38 results on '"Veerhuis R"'
Search Results
2. Decoration of Fibrin with Extracellular Chaperones
- Author
-
Talens, Simone, Leebeek, Frank, Veerhuis, R, Rijken, Dick, Talens, Simone, Leebeek, Frank, Veerhuis, R, and Rijken, Dick
- Published
- 2019
3. Amyloid-beta Oligomers Relate to Cognitive Decline in Alzheimer's Disease
- Author
-
Jongbloed, W., Bruggink, K.A., Kester, M.I., Visser, P.J., Scheltens, P., Blankenstein, M.A., Verbeek, M.M., Teunissen, C.E., Veerhuis, R., Jongbloed, W., Bruggink, K.A., Kester, M.I., Visser, P.J., Scheltens, P., Blankenstein, M.A., Verbeek, M.M., Teunissen, C.E., and Veerhuis, R.
- Abstract
Item does not contain fulltext, BACKGROUND: Amyloid-beta (Abeta)-oligomers are neurotoxic isoforms of Abeta and are a potential diagnostic biomarker for Alzheimer's disease (AD). OBJECTIVES: 1) Analyze the potential of Abeta-oligomer concentrations in cerebrospinal fluid (CSF) to diagnose and predict progression to AD in a large clinical study sample. 2) Monitor Abeta-oligomer concentrations over-time, both in early and advanced stages of AD. 3) Examine the relation between Abeta-oligomer levels in CSF and cognitive functioning. METHODS: 24 non-demented, 61 mild cognitive impairment (MCI), and 64 AD patients who underwent lumbar puncture and cognitive testing at baseline and follow-up were selected from the memory clinic based Amsterdam Dementia Cohort. CSF samples were analyzed for standard AD-biomarkers and Abeta-oligomer levels using a validated in-house Abeta-oligomer specific enzyme-linked immunosorbent assay. Abeta-oligomer levels were analyzed as indicators of disease progression (follow-up AD diagnosis) and cognitive decline, respectively. RESULTS: Patient groups did not differ in Abeta-oligomer concentrations at baseline or follow-up. Baseline CSF Abeta-oligomer levels were similar in MCI patients that develop AD as in stable MCI patients. MCI and AD patients showed an annual decrease in Abeta-oligomer levels of 9.4% and 6.8%, respectively. A decrease in Abeta-oligomer levels over time was strongly associated with more severe cognitive decline in AD patients. CONCLUSION: Despite the limited diagnostic potential of Abeta-oligomer levels in CSF to differentiate between patient groups, and between MCI-AD and MCI-stable patients, changes in CSF Abeta-oligomer levels were related to cognitive decline. Therefore, CSF Abeta-oligomers may aid in the selection of patients with a more aggressive disease course.
- Published
- 2015
4. Complement activation in Glioblastoma Multiforme pathophysiology: Evidence from serum levels and presence of complement activation products in tumor tissue
- Author
-
Bouwens van der Vlis, T.A.M., Trouw, L.A. (L.), Veerhuis, R., Dirven, C.M.F. (Clemens), Lamfers, M.L.M. (Martine), Al-Khawaja, H., Bouwens van der Vlis, T.A.M., Trouw, L.A. (L.), Veerhuis, R., Dirven, C.M.F. (Clemens), Lamfers, M.L.M. (Martine), and Al-Khawaja, H.
- Abstract
Inflammation plays a key role in the pathophysiology of Glioblastoma Multiforme (GBM). Here we focus on the contribution of the so far largely ignored complement system.ELISA and immunohistochemistry were combined to assess levels and localization of critical components of the initiation- and effector pathways of the complement cascade in sera and tumor tissue from GBM patients and matched controls.Serum levels of factor-B were decreased in GBM patients whereas C1q levels were increased. C1q and factor-B deposited in the tumor tissue. Deposition of C3 and C5b-9 suggests local complement activation. MBL deficiency, based on serum levels, was significantly less frequent among GBM patients compared to controls (14% vs. 33%). Therefore low levels of MBL may protect against the initiation/progression of GBM.
- Published
- 2015
- Full Text
- View/download PDF
5. Complement activation in Glioblastoma Multiforme pathophysiology: Evidence from serum levels and presence of complement activation products in tumor tissue
- Author
-
Bouwens, TAM, Trouw, LA, Veerhuis, R, Dirven, Clemens, Lamfers, Martine, Al-Khawaja, Hazem, Bouwens, TAM, Trouw, LA, Veerhuis, R, Dirven, Clemens, Lamfers, Martine, and Al-Khawaja, Hazem
- Abstract
Inflammation plays a key role in the pathophysiology of Glioblastoma Multiforme (GBM). Here we focus on the contribution of the so far largely ignored complement system. ELISA and immunohistochemistry were combined to assess levels and localization of critical components of the initiation- and effector pathways of the complement cascade in sera and tumor tissue from GBM patients and matched controls. Serum levels of factor-B were decreased in GBM patients whereas C1q levels were increased. C1q and factor-B deposited in the tumor tissue. Deposition of C3 and C5b-9 suggests local complement activation. MBL deficiency, based on serum levels, was significantly less frequent among GBM patients compared to controls (14% vs. 33%). Therefore low levels of MBL may protect against the initiation/progression of GBM. (C) 2014 The Authors. Published by Elsevier B.V. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/3.0/).
- Published
- 2015
6. Amyloid-beta oligomer detection by ELISA in cerebrospinal fluid and brain tissue
- Author
-
Bruggink, K.A., Jongbloed, W., Biemans, E.A.L.M., Veerhuis, R., Claassen, J.A.H.R., Kuiperij, H.B., Verbeek, M.M., Bruggink, K.A., Jongbloed, W., Biemans, E.A.L.M., Veerhuis, R., Claassen, J.A.H.R., Kuiperij, H.B., and Verbeek, M.M.
- Abstract
Item does not contain fulltext, Amyloid-beta (Abeta) deposits are important pathological hallmarks of Alzheimer's disease (AD). Abeta aggregates into fibrils; however, the intermediate oligomers are believed to be the most neurotoxic species and, therefore, are of great interest as potential biomarkers. Here, we have developed an enzyme-linked immunosorbent assay (ELISA) specific for Abeta oligomers by using the same capture and (labeled) detection antibody. The ELISA predominantly recognizes relatively small oligomers (10-25 kDa) and not monomers. In brain tissue of APP/PS1 transgenic mice, we found that Abeta oligomer levels increase with age. However, for measurements in human samples, pretreatment to remove human anti-mouse antibodies (HAMAs) was required. In HAMA-depleted human hippocampal extracts, the Abeta oligomer concentration was significantly increased in AD compared with nondemented controls. Abeta oligomer levels could also be quantified in pretreated cerebrospinal fluid (CSF) samples; however, no difference was detected between AD and control groups. Our data suggest that levels of small oligomers might not be suitable as biomarkers for AD. In addition, we demonstrate the importance of avoiding HAMA interference in assays to quantify Abeta oligomers in human body fluids.
- Published
- 2013
7. NG2 cells, a new trail for Alzheimer's disease mechanisms?
- Author
-
Nielsen, H.M., Ek, D., Avdic, U., Orbjorn, C., Hansson, O., Huitinga, I., Veerhuis, R., Rozemuller, A.J.M., Brun, A., Minthon, L., Wennstrom, M., Nielsen, H.M., Ek, D., Avdic, U., Orbjorn, C., Hansson, O., Huitinga, I., Veerhuis, R., Rozemuller, A.J.M., Brun, A., Minthon, L., and Wennstrom, M.
- Published
- 2013
8. Costimulatory effects of interferon-gamma and interleukin-1beta or tumor necrosis factor alpha on the synthesis of abetal-40 and abetal-42 by human astrocytes
- Author
-
Blasko, I., Veerhuis, R., Stampfer-Kountchev, M., Saurwein-Teissl, M., Eikelenboom, P., Grubeck-Loebenstein, B., and VU University medical center
- Published
- 2000
9. Small heat shock proteins induce a cerebral inflammatory reaction
- Author
-
Bruinsma, I.B., Jager, M. de, Carrano, A., Versleijen, A.A.M., Veerhuis, R., Boelens, W., Rozemuller, A.J., Waal, R.M. de, Verbeek, M.M., Bruinsma, I.B., Jager, M. de, Carrano, A., Versleijen, A.A.M., Veerhuis, R., Boelens, W., Rozemuller, A.J., Waal, R.M. de, and Verbeek, M.M.
- Abstract
Contains fulltext : 96353.pdf (publisher's version ) (Open Access)
- Published
- 2011
10. Apolipoprotein E protects cultured pericytes and astrocytes from D-Abeta(1-40)-mediated cell death.
- Author
-
Bruinsma, I.B., Wilhelmus, M.M.M., Kox, M., Veerhuis, R., Waal, R.M.W. de, Verbeek, M.M., Bruinsma, I.B., Wilhelmus, M.M.M., Kox, M., Veerhuis, R., Waal, R.M.W. de, and Verbeek, M.M.
- Abstract
Contains fulltext : 88262.pdf (publisher's version ) (Closed access), Cerebral amyloid angiopathy (CAA) is a common pathological finding in Alzheimer's disease and hereditary cerebral hemorrhage with amyloidosis of the Dutch type; in this latter condition it is caused by deposition of mutated amyloid beta protein (Abeta Glu22Gln; D-Abeta(1-40)). Previously, we found a dependence of the Abeta-mediated toxicity and apolipoprotein E (apoE) production by cultured pericytes on apoE genotype. Given their close association with the cerebrovascular wall both astrocytes and pericytes may be involved in CAA development, a process that includes Abeta deposition and clearance and that may be affected by interaction with locally produced apolipoprotein E (apoE). Although astrocytes are regarded as the major source of apolipoprotein E (apoE) in the brain, also pericytes produce apoE. In this study we compared the apoE production capacity, the effects of apoE on D-Abeta(1-40) internalization, D-Abeta(1-40) cell surface accumulation and the vulnerability for D-Abeta(1-40)-induced toxicity of either cell type in order to quantify the relative contributions of astrocytes and pericytes in the various processes that contribute to CAA formation. Strikingly, cultured astrocytes produced only 3-10% of the apoE amounts produced by pericytes. Furthermore, pericytes with the apoE epsilon4 allele produced three times less apoE and were more vulnerable to D-Abeta(1-40) treatment than pericytes without an epsilon4 allele. Such relations were not observed with astrocytes in vitro. Both pericytes and astrocytes, however, were protected from Abeta-induced cytotoxicity by high levels of pericyte-derived apoE, but not recombinant apoE. In addition, pericyte-derived apoE dose-dependently decreased both internalization of Abeta and Abeta accumulation at the cell surface in either cell type. The present data suggest that apoE produced by pericytes, rather than astrocyte-produced apoE, modulates Abeta cytotoxicity and Abeta removal near the vasculature in the brain. Furth
- Published
- 2010
11. Small heat shock proteins associated with cerebral amyloid angiopathy of hereditary cerebral hemorrhage with amyloidosis (Dutch type) induce interleukin-6 secretion.
- Author
-
Wilhelmus, M.M.M., Boelens, W.C., Kox, M., Maat-Schieman, M.L., Veerhuis, R., Waal, R.M.W. de, Verbeek, M.M., Wilhelmus, M.M.M., Boelens, W.C., Kox, M., Maat-Schieman, M.L., Veerhuis, R., Waal, R.M.W. de, and Verbeek, M.M.
- Abstract
Contains fulltext : 81507.pdf (publisher's version ) (Closed access), In hereditary cerebral hemorrhage with amyloidosis of the Dutch type (HCHWA-D), severe cerebral amyloid angiopathy (CAA) is associated with an inflammatory reaction. Small heat shock proteins (sHsps) are molecular chaperones and association of HspB8 with CAA in HCHWA-D has been observed. The aims of this study were to investigate (1) if other sHsps are associated with the pathological lesions in HCHWA-D brains, (2) if the amyloid-beta protein (A beta) increases production of sHsps in cultured cerebral cells and (3) if sHsps are involved in the cerebral inflammatory processes in both Alzheimer's disease (AD) and HCHWA-D. We conclude that Hsp20, HspB8 and HspB2 are present in CAA in HCHWA-D, and that A beta did not affect cellular sHsps expression in cultured human brain pericytes and astrocytes. In addition, we demonstrated that Hsp20, HspB2 and HspB8 induced interleukin-6 production in cultured pericytes and astrocytes, which could be antagonized by dexamethasone, whereas other sHsps and A beta were inactive, suggesting that sHsps may be among the key mediators of the local inflammatory response associated with HCHWA-D and AD lesions.
- Published
- 2009
12. Lipoprotein receptor-related protein-1 mediates amyloid-beta-mediated cell death of cerebrovascular cells.
- Author
-
Wilhelmus, M.M.M., Otte-Holler, I., Triel, J.J. van, Veerhuis, R., Maat-Schieman, M.L., Bu, G., Waal, R.M.W. de, Verbeek, M.M., Wilhelmus, M.M.M., Otte-Holler, I., Triel, J.J. van, Veerhuis, R., Maat-Schieman, M.L., Bu, G., Waal, R.M.W. de, and Verbeek, M.M.
- Abstract
Contains fulltext : 51712.pdf (publisher's version ) (Closed access), Inefficient clearance of A beta, caused by impaired blood-brain barrier crossing into the circulation, seems to be a major cause of A beta accumulation in the brain of late-onset Alzheimer's disease patients and hereditary cerebral hemorrhage with amyloidosis Dutch type. We observed association of receptor for advanced glycation end products, CD36, and low-density lipoprotein receptor (LDLR) with cerebral amyloid angiopathy in both Alzheimer's disease and hereditary cerebral hemorrhage with amyloidosis Dutch type brains and increased low-density lipoprotein receptor-related protein-1 (LRP-1) expression by perivascular cells in cerebral amyloid angiopathy. We investigated if these A beta receptors are involved in A beta internalization and in A beta-mediated cell death of human cerebrovascular cells and astrocytes. Expression of both the LRP-1 and LDLR by human brain pericytes and leptomeningeal smooth muscle cells, but not by astrocytes, increased on incubation with A beta. Receptor-associated protein specifically inhibited A beta-mediated up-regulation of LRP-1, but not of LDLR, and receptor-associated protein also decreased A beta internalization and A beta-mediated cell death. We conclude that especially LRP-1 and, to a minor extent, LDLR are involved in A beta internalization by and A beta-mediated cell death of cerebral perivascular cells. Although perivascular cells may adapt their A beta internalization capacity to the levels of A beta present, saturated LRP-1/LDLR-mediated uptake of A beta results in degeneration of perivascular cells.
- Published
- 2007
13. Small heat shock proteins associated with cerebral amyloid angiopathy of hereditary cerebral hemorrhage with amyloidosis (Dutch type) induce interleukin-6 secretion
- Author
-
Wilhelmus, M.M.M., Boelens, W.C., Kox, M., Maat-Schieman, M.L., Veerhuis, R., Waal, R.M.W. de, Verbeek, M.M., Wilhelmus, M.M.M., Boelens, W.C., Kox, M., Maat-Schieman, M.L., Veerhuis, R., Waal, R.M.W. de, and Verbeek, M.M.
- Abstract
Contains fulltext : 36466.pdf (publisher's version ) (Closed access)
- Published
- 2007
14. Decreased cerebrospinal fluid amyloid beta (1-40) levels in frontotemporal lobar degeneration
- Author
-
Pijnenburg, Y A L, primary, Schoonenboom, S N M, additional, Mehta, P D, additional, Mehta, S P, additional, Mulder, C, additional, Veerhuis, R, additional, Blankenstein, M A, additional, and Scheltens, P, additional
- Published
- 2006
- Full Text
- View/download PDF
15. Distribution of A-betha-associated proteins in cerebrovascular amyloid of Alzheimer's disease
- Author
-
Verbeek, M.M., Otte-Holler, I., Veerhuis, R., Ruiter, D.J., Waal, R.M.W. de, Verbeek, M.M., Otte-Holler, I., Veerhuis, R., Ruiter, D.J., and Waal, R.M.W. de
- Abstract
Item does not contain fulltext
- Published
- 1998
16. <em>In vivo</em> degradation of rat C1q induced by intravenous injection of soluble IgG aggregates.
- Author
-
Veerhuis, R., Van Es, L. A., and Daha, M. R.
- Subjects
- *
IMMUNE complexes , *IMMUNOGLOBULIN G , *IMMUNOGLOBULINS , *LABORATORY rats , *ANTIGENS , *IMMUNOLOGY - Abstract
Immune complexes are able to bind and activate the first component of complement, Cl. Upon activation of C&1macr;, C&1macr;r and C&1macr;s are rapidly inactivated by C&1macr;-In which also forms a complex with these two subcomponents, resulting in their release from C&1macr;-immune aggregate complexes. The fate of C1q after the binding of C1 to immune complexes in vivo is not clear and, therefore the clearance of radiolabelled rat C1q was investigated in normal rats and in rats receiving soluble aggregated human IgG. 25I-labelled rat Clq was cleared with a half-life (T½) of 12.4 hr in normal rats. Injection of AIgG into rats that had previously received 251-C1q accelerated the clearance of 251-C1q, resulting, finally, in a T½ of 53 min. The levels of circulating endogenous C1q were also followed using haemolytic titrations and immunochemical measurements. Directly after injection of AIgG into rats, there was a rapid decrease in C1q haemolytic activity to less than 25% of the initial value after 10 min. The rate of disappearance of C1q antigen, was, however, much slower, the lowest concentration being 30% at 2 hr. C1q haemolytic activity and the C1q antigen level returned to virtually normal values after 24 hr. Plasma samples were taken at different time intervals after the injection of AIgG and subjected to gel filtration on Sephacryl S-400 columns. It was found that, in the 10 mm samples. C1q antigen and C1q haemolytic activity, each with an estimated molecular weight (MW) of 400,000, were detected together. in addition, there was C1q antigen with a MW of less than 69,000 without C1q haemolytic activity. SDS-PAGE analysis of the various serum samples indicated that the low MW C1q antigen had an apparent MW of 25,000. Measurement of uptake of 25I-C1q in various organs indicated that the main site of clearance of 25I-C1q is the liver. [ABSTRACT FROM AUTHOR]
- Published
- 1985
17. In vivo degradation of rat C1q induced by intravenous injection of soluble IgG aggregates
- Author
-
Veerhuis, R, van Es, L A, and Daha, M R
- Subjects
Male ,Time Factors ,Complement Activating Enzymes ,Complement C1q ,chemical and pharmacologic phenomena ,Rats, Inbred Strains ,Antigen-Antibody Complex ,urologic and male genital diseases ,Hemolysis ,Rats ,fluids and secretions ,Liver ,immune system diseases ,Immunoglobulin G ,Chromatography, Gel ,Animals ,Electrophoresis, Polyacrylamide Gel ,skin and connective tissue diseases ,Complement Activation ,Research Article - Abstract
Immune complexes are able to bind and activate the first component of complement, C1. Upon activation of C1, C1r and C1s are rapidly inactivated by C1-In which also forms a complex with these two subcomponents, resulting in their release from C1-immune aggregate complexes. The fate of C1q after the binding C1 to immune complexes in vivo is not clear and, therefore the clearance of radiolabelled rat C1q was investigated in normal rats and in rats receiving soluble aggregated human IgG. 125I-labelled rat C1q was cleared with a half-life (T 1/2) of 12.4 hr in normal rats. Injection of AIgG into rats that had previously received 125I-C1q accelerated the clearance of 125I-C1q, resulting, finally, in a T 1/2 of 53 min. The levels of circulating endogenous C1q were also followed using haemolytic titrations and immunochemical measurements. Directly after injection of AIgG into rats, there was a rapid decrease in C1q haemolytic activity to less than 25% of the initial value after 10 min. The rate of disappearance of C1q antigen, was, however, much slower, the lowest concentration being 30% at 2 hr. C1q haemolytic activity and the C1q antigen level returned to virtually normal values after 24 hr. Plasma samples were taken at different time intervals after the injection of AIgG and subjected to gel filtration on Sephacryl S-400 columns. It was found that, in the 10 min samples, C1q antigen and C1q haemolytic activity, each with an estimated molecular weight (MW) of 400,000, were detected together. In addition, there was C1q antigen with a MW of less than 69,000 without C1q haemolytic activity. SDS-PAGE analysis of the various serum samples indicated that the low MW C1q antigen had an apparent MW of 25,000. Measurement of uptake of 125I-C1q in various organs indicated that the main site of clearance of 125I-C1q is the liver.
- Published
- 1985
18. Maximal COX-2 and ppRb expression in neurons occurs during early Braak stages prior to the maximal activation of astrocytes and microglia in Alzheimer's disease
- Author
-
Arendt Thomas, Veerhuis Robert, van Haastert Elise S, Hoozemans Jeroen JM, Scheper Wiep, Eikelenboom Piet, and Rozemuller Annemieke JM
- Subjects
Alzheimer's disease ,astrocytes ,cell cycle ,cyclooxygenase-2 ,microglia ,retinoblastoma protein ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Neuronal expression of cyclooxygenase-2 (COX-2) and cell cycle proteins is suggested to contribute to neurodegeneration during Alzheimer's disease (AD). The stimulus that induces COX-2 and cell cycle protein expression in AD is still elusive. Activated glia cells are shown to secrete substances that can induce expression of COX-2 and cell cycle proteins in vitro. Using post mortem brain tissue we have investigated whether activation of microglia and astrocytes in AD brain can be correlated with the expression of COX-2 and phosphorylated retinoblastoma protein (ppRb). The highest levels of neuronal COX-2 and ppRb immunoreactivity are observed in the first stages of AD pathology (Braak 0–II, Braak A). No significant difference in COX-2 or ppRb neuronal immunoreactivity is observed between Braak stage 0 and later Braak stages for neurofibrillary changes or amyloid plaques. The mean number of COX-2 or ppRb immunoreactive neurons is significantly decreased in Braak stage C compared to Braak stage A for amyloid deposits. Immunoreactivity for glial markers KP1, CR3/43 and GFAP appears in the later Braak stages and is significantly increased in Braak stage V-VI compared to Braak stage 0 for neurofibrillary changes. In addition, a significant negative correlation is observed between the presence of KP1, CR3/43 and GFAP immunoreactivity and the presence of neuronal immunoreactivity for COX-2 and ppRb. These data show that maximal COX-2 and ppRb immunoreactivity in neurons occurs during early Braak stages prior to the maximal activation of astrocytes and microglia. In contrast to in vitro studies, post mortem data do not support a causal relation between the activation of microglia and astrocytes and the expression of neuronal COX-2 and ppRb in the pathological cascade of AD.
- Published
- 2005
- Full Text
- View/download PDF
19. Impaired Innate Immunity Mechanisms in the Brain of Alzheimer's Disease
- Author
-
Ilaria Carbone, Federico Licastro, Elisa Porcellini, Martina Romagnoli, Robert Veerhuis, Romagnoli M, Porcellini E, Carbone I, Veerhuis R, Licastro F, Clinical chemistry, and Amsterdam Neuroscience - Neurodegeneration
- Subjects
Male ,Apolipoprotein E ,Interferon Regulatory Factor-7 ,Biology ,Hippocampal formation ,Hippocampus ,Article ,Catalysis ,Virus ,Inorganic Chemistry ,lcsh:Chemistry ,Immune system ,Alzheimer Disease ,antiviral genes ,Gene expression ,Humans ,RNA, Messenger ,Physical and Theoretical Chemistry ,Allele ,Molecular Biology ,lcsh:QH301-705.5 ,Alleles ,Spectroscopy ,Aged ,Aged, 80 and over ,Temporal cortex ,Mediator Complex ,Innate immune system ,Organic Chemistry ,Brain ,Interferon-alpha ,General Medicine ,Middle Aged ,brain immunity ,Immunity, Innate ,Computer Science Applications ,lcsh:Biology (General) ,lcsh:QD1-999 ,Immunology ,antiviral gene ,gene expression ,Female ,Interferons ,viral infection ,Alzheimer’s disease - Abstract
Among environmental factors likely associated with Alzheimer&rsquo, s disease (AD), persistent virus infections, and age-related progressive decline of immune competence might play a pivotal role. However, AD antimicrobial brain immune responses are poorly investigated. The present study focused on genes involved in antimicrobial defenses, especially against virus infections, in the AD brain. In particular, mRNA levels of IRF7, MED23, IL28B, and IFN-&alpha, genes were analyzed in hippocampus and temporal cortex brain samples from AD and non-demented controls. All subjects were also genotyped for APOE &epsilon, IRF7, MED23, and IL28B gene polymorphisms. Most AD patients showed decreased mRNA levels of all investigated genes in the hippocampus and temporal cortex. However, a small group of AD patients showed increased hippocampal mRNA expression of MED23, IL28B, and IFN-&alpha, mRNA levels of MED23, IL28B, IFN-&alpha, from the hippocampus and those of MED23 from the temporal cortex were further decreased in APOE &epsilon, 4 allele AD carriers. Moreover, rs6598008 polymorphism of IRF7 was significantly associated with decreased hippocampal expression of IRF7, MED23, IL28B, and IFN-&alpha, These findings suggest that AD brains show impaired innate antimicrobial gene expression profiles, and individual genetic makeup, such as positivity for the APOE &epsilon, 4 and IRF7 A alleles, might affect brain immune efficiency.
- Published
- 2020
20. Neurodegeneration: Biochemical signals from the brain
- Author
-
Jongbloed, W., Blankenstein, M.A., Scheltens, P., Veerhuis, R., Neuroscience Campus Amsterdam - Neurodegeneration, Blankenstein, Marinus Aloysius, Scheltens, Philip, Veerhuis, Robert, Clinical chemistry, Neurology, and NCA - neurodegeneration
- Abstract
promotiedatum: 8-5-2014 � prom-id: 11252
- Published
- 2014
21. Amyloid associated proteins in Alzheimer's Disease
- Author
-
Mulder, S.D., Blankenstein, M.A., Scheltens, P., Veerhuis, R., Neuroscience Campus Amsterdam - Neurodegeneration, Blankenstein, Marinus Aloysius, Scheltens, Philip, Veerhuis, Robert, Clinical chemistry, and NCA - Neurodegeneration
- Published
- 2011
22. Biochemical markers in dementia: from mice to men
- Author
-
Verwey, N.A., Scheltens, Philip, Blankenstein, Marinus Aloysius, Veerhuis, Robert, van der Flier, Wiesje, Neurology, NCA - Neurodegeneration, Scheltens, P., Blankenstein, M.A., Veerhuis, R., van der Flier, W.M., and Neuroscience Campus Amsterdam - Neurodegeneration
- Published
- 2010
23. LRRK2 Kinase Inhibition Attenuates Astrocytic Activation in Response to Amyloid β 1-42 Fibrils.
- Author
-
Filippini A, Salvi V, Dattilo V, Magri C, Castrezzati S, Veerhuis R, Bosisio D, Gennarelli M, and Russo I
- Subjects
- Mice, Humans, Animals, Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 metabolism, Neuroinflammatory Diseases, Brain metabolism, Amyloid beta-Peptides, Alzheimer Disease pathology
- Abstract
Intracerebral accumulation of amyloid-β in the extracellular plaques of Alzheimer's disease (AD) brains represents the main cause of reactive astrogliosis and neuroinflammatory response. Of relevance, leucine-rich repeat kinase 2 (LRRK2), a kinase linked to genetic and sporadic Parkinson's disease (PD), has been identified as a positive mediator of neuroinflammation upon different inflammatory stimuli, however its pathogenicity in AD remains mainly unexplored. In this study, by using pharmacological inhibition of LRRK2 and murine primary astrocytes, we explored whether LRRK2 regulates astrocytic activation in response to amyloid-β
1-42 (Aβ1-42 ). Our results showed that murine primary astrocytes become reactive and recruit serine 935 phosphorylated LRRK2 upon Aβ1-42 fibril exposure. Moreover, we found that pharmacological inhibition of LRRK2, with two different kinase inhibitors, can attenuate Aβ1-42 -mediated inflammation and favor the clearance of Aβ1-42 fibrils in astrocytes. Overall, our findings report that LRRK2 kinase activity modulates astrocytic reactivity and functions in the presence of Aβ1-42 deposits and indicate that PD-linked LRRK2 might contribute to AD-related neuroinflammation and pathogenesis.- Published
- 2023
- Full Text
- View/download PDF
24. The potential convergence of NLRP3 inflammasome, potassium, and dopamine mechanisms in Parkinson's disease.
- Author
-
Pike AF, Szabò I, Veerhuis R, and Bubacco L
- Abstract
The pathology of Parkinson's disease (PD) is characterized by α-synuclein aggregation, microglia-mediated neuroinflammation, and dopaminergic neurodegeneration in the substantia nigra with collateral striatal dopamine signaling deficiency. Microglial NLRP3 inflammasome activation has been linked independently to each of these facets of PD pathology. The voltage-gated potassium channel Kv1.3, upregulated in microglia by α-synuclein and facilitating potassium efflux, has also been identified as a modulator of neuroinflammation and neurodegeneration in models of PD. Evidence increasingly suggests that microglial Kv1.3 is mechanistically coupled with NLRP3 inflammasome activation, which is contingent on potassium efflux. Potassium conductance also influences dopamine release from midbrain dopaminergic neurons. Dopamine, in turn, has been shown to inhibit NLRP3 inflammasome activation in microglia. In this review, we provide a literature framework for a hypothesis in which Kv1.3 activity-induced NLRP3 inflammasome activation, evoked by stimuli such as α-synuclein, could lead to microglia utilizing dopamine from adjacent dopaminergic neurons to counteract this process and fend off an activated state. If this is the case, a sufficient dopamine supply would ensure that microglia remain under control, but as dopamine is gradually siphoned from the neurons by microglial demand, NLRP3 inflammasome activation and Kv1.3 activity would progressively intensify to promote each of the three major facets of PD pathology: α-synuclein aggregation, microglia-mediated neuroinflammation, and dopaminergic neurodegeneration. Risk factors overlapping to varying degrees to render brain regions susceptible to such a mechanism would include a high density of microglia, an initially sufficient supply of dopamine, and poor insulation of the dopaminergic neurons by myelin., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
25. Dopamine signaling modulates microglial NLRP3 inflammasome activation: implications for Parkinson's disease.
- Author
-
Pike AF, Longhena F, Faustini G, van Eik JM, Gombert I, Herrebout MAC, Fayed MMHE, Sandre M, Varanita T, Teunissen CE, Hoozemans JJM, Bellucci A, Veerhuis R, and Bubacco L
- Subjects
- Animals, Dopamine metabolism, Mice, Mice, Inbred C57BL, Microglia metabolism, NLR Family, Pyrin Domain-Containing 3 Protein metabolism, Inflammasomes metabolism, Parkinson Disease pathology
- Abstract
Background: Parkinson's disease (PD) is characterized by the loss of nigral dopaminergic neurons leading to impaired striatal dopamine signaling, α-synuclein- (α-syn-) rich inclusions, and neuroinflammation. Degenerating neurons are surrounded by activated microglia with increased secretion of interleukin-1β (IL-1β), driven largely by the NLRP3 inflammasome. A critical role for microglial NLRP3 inflammasome activation in the progression of both dopaminergic neurodegeneration and α-syn pathology has been demonstrated in parkinsonism mouse models. Fibrillar α-syn activates this inflammasome in mouse and human macrophages, and we have shown previously that the same holds true for primary human microglia. Dopamine blocks microglial NLRP3 inflammasome activation in the MPTP model, but its effects in this framework, highly relevant to PD, remain unexplored in primary human microglia and in other in vivo parkinsonism models., Methods: Biochemical techniques including quantification of IL-1β secretion and confocal microscopy were employed to gain insight into dopamine signaling-mediated inhibition of the NLRP3 inflammasome mechanism in primary human microglia and the SYN120 transgenic mouse model. Dopamine and related metabolites were applied to human microglia together with various inflammasome activating stimuli. The involvement of the receptors through which these catecholamines were predicted to act were assessed with agonists in both species., Results: We show in primary human microglia that dopamine, L-DOPA, and high extracellular K
+ , but not norepinephrine and epinephrine, block canonical, non-canonical, and α-syn-mediated NLRP3 inflammasome-driven IL-1β secretion. This suggests that dopamine acts as an inflammasome inhibitor in human microglia. Accordingly, we provide evidence that dopamine exerts its inhibitory effect through dopamine receptor D1 and D2 (DRD1 and DRD2) signaling. We also show that aged mice transgenic for human C-terminally truncated (1-120) α-syn (SYN120 tg mice) display increased NLRP3 inflammasome activation in comparison to WT mice that is diminished upon DRD1 agonism., Conclusions: Dopamine inhibits canonical, non-canonical, and α-syn-mediated activation of the NLRP3 inflammasome in primary human microglia, as does high extracellular K+ . We suggest that dopamine serves as an endogenous repressor of the K+ efflux-dependent microglial NLRP3 inflammasome activation that contributes to dopaminergic neurodegeneration in PD, and that this reciprocation may account for the specific vulnerability of these neurons to disease pathology., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
26. APOE ε4 genotype-dependent cerebrospinal fluid proteomic signatures in Alzheimer's disease.
- Author
-
Konijnenberg E, Tijms BM, Gobom J, Dobricic V, Bos I, Vos S, Tsolaki M, Verhey F, Popp J, Martinez-Lage P, Vandenberghe R, Lleó A, Frölich L, Lovestone S, Streffer J, Bertram L, Blennow K, Teunissen CE, Veerhuis R, Smit AB, Scheltens P, Zetterberg H, and Visser PJ
- Subjects
- Aged, Aged, 80 and over, Amyloid beta-Peptides, Biomarkers, Genotype, Humans, Proteomics, Reproducibility of Results, tau Proteins, Alzheimer Disease genetics, Apolipoprotein E4 genetics
- Abstract
Background: Aggregation of amyloid β into plaques in the brain is one of the earliest pathological events in Alzheimer's disease (AD). The exact pathophysiology leading to dementia is still uncertain, but the apolipoprotein E (APOE) ε4 genotype plays a major role. We aimed to identify the molecular pathways associated with amyloid β aggregation using cerebrospinal fluid (CSF) proteomics and to study the potential modifying effects of APOE ε4 genotype., Methods: We tested 243 proteins and protein fragments in CSF comparing 193 subjects with AD across the cognitive spectrum (65% APOE ε4 carriers, average age 75 ± 7 years) against 60 controls with normal CSF amyloid β, normal cognition, and no APOE ε4 allele (average age 75 ± 6 years)., Results: One hundred twenty-nine proteins (53%) were associated with aggregated amyloid β. APOE ε4 carriers with AD showed altered concentrations of proteins involved in the complement pathway and glycolysis when cognition was normal and lower concentrations of proteins involved in synapse structure and function when cognitive impairment was moderately severe. APOE ε4 non-carriers with AD showed lower expression of proteins involved in synapse structure and function when cognition was normal and lower concentrations of proteins that were associated with complement and other inflammatory processes when cognitive impairment was mild. Repeating analyses for 114 proteins that were available in an independent EMIF-AD MBD dataset (n = 275) showed that 80% of the proteins showed group differences in a similar direction, but overall, 28% effects reached statistical significance (ranging between 6 and 87% depending on the disease stage and genotype), suggesting variable reproducibility., Conclusions: These results imply that AD pathophysiology depends on APOE genotype and that treatment for AD may need to be tailored according to APOE genotype and severity of the cognitive impairment.
- Published
- 2020
- Full Text
- View/download PDF
27. Impaired Innate Immunity Mechanisms in the Brain of Alzheimer's Disease.
- Author
-
Romagnoli M, Porcellini E, Carbone I, Veerhuis R, and Licastro F
- Subjects
- Aged, Aged, 80 and over, Alleles, Alzheimer Disease genetics, Female, Humans, Immunity, Innate genetics, Interferon Regulatory Factor-7 genetics, Interferon Regulatory Factor-7 metabolism, Interferon-alpha genetics, Interferon-alpha metabolism, Interferons genetics, Interferons metabolism, Male, Mediator Complex genetics, Mediator Complex metabolism, Middle Aged, RNA, Messenger genetics, RNA, Messenger metabolism, Alzheimer Disease immunology, Alzheimer Disease metabolism, Brain metabolism, Brain pathology, Hippocampus metabolism, Immunity, Innate physiology
- Abstract
Among environmental factors likely associated with Alzheimer's disease (AD), persistent virus infections, and age-related progressive decline of immune competence might play a pivotal role. However, AD antimicrobial brain immune responses are poorly investigated. The present study focused on genes involved in antimicrobial defenses, especially against virus infections, in the AD brain. In particular, mRNA levels of IRF7, MED23, IL28B, and IFN-α genes were analyzed in hippocampus and temporal cortex brain samples from AD and non-demented controls. All subjects were also genotyped for APOE ε, IRF7, MED23, and IL28B gene polymorphisms. Most AD patients showed decreased mRNA levels of all investigated genes in the hippocampus and temporal cortex. However, a small group of AD patients showed increased hippocampal mRNA expression of MED23, IL28B, and IFN-α. mRNA levels of MED23, IL28B, IFN-α from the hippocampus and those of MED23 from the temporal cortex were further decreased in APOE ε4 allele AD carriers. Moreover, rs6598008 polymorphism of IRF7 was significantly associated with decreased hippocampal expression of IRF7, MED23, IL28B, and IFN-α. These findings suggest that AD brains show impaired innate antimicrobial gene expression profiles, and individual genetic makeup, such as positivity for the APOE ε4 and IRF7 A alleles, might affect brain immune efficiency.
- Published
- 2020
- Full Text
- View/download PDF
28. Effects of an Aβ-antibody fragment on Aβ aggregation and astrocytic uptake are modulated by apolipoprotein E and J mimetic peptides.
- Author
-
Montoliu-Gaya L, Mulder SD, Veerhuis R, and Villegas S
- Subjects
- Adult, Alzheimer Disease genetics, Alzheimer Disease metabolism, Alzheimer Disease pathology, Amyloid beta-Peptides chemistry, Amyloid beta-Peptides genetics, Apolipoproteins E genetics, Apolipoproteins E metabolism, Apolipoproteins E pharmacology, Astrocytes metabolism, Astrocytes pathology, Cloning, Molecular, Clusterin genetics, Clusterin metabolism, Clusterin pharmacology, Endocytosis drug effects, Escherichia coli genetics, Escherichia coli metabolism, Female, Gene Expression, Genetic Vectors chemistry, Genetic Vectors metabolism, Humans, Male, Middle Aged, Molecular Mimicry, Peptide Fragments chemistry, Peptide Fragments genetics, Peptides genetics, Peptides metabolism, Primary Cell Culture, Protein Folding, Recombinant Proteins biosynthesis, Recombinant Proteins genetics, Recombinant Proteins pharmacology, Single-Chain Antibodies biosynthesis, Single-Chain Antibodies genetics, Amyloid beta-Peptides metabolism, Astrocytes drug effects, Peptide Fragments metabolism, Peptides pharmacology, Protein Aggregates drug effects, Single-Chain Antibodies pharmacology
- Abstract
Aβ-Immunotherapy has long been studied in the treatment of Alzheimer's disease (AD), but not how other molecules involved in the disease can affect antibody performance. We previously designed an antibody fragment, scFv-h3D6, and showed that it precludes Aβ-induced cytotoxicity by withdrawing Aβ oligomers from the amyloid pathway towards a non-toxic, worm-like pathway. ScFv-h3D6 was effective at the behavioral, cellular, and molecular levels in the 3xTg-AD mouse model. Because scFv-h3D6 treatment restored apolipoprotein E (apoE) and J (apoJ) concentrations to non-pathological values, and Aβ internalization by glial cells was found to be decreased in the presence of these apolipoproteins, we now aimed to test the influence of scFv-h3D6 on Aβ aggregation and cellular uptake by primary human astrocytes in the presence of therapeutic apoE and apoJ mimetic peptides (MPs). Firstly, we demonstrated by CD and FTIR that the molecules used in this work were well folded. Next, interactions between apoE or apoJ-MP, scFv-h3D6 and Aβ were studied by CD. The conformational change induced by the interaction of Aβ with apoE-MP was much bigger than the induced with apoJ-MP, in line with the observed formation of protective worm-like fibrils by the scFv-h3D6/Aβ complex in the presence of apoJ-MP, but not of apoE-MP. ScFv-h3D6, apoJ-MP, and apoE-MP to a different extent reduced Aβ uptake by astrocytes, and apoE-MP partially interfered with the dramatic reduction by scFv-h3D6 while apoJ-MP had no effect on scFv-h3D6 action. As sustained Aβ uptake by astrocytes may impair their normal functions, and ultimately neuronal viability, this work shows another beneficence of scFv-h3D6 treatment, which is not further improved by the use of apoE or apoJ mimetic peptides.
- Published
- 2017
- Full Text
- View/download PDF
29. Facilitating the Validation of Novel Protein Biomarkers for Dementia: An Optimal Workflow for the Development of Sandwich Immunoassays.
- Author
-
Del Campo M, Jongbloed W, Twaalfhoven HA, Veerhuis R, Blankenstein MA, and Teunissen CE
- Abstract
Different neurodegenerative disorders, such as Alzheimer's disease (AD) and frontotemporal dementia (FTD), lead to dementia syndromes. Dementia will pose a huge impact on society and thus it is essential to develop novel tools that are able to detect the earliest, most sensitive, discriminative, and dynamic biomarkers for each of the disorders. To date, the most common assays used in large-scale protein biomarker analysis are enzyme-linked immunosorbent assays (ELISA), such as the sandwich immunoassays, which are sensitive, practical, and easily implemented. However, due to the novelty of many candidate biomarkers identified during proteomics screening, such assays or the antibodies that specifically recognize the desired marker are often not available. The development and optimization of a new ELISA should be carried out with considerable caution since a poor planning can be costly, ineffective, time consuming, and it may lead to a misinterpretation of the findings. Previous guidelines described either the overall biomarker development in more general terms (i.e., the process from biomarker discovery to validation) or the specific steps of performing an ELISA procedure. However, a workflow describing and guiding the main issues in the development of a novel ELISA is missing. Here, we describe a specific and detailed workflow to develop and validate new ELISA for a successful and reliable validation of novel dementia biomarkers. The proposed workflow highlights the main issues in the development of an ELISA and covers several critical aspects, including production, screening, and selection of specific antibodies until optimal fine-tuning of the assay. Although these recommendations are designed to analyze novel biomarkers for dementia in cerebrospinal fluid, they are generally applicable for the development of immunoassays for biomarkers in other human body fluids or tissues. This workflow is designed to maximize the quality of the developed ELISA using a time- and cost-efficient strategy. This will facilitate the validation of the dementia biomarker candidates ultimately allowing accurate diagnostic conclusions.
- Published
- 2015
- Full Text
- View/download PDF
30. NG2 cells, a new trail for Alzheimer's disease mechanisms?
- Author
-
Nielsen HM, Ek D, Avdic U, Orbjörn C, Hansson O, Veerhuis R, Rozemuller AJ, Brun A, Minthon L, and Wennström M
- Subjects
- Aged, Aged, 80 and over, Alzheimer Disease pathology, Astrocytes pathology, Astrocytes physiology, Biomarkers cerebrospinal fluid, Brain pathology, Cell Survival physiology, Cells, Cultured, Female, Humans, Male, Middle Aged, Neuroglia pathology, Phosphorylation, Plaque, Amyloid pathology, tau Proteins cerebrospinal fluid, Alzheimer Disease physiopathology, Amyloid beta-Peptides metabolism, Antigens metabolism, Brain physiopathology, Neuroglia physiology, Peptide Fragments metabolism, Plaque, Amyloid physiopathology, Proteoglycans metabolism
- Abstract
Background: Neuron Glial 2 (NG2) cells are glial cells known to serve as oligodendrocyte progenitors as well as modulators of the neuronal network. Altered NG2 cell morphology and up-regulation as well as increased shedding of the proteoglycan NG2 expressed on the cell surface have been described in rodent models of brain injury. Here we describe alterations in the human NG2 cell population in response to pathological changes characteristic of Alzheimer's disease (AD)., Results: Immunohistological stainings of postmortem brain specimens from clinically diagnosed and postmortem verified AD patients and non-demented controls revealed reduced NG2 immunoreactivity as well as large numbers of NG2 positive astrocytes in individuals with high amyloid beta plaque load. Since fibrillar amyloid beta (Aβ)1-42 is the major component of AD-related senile plaques, we exposed human NG2 cells to oligomer- and fibril enriched preparations of Aβ1-42. We found that both oligomeric and fibrillar Aβ1-42 induced changes in NG2 cell morphology. Further, in vitro exposure to fibrillar Aβ1-42 decreased the NG2 concentrations in both cell lysates and supernatants. Interestingly, we also found significantly decreased levels of soluble NG2 in the cerebrospinal fluid (CSF) from clinically diagnosed AD patients compared to non-demented individuals. Additionally, the CSF NG2 levels were found to significantly correlate with the core AD biomarkers Aß1-42, T-tau and P-tau., Conclusion: Our results demonstrate major alterations in the NG2 cell population in relation to AD pathology which highlights the NG2 cell population as a new attractive research target in the search for cellular mechanisms associated with AD pathogenesis.
- Published
- 2013
- Full Text
- View/download PDF
31. Whether, when and how chronic inflammation increases the risk of developing late-onset Alzheimer's disease.
- Author
-
Eikelenboom P, Hoozemans JJ, Veerhuis R, van Exel E, Rozemuller AJ, and van Gool WA
- Abstract
Neuropathological studies have revealed the presence of a broad variety of inflammation-related proteins (complement factors, acute-phase proteins, pro-inflammatory cytokines) in Alzheimer's disease (AD) brains. These constituents of innate immunity are involved in several crucial pathogenic events of the underlying pathological cascade in AD, and recent studies have shown that innate immunity is involved in the etiology of late-onset AD. Genome-wide association studies have demonstrated gene loci that are linked to the complement system. Neuropathological and experimental studies indicate that fibrillar amyloid-β (Aβ) can activate the innate immunity-related CD14 and Toll-like receptor signaling pathways of glial cells for pro-inflammatory cytokine production. The production capacity of this pathway is under genetic control and offspring with a parental history of late-onset AD have a higher production capacity for pro-inflammatory cytokines. The activation of microglia by fibrillar Aβ deposits in the early preclinical stages of AD can make the brain susceptible later on for a second immune challenge leading to enhanced production of pro-inflammatory cytokines. An example of a second immune challenge could be systemic inflammation in patients with preclinical AD. Prospective epidemiological studies show that elevated serum levels of acute phase reactants can be considered as a risk factor for AD. Clinical studies suggest that peripheral inflammation increases the risk of dementia, especially in patients with preexistent cognitive impairment, and accelerates further deterioration in demented patients. The view that peripheral inflammation can increase the risk of dementia in older people provides scope for prevention.
- Published
- 2012
- Full Text
- View/download PDF
32. Goodpasture antigen-binding protein/ceramide transporter binds to human serum amyloid P-component and is present in brain amyloid plaques.
- Author
-
Mencarelli C, Bode GH, Losen M, Kulharia M, Molenaar PC, Veerhuis R, Steinbusch HW, De Baets MH, Nicolaes GA, and Martinez-Martinez P
- Subjects
- Alzheimer Disease genetics, Animals, Humans, Mice, Mice, Transgenic, Multiprotein Complexes genetics, Protein Binding, Protein Serine-Threonine Kinases genetics, Protein Structure, Tertiary, Serum Amyloid P-Component genetics, Alzheimer Disease blood, Brain metabolism, Multiprotein Complexes blood, Protein Serine-Threonine Kinases blood, Serum Amyloid P-Component metabolism
- Abstract
Serum amyloid P component (SAP) is a non-fibrillar glycoprotein belonging to the pentraxin family of the innate immune system. SAP is present in plasma, basement membranes, and amyloid deposits. This study demonstrates, for the first time, that the Goodpasture antigen-binding protein (GPBP) binds to human SAP. GPBP is a nonconventional Ser/Thr kinase for basement membrane type IV collagen. Also GPBP is found in plasma and in the extracellular matrix. In the present study, we demonstrate that GPBP specifically binds SAP in its physiological conformations, pentamers and decamers. The START domain in GPBP is important for this interaction. SAP and GPBP form complexes in blood and partly colocalize in amyloid plaques from Alzheimer disease patients. These data suggest the existence of complexes of SAP and GPBP under physiological and pathological conditions. These complexes are important for understanding basement membrane, blood physiology, and plaque formation in Alzheimer disease.
- Published
- 2012
- Full Text
- View/download PDF
33. Small heat shock proteins induce a cerebral inflammatory reaction.
- Author
-
Bruinsma IB, de Jager M, Carrano A, Versleijen AA, Veerhuis R, Boelens W, Rozemuller AJ, de Waal RM, and Verbeek MM
- Subjects
- Aged, Aged, 80 and over, Astrocytes metabolism, Astrocytes pathology, Cells, Cultured, Female, HSP20 Heat-Shock Proteins physiology, HSP27 Heat-Shock Proteins physiology, Heat-Shock Proteins physiology, Humans, Male, Molecular Chaperones, Myocytes, Smooth Muscle metabolism, Myocytes, Smooth Muscle pathology, Protein Serine-Threonine Kinases physiology, Recombinant Proteins pharmacology, Alzheimer Disease metabolism, Alzheimer Disease pathology, Brain metabolism, Brain pathology, Cerebral Amyloid Angiopathy metabolism, Cerebral Amyloid Angiopathy pathology, Heat-Shock Proteins, Small physiology, Inflammation Mediators physiology
- Abstract
More than 80% of Alzheimer's disease (AD) patients have some degree of cerebral amyloid angiopathy (CAA). In addition to arteries and veins, capillaries can also be affected. Capillary CAA (capCAA), rather than CAA in larger vessels, is associated with flame-like amyloid-beta (Aβ) deposits that may extend beyond the vessel wall and radiate into the neuropil, a phenomenon also known as "dyshoric angiopathy." Aβ deposits in AD, parenchymal as well as (cap)CAA and dyshoric angiopathy, are associated with a local inflammatory reaction, including activation of microglial cells and astrocytes that, among others, produce cytokines and reactive oxygen species. This neuroinflammatory reaction may account for at least part of the cognitive decline. In previous studies we observed that small heat shock proteins (sHsps) are associated with Aβ deposits in AD. In this study the molecular chaperones Hsp20, HspB8 and HspB2B3 were found to colocalize with CAA and capCAA in AD brains. In addition, Hsp20, HspB8 and HspB2B3 colocalized with intercellular adhesion molecule 1 (ICAM-1) in capCAA-associated dyshoric angiopathy. Furthermore, we demonstrated that Hsp20, HspB8 and HspB2B3 induced production of interleukin 8, soluble ICAM-1 and monocyte chemoattractant protein 1 by human leptomeningeal smooth muscle cells and human brain astrocytes in vitro and that Hsp27 inhibited production of transforming growth factor beta 1 and CD40 ligand. Our results suggest a central role for sHsps in the neuroinflammatory reaction in AD and CAA and thus in contributing to cognitive decline.
- Published
- 2011
- Full Text
- View/download PDF
34. Lipoprotein receptor-related protein-1 mediates amyloid-beta-mediated cell death of cerebrovascular cells.
- Author
-
Wilhelmus MM, Otte-Höller I, van Triel JJ, Veerhuis R, Maat-Schieman ML, Bu G, de Waal RM, and Verbeek MM
- Subjects
- Aged, Aged, 80 and over, Amyloid beta-Peptides analysis, Apoptosis, Blood Vessels metabolism, Blood Vessels pathology, Blood-Brain Barrier metabolism, Brain blood supply, Brain metabolism, Brain pathology, CD36 Antigens metabolism, Female, Humans, Low Density Lipoprotein Receptor-Related Protein-1 analysis, Male, Pericytes chemistry, Pericytes metabolism, Pericytes pathology, Receptors, LDL analysis, Receptors, LDL metabolism, Alzheimer Disease metabolism, Alzheimer Disease pathology, Amyloid beta-Peptides metabolism, Cerebral Amyloid Angiopathy, Familial metabolism, Cerebral Amyloid Angiopathy, Familial pathology, Low Density Lipoprotein Receptor-Related Protein-1 metabolism
- Abstract
Inefficient clearance of A beta, caused by impaired blood-brain barrier crossing into the circulation, seems to be a major cause of A beta accumulation in the brain of late-onset Alzheimer's disease patients and hereditary cerebral hemorrhage with amyloidosis Dutch type. We observed association of receptor for advanced glycation end products, CD36, and low-density lipoprotein receptor (LDLR) with cerebral amyloid angiopathy in both Alzheimer's disease and hereditary cerebral hemorrhage with amyloidosis Dutch type brains and increased low-density lipoprotein receptor-related protein-1 (LRP-1) expression by perivascular cells in cerebral amyloid angiopathy. We investigated if these A beta receptors are involved in A beta internalization and in A beta-mediated cell death of human cerebrovascular cells and astrocytes. Expression of both the LRP-1 and LDLR by human brain pericytes and leptomeningeal smooth muscle cells, but not by astrocytes, increased on incubation with A beta. Receptor-associated protein specifically inhibited A beta-mediated up-regulation of LRP-1, but not of LDLR, and receptor-associated protein also decreased A beta internalization and A beta-mediated cell death. We conclude that especially LRP-1 and, to a minor extent, LDLR are involved in A beta internalization by and A beta-mediated cell death of cerebral perivascular cells. Although perivascular cells may adapt their A beta internalization capacity to the levels of A beta present, saturated LRP-1/LDLR-mediated uptake of A beta results in degeneration of perivascular cells.
- Published
- 2007
- Full Text
- View/download PDF
35. DNA polymerase-beta is expressed early in neurons of Alzheimer's disease brain and is loaded into DNA replication forks in neurons challenged with beta-amyloid.
- Author
-
Copani A, Hoozemans JJ, Caraci F, Calafiore M, Van Haastert ES, Veerhuis R, Rozemuller AJ, Aronica E, Sortino MA, and Nicoletti F
- Subjects
- Adult, Aged, Aged, 80 and over, Alzheimer Disease genetics, Alzheimer Disease pathology, Brain drug effects, Brain pathology, Cells, Cultured, DNA Polymerase beta genetics, DNA Polymerase beta metabolism, DNA Replication drug effects, Female, Gene Expression Regulation, Enzymologic drug effects, Gene Expression Regulation, Enzymologic physiology, Humans, Male, Middle Aged, Neurons drug effects, Neurons pathology, Retinoblastoma Protein metabolism, Alzheimer Disease enzymology, Amyloid beta-Peptides toxicity, Brain enzymology, DNA Polymerase beta biosynthesis, DNA Replication physiology, Neurons enzymology
- Abstract
Cultured neurons exposed to synthetic beta-amyloid (Abeta) fragments reenter the cell cycle and initiate a pathway of DNA replication that involves the repair enzyme DNA polymerase-beta (DNA pol-beta) before undergoing apoptotic death. In this study, by performing coimmunoprecipitation experiments on cross-linked nucleoprotein fragments from Abeta-treated neurons, we demonstrate that DNA pol-beta coimmunoprecipitates with cell division cycle 45 (Cdc45) and with DNA primase in short nucleoprotein fragments. This indicates that DNA pol-beta is loaded into neuronal DNA replication forks after Abeta treatment. In response to Abeta the canonical DNA-synthesizing enzyme DNA pol-delta also was loaded into neuronal replication forks, but at later times than DNA pol-beta. Methoxyamine, an inhibitor of the apurinic/apyrimidinic endonuclease that allows for the recruitment of DNA pol-beta during the process of base excision repair (BER), failed to affect coimmunoprecipitation between DNA pol-beta and Cdc45, indicating that DNA pol-beta loading to the replication forks is independent of DNA breaks. However, methoxyamine reduced DNA replication and ensuing apoptosis in neurons exposed to Abeta, suggesting that an efficient BER process allows DNA replication to proceed up to the threshold for death. These data demonstrate that DNA pol-beta is an essential component of the DNA replication machinery in Abeta-treated neurons and additionally support the hypothesis of a close association of cell cycle events with neuronal death in Alzheimer's disease (AD). Accordingly, by investigating the neuronal expression of DNA pol-beta, along with phosphorylated retinoblastoma protein and neurofibrillary changes in AD brain, we show an early involvement of DNA pol-beta in the pathogenesis of AD.
- Published
- 2006
- Full Text
- View/download PDF
36. Maximal COX-2 and ppRb expression in neurons occurs during early Braak stages prior to the maximal activation of astrocytes and microglia in Alzheimer's disease.
- Author
-
Hoozemans JJ, van Haastert ES, Veerhuis R, Arendt T, Scheper W, Eikelenboom P, and Rozemuller AJ
- Abstract
Neuronal expression of cyclooxygenase-2 (COX-2) and cell cycle proteins is suggested to contribute to neurodegeneration during Alzheimer's disease (AD). The stimulus that induces COX-2 and cell cycle protein expression in AD is still elusive. Activated glia cells are shown to secrete substances that can induce expression of COX-2 and cell cycle proteins in vitro. Using post mortem brain tissue we have investigated whether activation of microglia and astrocytes in AD brain can be correlated with the expression of COX-2 and phosphorylated retinoblastoma protein (ppRb). The highest levels of neuronal COX-2 and ppRb immunoreactivity are observed in the first stages of AD pathology (Braak 0-II, Braak A). No significant difference in COX-2 or ppRb neuronal immunoreactivity is observed between Braak stage 0 and later Braak stages for neurofibrillary changes or amyloid plaques. The mean number of COX-2 or ppRb immunoreactive neurons is significantly decreased in Braak stage C compared to Braak stage A for amyloid deposits. Immunoreactivity for glial markers KP1, CR3/43 and GFAP appears in the later Braak stages and is significantly increased in Braak stage V-VI compared to Braak stage 0 for neurofibrillary changes. In addition, a significant negative correlation is observed between the presence of KP1, CR3/43 and GFAP immunoreactivity and the presence of neuronal immunoreactivity for COX-2 and ppRb. These data show that maximal COX-2 and ppRb immunoreactivity in neurons occurs during early Braak stages prior to the maximal activation of astrocytes and microglia. In contrast to in vitro studies, post mortem data do not support a causal relation between the activation of microglia and astrocytes and the expression of neuronal COX-2 and ppRb in the pathological cascade of AD.
- Published
- 2005
- Full Text
- View/download PDF
37. How chronic inflammation can affect the brain and support the development of Alzheimer's disease in old age: the role of microglia and astrocytes.
- Author
-
Blasko I, Stampfer-Kountchev M, Robatscher P, Veerhuis R, Eikelenboom P, and Grubeck-Loebenstein B
- Subjects
- Aging pathology, Aging physiology, Alzheimer Disease drug therapy, Alzheimer Disease physiopathology, Amyloid beta-Peptides metabolism, Amyloid beta-Peptides physiology, Amyloid beta-Protein Precursor metabolism, Amyloid beta-Protein Precursor physiology, Anti-Inflammatory Agents therapeutic use, Astrocytes metabolism, Astrocytes pathology, Astrocytes physiology, Brain drug effects, Brain pathology, Chronic Disease, Humans, Immunity, Innate physiology, Inflammation drug therapy, Inflammation physiopathology, Microglia metabolism, Microglia pathology, Microglia physiology, Aging immunology, Alzheimer Disease etiology, Brain physiopathology, Inflammation complications
- Abstract
A huge amount of evidence has implicated amyloid beta (A beta) peptides and other derivatives of the amyloid precursor protein (beta APP) as central to the pathogenesis of Alzheimer's disease (AD). It is also widely recognized that age is the most important risk factor for AD and that the innate immune system plays a role in the development of neurodegeneration. Little is known, however, about the molecular mechanisms that underlie age-related changes of innate immunity and how they affect brain pathology. Aging is characteristically accompanied by a shift within innate immunity towards a pro-inflammatory status. Pro-inflammatory mediators such as tumour necrosis factor-alpha or interleukin-1 beta can then in combination with interferon-gamma be toxic on neurons and affect the metabolism of beta APP such that increased concentrations of amyloidogenic peptides are produced by neuronal cells as well as by astrocytes. A disturbed balance between the production and the degradation of A beta can trigger chronic inflammatory processes in microglial cells and astrocytes and thus initiate a vicious circle. This leads to a perpetuation of the disease.
- Published
- 2004
- Full Text
- View/download PDF
38. In vivo degradation of rat C1q induced by intravenous injection of soluble IgG aggregates.
- Author
-
Veerhuis R, van Es LA, and Daha MR
- Subjects
- Animals, Antigen-Antibody Complex, Chromatography, Gel, Complement Activation, Complement C1q, Electrophoresis, Polyacrylamide Gel, Hemolysis, Liver metabolism, Male, Rats, Rats, Inbred Strains, Time Factors, Complement Activating Enzymes metabolism, Immunoglobulin G metabolism
- Abstract
Immune complexes are able to bind and activate the first component of complement, C1. Upon activation of C1, C1r and C1s are rapidly inactivated by C1-In which also forms a complex with these two subcomponents, resulting in their release from C1-immune aggregate complexes. The fate of C1q after the binding C1 to immune complexes in vivo is not clear and, therefore the clearance of radiolabelled rat C1q was investigated in normal rats and in rats receiving soluble aggregated human IgG. 125I-labelled rat C1q was cleared with a half-life (T 1/2) of 12.4 hr in normal rats. Injection of AIgG into rats that had previously received 125I-C1q accelerated the clearance of 125I-C1q, resulting, finally, in a T 1/2 of 53 min. The levels of circulating endogenous C1q were also followed using haemolytic titrations and immunochemical measurements. Directly after injection of AIgG into rats, there was a rapid decrease in C1q haemolytic activity to less than 25% of the initial value after 10 min. The rate of disappearance of C1q antigen, was, however, much slower, the lowest concentration being 30% at 2 hr. C1q haemolytic activity and the C1q antigen level returned to virtually normal values after 24 hr. Plasma samples were taken at different time intervals after the injection of AIgG and subjected to gel filtration on Sephacryl S-400 columns. It was found that, in the 10 min samples, C1q antigen and C1q haemolytic activity, each with an estimated molecular weight (MW) of 400,000, were detected together. In addition, there was C1q antigen with a MW of less than 69,000 without C1q haemolytic activity. SDS-PAGE analysis of the various serum samples indicated that the low MW C1q antigen had an apparent MW of 25,000. Measurement of uptake of 125I-C1q in various organs indicated that the main site of clearance of 125I-C1q is the liver.
- Published
- 1985
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.