entorhinal cortex from AD patients and non-demented controls were analyzed by immunohistological stainings. Impact of different Aß42 aggregation forms on enzymatic activity (α-amylase, pyruvate kinase, and lactate dehydrogenase), lactate secretion, and accumulation of large glycogen units in cultured astrocytes were analyzed by activity assays, ELISA, and immunocytochemistry, respectively. RESULTS: AD patients showed increased number of α-amylase positive glial cells. The glial cells co-expressed the astrocytic marker glial fibrillary acidic protein, displayed hypertrophic features, and increased amount of large glycogen units. We further found increased load of large glycogen units, α-amylase immunoreactivity and α-amylase activity in cultured astrocytes stimulated with fibril Aß42, with increased pyruvate kinase activity, but unaltered lactate release as downstream events. The fibril Aß42-induced α-amylase activity was attenuated by ß-adrenergic receptor antagonist propranolol. DISCUSSION: We hypothesize that astrocytes respond to fibril Aß42 in Aß plaques by increasing their α-amylase production to either liberate energy or regulate functions needed in reactive processes. These findings indicate α-amylase as an important actor involved in AD associated neuroinflammation.


Subject(s)
Alzheimer Disease/enzymology , Amyloid beta-Peptides/toxicity , Astrocytes/enzymology , Entorhinal Cortex/enzymology , Glycogenolysis/physiology , Peptide Fragments/toxicity , alpha-Amylases/analysis , Alzheimer Disease/pathology , Astrocytes/drug effects , Astrocytes/pathology , Cells, Cultured , Cohort Studies , Entorhinal Cortex/pathology , Glycogenolysis/drug effects , Humans , alpha-Amylases/metabolism
6.
Neurobiol Aging ; 69: 94-101, 2018 09.
Article in English | MEDLINE | ID: mdl-29864717

ABSTRACT

Islet amyloid polypeptide (IAPP) forms toxic aggregates in the brain of patients with Alzheimer's disease (AD). Whether IAPP also affects the retina in these patients is still unknown. Levels of IAPP in soluble and insoluble homogenate fractions of retina and hippocampus from AD patients and nondemented controls were analyzed using ELISA. Number of pericytes and vessel length were determined by analysis of immunostained retina and hippocampus. Insoluble retinal fractions of AD patients contained lower levels of unmodified IAPP, whereas soluble retinal fractions contained increased levels of the same. Total IAPP levels and pericyte numbers in retina mirrored corresponding variables in the hippocampus. Moreover, levels of total unmodified IAPP correlated negatively with the vessel length both in retina and hippocampus across the group and positively with pericyte numbers in retina in AD patients. Our studies indicate that changes in brain IAPP are reflected by corresponding levels in the retina. Our results also suggest modification of IAPP as an important event implicated in vascular changes associated with AD.


Subject(s)
Alzheimer Disease/metabolism , Hippocampus/blood supply , Hippocampus/metabolism , Islet Amyloid Polypeptide/metabolism , Retina/metabolism , Retinal Vessels/pathology , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Female , Hippocampus/pathology , Humans , Male , Middle Aged , Pericytes/pathology , Retina/pathology
7.
Aging Cell ; 17(3): e12728, 2018 06.
Article in English | MEDLINE | ID: mdl-29453790

ABSTRACT

The population of brain pericytes, a cell type important for vessel stability and blood brain barrier function, has recently been shown altered in patients with Alzheimer's disease (AD). The underlying reason for this alteration is not fully understood, but progressive accumulation of the AD characteristic peptide amyloid-beta (Aß) has been suggested as a potential culprit. In the current study, we show reduced number of hippocampal NG2+ pericytes and an association between NG2+ pericyte numbers and Aß1-40 levels in AD patients. We further demonstrate, using in vitro studies, an aggregation-dependent impact of Aß1-40 on human NG2+ pericytes. Fibril-EP Aß1-40 exposure reduced pericyte viability and proliferation and increased caspase 3/7 activity. Monomer Aß1-40 had quite the opposite effect: increased pericyte viability and proliferation and reduced caspase 3/7 activity. Oligomer-EP Aß1-40 had no impact on either of the cellular events. Our findings add to the growing number of studies suggesting a significant impact on pericytes in the brains of AD patients and suggest different aggregation forms of Aß1-40 as potential key regulators of the brain pericyte population size.


Subject(s)
Amyloid beta-Peptides/metabolism , Antigens/metabolism , Pericytes/metabolism , Proteoglycans/metabolism , Aged , Aged, 80 and over , Cell Culture Techniques , Female , Humans , Male , Middle Aged
8.
Brain Pathol ; 28(6): 920-932, 2018 11.
Article in English | MEDLINE | ID: mdl-29485701

ABSTRACT

Reduced glucose metabolism and formation of polyglucosan bodies (PGB) are, beside amyloid beta plaques and neurofibrillary tangles, well-known pathological findings associated with Alzheimer's disease (AD). Since both glucose availability and PGB are regulated by enzymatic degradation of glycogen, we hypothesize that dysfunctional glycogen degradation is a critical event in AD progression. We therefore investigated whether alpha (α)-amylase, an enzyme known to efficiently degrade polysaccharides in the gastrointestinal tract, is expressed in the hippocampal CA1/subiculum and if the expression is altered in AD patients. Using immunohistochemical staining techniques, we show the presence of the α-amylase isotypes AMY1A and AMY2A in neuronal dendritic spines, pericytes and astrocytes. Moreover, AD patients showed reduced gene expression of α-amylase, but conversely increased protein levels of α-amylase as well as increased activity of the enzyme compared with non-demented controls. Lastly, we observed increased, albeit not significant, load of periodic acid-Schiff positive PGB in the brain of AD patients, which correlated with increased α-amylase activity. These findings show that α-amylase is expressed and active in the human brain, and suggest the enzyme to be affected, alternatively play a role, in the neurodegenerative Alzheimer's disease pathology.


Subject(s)
Alzheimer Disease/enzymology , CA1 Region, Hippocampal/enzymology , Energy Metabolism , Pancreatic alpha-Amylases/metabolism , Salivary alpha-Amylases/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Astrocytes/enzymology , Cohort Studies , Dendritic Spines/enzymology , Female , Gene Expression , Glucans/biosynthesis , Glucose/metabolism , Glycogen/metabolism , Humans , Male , Middle Aged , Neurofibrillary Tangles/pathology , Pancreatic alpha-Amylases/genetics , Pericytes/enzymology , Plaque, Amyloid/pathology , Salivary alpha-Amylases/genetics
9.
J Cereb Blood Flow Metab ; 37(4): 1470-1482, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27354094

ABSTRACT

Amylin, a pancreatic ß-cell-derived peptide hormone, forms inclusions in brain microvessels of patients with dementia who have been diagnosed with type 2 diabetes and Alzheimer's disease. The cellular localization of these inclusions and the consequences thereof are not yet known. Using immunohistochemical staining of hippocampus and parahippocampal cortex from patients with Alzheimer's disease and non-demented controls, we show that amylin cell inclusions are found in pericytes. The number of amylin cell inclusions did not differ between patients with Alzheimer's disease and controls, but amylin-containing pericytes displayed nuclear changes associated with cell death and reduced expression of the pericyte marker neuron-glial antigen 2. The impact of amylin on pericyte viability was further demonstrated in in vitro studies, which showed that pericyte death increased in presence of fibril- and oligomer amylin. Furthermore, oligomer amylin increased caspase 3/7 activity, reduced lysate neuron-glial antigen 2 levels and impaired autophagy. Our findings contribute to increased understanding of how aggregated amylin affects brain vasculature and highlight amylin as a potential factor involved in microvascular pathology in dementia progression.


Subject(s)
Alzheimer Disease/metabolism , Antigens/biosynthesis , Hippocampus/metabolism , Islet Amyloid Polypeptide/metabolism , Pericytes/metabolism , Proteoglycans/biosynthesis , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Autophagy/drug effects , Case-Control Studies , Cell Culture Techniques , Cell Survival/drug effects , Cells, Cultured , Female , Hippocampus/blood supply , Hippocampus/pathology , Humans , Immunohistochemistry , Islet Amyloid Polypeptide/toxicity , Male , Microvessels/metabolism , Microvessels/pathology , Middle Aged , Pericytes/pathology
10.
J Biol Chem ; 291(41): 21644-21655, 2016 Oct 07.
Article in English | MEDLINE | ID: mdl-27566545

ABSTRACT

C4BP (C4b-binding protein) is a polymer of seven identical α chains and one unique ß chain synthesized in liver and pancreas. We showed previously that C4BP enhances islet amyloid polypeptide (IAPP) fibril formation in vitro Now we report that polymeric C4BP strongly inhibited lysis of human erythrocytes incubated with monomeric IAPP, whereas no lysis was observed after incubation with preformed IAPP fibrils. In contrast, incubation with the monomeric α-chain of C4BP was less effective. These data indicate that polymeric C4BP with multiple binding sites for IAPP neutralizes lytic activity of IAPP. Furthermore, addition of monomeric IAPP to a rat insulinoma cell line (INS-1) resulted in decreased cell viability, which was restored in the presence of physiological concentrations of C4BP. Treatment of INS-1 cells and primary rat islets with IAPP also diminished their ability to secrete insulin upon stimulation with glucose, which was reversed in the presence of C4BP. Further, C4BP was internalized together with IAPP into INS-1 cells. Pathway analyses of mRNA expression microarray data indicated that cells exposed to C4BP and IAPP in comparison with IAPP alone increased expression of genes involved in cholesterol synthesis. Depletion of cholesterol through methyl-ß-cyclodextrin or cholesterol oxidase abolished the protective effect of C4BP on IAPP cytotoxicity of INS-1 cells. Also, inhibition of phosphoinositide 3-kinase but not NF-κB had a similar effect. Taken together, C4BP protects ß-cells from IAPP cytotoxicity by modulating IAPP fibril formation extracellularly and also, after uptake by the cells, by enhancing cholesterol synthesis.


Subject(s)
Cholesterol/biosynthesis , Complement C4b-Binding Protein/metabolism , Gene Expression Regulation/physiology , Insulin-Secreting Cells/metabolism , Islet Amyloid Polypeptide/biosynthesis , Animals , Cell Line, Tumor , Cholesterol Oxidase/metabolism , Humans , Male , Phosphatidylinositol 3-Kinases/metabolism , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL