Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Annemieke Rozemuller is active.

Publication


Featured researches published by Annemieke Rozemuller.


Acta Neuropathologica | 2010

Nomenclature and nosology for neuropathologic subtypes of frontotemporal lobar degeneration: an update

Ian R. A. Mackenzie; Manuela Neumann; Eileen H. Bigio; Nigel J. Cairns; Irina Alafuzoff; Jillian J. Kril; Gabor G. Kovacs; Bernardino Ghetti; Glenda M. Halliday; Ida E. Holm; Wouter Kamphorst; Tamas Revesz; Annemieke Rozemuller; Samir Kumar-Singh; Haruhiko Akiyama; Atik Baborie; Salvatore Spina; Dennis W. Dickson; John Q. Trojanowski; David Mann

One year ago, in this journal, we published a recommended nomenclature for the neuropathologic subtypes of frontotemporal lobar degeneration (FTLD) [7]. A major impetus behind this was to resolve the confusion that had arisen around the use of the term “FTLD with ubiquitinated inclusions” (FTLD-U), following the discovery that the molecular pathology of these cases was heterogeneous, with most, but not all, being characterized by pathological TDP-43 [6, 11]. In addition, a system of nosology was introduced that grouped the FTLD subtypes into broad categories, based on the molecular defect that is most characteristic, according to current evidence. This system provided a concise and consistent terminology that has now been widely adopted in the literature. Another anticipated advantage was the ability to readily accommodate new discoveries. At the time, we did not anticipate how quickly this attribute would be put to use.


American Journal of Pathology | 2009

The Unfolded Protein Response Is Activated in Pretangle Neurons in Alzheimer's Disease Hippocampus

Jeroen J.M. Hoozemans; Elise S. van Haastert; Diana A.T. Nijholt; Annemieke Rozemuller; Piet Eikelenboom; Wiep Scheper

Accumulation of misfolded proteins in the endoplasmic reticulum triggers a cellular stress response called the unfolded protein response (UPR) that protects the cell against the toxic buildup of misfolded proteins. Previously, we reported that UPR activation is increased in Alzheimers disease (AD) patients. How the UPR relates to the pathological hallmarks of AD is still elusive. In the present study, the involvement of UPR activation in neurofibrillary degeneration in AD was investigated. Immunoreactivity for the phosphorylated UPR activation markers pancreatic ER kinase (pPERK), eukaryotic initiation factor 2alpha, and inositol-requiring enzyme 1alpha was observed in hippocampal neurons associated with granulovacuolar degeneration. The percentage of pPERK-immunoreactive neurons was increased in AD cases compared with nondemented control cases and with the Braak stage for neurofibrillary changes. Although absent from neurofibrillary tangles, pPERK immunoreactivity was most abundant in neurons with diffuse localization of phosphorylated tau protein. Additional analyses showed that pPERK immunoreactivity was associated with ubiquitin and the ubiquitin binding protein p62. A strong co-occurrence of immunoreactivity for both pPERK and glycogen synthase kinase 3beta in neurons was also observed. Together, these data indicate that UPR activation in AD neurons occurs at an early stage of neurofibrillary degeneration and suggest that the prolonged activation of the UPR is involved in both tau phosphorylation and neurodegeneration in AD pathogenesis.


Acta Neuropathologica | 2009

Nomenclature for neuropathologic subtypes of frontotemporal lobar degeneration: Consensus recommendations

Ian R. Mackenzie; Manuela Neumann; Eileen H. Bigio; Nigel J. Cairns; Irina Alafuzoff; Jillian J. Kril; Gabor G. Kovacs; Bernardino Ghetti; Glenda M. Halliday; Ida E. Holm; Wouter Kamphorst; Tamas Revesz; Annemieke Rozemuller; Samir Kumar-Singh; Haruhiko Akiyama; Atik Baborie; Salvatore Spina; Dennis W. Dickson; John Q. Trojanowski; David Mann

Nomenclature for neuropathologic subtypes of frontotemporal lobar degeneration : consensus recommendations


Free Radical Biology and Medicine | 2008

Nrf2-induced antioxidant protection : A promising target to counteract ROS-mediated damage in neurodegenerative disease?

Helga E. de Vries; Maarten E. Witte; David Hondius; Annemieke Rozemuller; Benjamin Drukarch; Jeroen J.M. Hoozemans; Jack van Horssen

Neurodegenerative diseases share various pathological features, such as accumulation of aberrant protein aggregates, microglial activation, and mitochondrial dysfunction. These pathological processes are associated with generation of reactive oxygen species (ROS), which cause oxidative stress and subsequent damage to essential molecules, such as lipids, proteins, and DNA. Hence, enhanced ROS production and oxidative injury play a cardinal role in the onset and progression of neurodegenerative disorders. To maintain a proper redox balance, the central nervous system is endowed with an antioxidant defense mechanism consisting of endogenous antioxidant enzymes. Expression of most antioxidant enzymes is tightly controlled by the antioxidant response element (ARE) and is activated by nuclear factor E2-related factor 2 (Nrf2). In past years reports have highlighted the protective effects of Nrf2 activation in reducing oxidative stress in both in vitro and in vivo models of neurodegenerative disorders. Here we provide an overview of the involvement of ROS-induced oxidative damage in Alzheimers disease, Parkinsons disease, and Huntingtons disease and we discuss the potential therapeutic effects of antioxidant enzymes and compounds that activate the Nrf2-ARE pathway.


Journal of Neural Transmission | 2006

The significance of neuroinflammation in understanding Alzheimer’s disease

Piet Eikelenboom; Robert Veerhuis; Wiep Scheper; Annemieke Rozemuller; W.A. van Gool; J. J. M. Hoozemans

Summary.The interest of scientists in the involvement of inflammation-related mechanisms in the pathogenesis of Alzheimer’s disease (AD) goes back to the work of one of the pioneers of the study of this disease. About hundred years ago Oskar Fischer stated that the crucial step in the plaque formation is the extracellular deposition of a foreign substance that provokes an inflammatory reaction followed by a regenerative response of the surrounding nerve fibers. Eighty years later immunohistochemical studies revealed that amyloid plaques are indeed co-localized with a broad variety of inflammation-related proteins (complement factors, acute-phase proteins, pro-inflammatory cytokines) and clusters of activated microglia. These findings have led to the view that the amyloid plaque is the nidus of a non-immune mediated chronic inflammatory response locally induced by fibrillar Aβ deposits. Recent neuropathological studies show a close relationship between fibrillar Aβ deposits, inflammation and neuroregeneration in relatively early stages of AD pathology preceding late AD stages characterized by extensive tau-related neurofibrillary changes.In the present work we will review the role of inflammation in the early stage of AD pathology and particularly the role of inflammation in Aβ metabolism and deposition. We also discuss the possibilities of inflammation-based therapeutic strategies in AD.


Brain | 2012

The clinical and pathological phenotype of C9ORF72 hexanucleotide repeat expansions.

Javier Simón-Sánchez; Elise G.P. Dopper; Petra E. Cohn-Hokke; Renate K. Hukema; Nayia Nicolaou; Harro Seelaar; J. Roos A. de Graaf; Inge de Koning; Natasja M. van Schoor; Dorly J. H. Deeg; Marion Smits; Joost Raaphorst; Leonard H. van den Berg; Helenius J. Schelhaas; Christine E. M. de Die-Smulders; Danielle Majoor-Krakauer; Annemieke Rozemuller; Rob Willemsen; Yolande A.L. Pijnenburg; Peter Heutink; John C. van Swieten

There is increasing evidence that frontotemporal dementia and amyotrophic lateral sclerosis are part of a disease continuum. Recently, a hexanucleotide repeat expansion in C9orf72 was identified as a major cause of both sporadic and familial frontotemporal dementia and amyotrophic lateral sclerosis. The aim of this study was to investigate clinical and neuropathological characteristics of hexanucleotide repeat expansions in C9orf72 in a large cohort of Dutch patients with frontotemporal dementia. Repeat expansions were successfully determined in a cohort of 353 patients with sporadic or familial frontotemporal dementia with or without amyotrophic lateral sclerosis, and 522 neurologically normal controls. Immunohistochemistry was performed in a series of 10 brains from patients carrying expanded repeats using a panel of antibodies. In addition, the presence of RNA containing GGGGCC repeats in paraffin-embedded sections of post-mortem brain tissue was investigated using fluorescence in situ hybridization with a locked nucleic acid probe targeting the GGGGCC repeat. Hexanucleotide repeat expansions in C9orf72 were found in 37 patients with familial (28.7%) and five with sporadic frontotemporal dementia (2.2%). The mean age at onset was 56.9 ± 8.3 years (range 39-76), and disease duration 7.6 ± 4.6 years (range 1-22). The clinical phenotype of these patients varied between the behavioural variant of frontotemporal dementia (n = 34) and primary progressive aphasia (n = 8), with concomitant amyotrophic lateral sclerosis in seven patients. Predominant temporal atrophy on neuroimaging was present in 13 of 32 patients. Pathological examination of the 10 brains from patients carrying expanded repeats revealed frontotemporal lobar degeneration with neuronal transactive response DNA binding protein-positive inclusions of variable type, size and morphology in all brains. Fluorescence in situ hybridization analysis of brain material from patients with the repeat expansion, a microtubule-associated protein tau or a progranulin mutation, and controls did not show RNA-positive inclusions specific for brains with the GGGGCC repeat expansion. The hexanucleotide repeat expansion in C9orf72 is an important cause of frontotemporal dementia with and without amyotrophic lateral sclerosis, and is sometimes associated with primary progressive aphasia. Neuropathological hallmarks include neuronal and glial inclusions, and dystrophic neurites containing transactive response DNA binding protein. Future studies are needed to explain the wide variation in clinical presentation.


Neurology | 2012

Cerebrospinal fluid markers for differential dementia diagnosis in a large memory clinic cohort

Niki S.M. Schoonenboom; Fransje Reesink; N. A. Verwey; M. I. Kester; Charlotte E. Teunissen; P.M. van de Ven; Yolande A.L. Pijnenburg; Marinus A. Blankenstein; Annemieke Rozemuller; P. Scheltens; W.M. van der Flier

Objective: To determine how amyloid β 42 (Aβ42), total tau (t-tau), and phosphorylated tau (p-tau) levels in CSF behave in a large cohort of patients with different types of dementia. Methods: Baseline CSF was collected from 512 patients with Alzheimer disease (AD) and 272 patients with other types of dementia (OD), 135 patients with a psychiatric disorder (PSY), and 275 patients with subjective memory complaints (SMC). Aβ42, t-tau, and p-tau (at amino acid 181) were measured in CSF by ELISA. Autopsy was obtained in a subgroup of 17 patients. Results: A correct classification of patients with AD (92%) and patients with OD (66%) was accomplished when CSF Aβ42 and p-tau were combined. Patients with progressive supranuclear palsy had normal CSF biomarker values in 90%. Patients with Creutzfeldt-Jakob disease demonstrated an extremely high CSF t-tau at a relatively normal CSF p-tau. CSF AD biomarker profile was seen in 47% of patients with dementia with Lewy bodies (DLB), 38% in corticobasal degeneration (CBD), and almost 30% in frontotemporal lobar degeneration (FTLD) and vascular dementia (VaD). PSY and SMC patients had normal CSF biomarkers in 91% and 88%. Older patients are more likely to have a CSF AD profile. Concordance between clinical and neuropathologic diagnosis was 85%. CSF markers reflected neuropathology in 94%. Conclusion: CSF Aβ42, t-tau, and p-tau are useful in differential dementia diagnosis. However, in DLB, FTLD, VaD, and CBD, a substantial group exhibit a CSF AD biomarker profile, which requires more autopsy confirmation in the future.


Acta Neuropathologica | 2001

Cyclooxygenase expression in microglia and neurons in Alzheimer's disease and control brain

Jeroen J.M. Hoozemans; Annemieke Rozemuller; I. Janssen; C. J. A. De Groot; Robert Veerhuis; Piet Eikelenboom

Abstract. Epidemiological studies suggest that non-steroidal anti-inflammatory drugs (NSAIDs) lower the risk of developing Alzheimers disease (AD). Most NSAIDs act upon local inflammatory events by inhibiting the expression or activation of cylooxygenase (COX). In the present study the expression of COX-1 and COX-2 in AD and non-demented control temporal and frontal cortex was investigated using immunohistochemistry. COX-1 expression was detected in microglial cells, while COX-2 expression was found in neuronal cells. In AD brains, COX-1-positive microglial cells were primarily associated with amyloid β plaques, while the number of COX-2-positive neurons was increased compared to that in control brains. No COX expression was detected in astrocytes. In vitro, primary human microglial and astrocyte cultures, and human neuroblastoma cells (SK-N-SH) were found to secrete prostaglandin E2 (PGE2), especially when stimulated. PGE2 synthesis by astrocytes and SK-N-SH cells was stimulated by interleukin-1β. Microglial cell PGE2 synthesis was stimulated by lipopolysaccharide only. Although astrocytes are used in studies in vitro to investigate the role of COX in AD, there are no indications that these cells express COX-1 or COX-2 in vivo. The different distribution patterns of COX-1 and COX-2 in AD could implicate that these enzymes are involved in different cellular processes in the pathogenesis of AD.


Acta Neuropathologica | 2009

Staging/typing of Lewy body related α-synuclein pathology: a study of the BrainNet Europe Consortium

Irina Alafuzoff; Thomas Arzberger; Safa Al-Sarraj; Jeanne E. Bell; Istvan Bodi; Nenad Bogdanovic; Orso Bugiani; Isidro Ferrer; Ellen Gelpi; Stephen M. Gentleman; Giorgio Giaccone; James Ironside; Nikolaos Kavantzas; Andrew J. King; Penelope Korkolopoulou; Gabor G. Kovacs; David Meyronet; Camelia Maria Monoranu; Piero Parchi; Laura Parkkinen; Efstratios Patsouris; Wolfgang Roggendorf; Annemieke Rozemuller; Christine Stadelmann-Nessler; Nathalie Streichenberger; Dietmar R. Thal; Hans A. Kretzschmar

When 22 members of the BrainNet Europe (BNE) consortium assessed 31 cases with α-synuclein (αS) immunoreactive (IR) pathology applying the consensus protocol described by McKeith and colleagues in 2005, the inter-observer agreement was 80%, being lowest in the limbic category (73%). When applying the staging protocol described by Braak and colleagues in 2003, agreement was only 65%, and in some cases as low as 36%. When modifications of these strategies, i.e., McKeith’s protocol by Leverenz and colleagues from 2009, Braak’s staging by Müller and colleagues from 2005 were applied then the agreement increased to 78 and 82%, respectively. In both of these modifications, a reduced number of anatomical regions/blocks are assessed and still in a substantial number of cases, the inter-observer agreement differed significantly. Over 80% agreement in both typing and staging of αS pathology could be achieved when applying a new protocol, jointly designed by the BNE consortium. The BNE-protocol assessing αS-IR lesions in nine blocks offered advantages over the previous modified protocols because the agreement between the 22 observers was over 80% in most cases. Furthermore, in the BNE-protocol, the αS pathology is assessed as being present or absent and thus the quality of staining and the assessment of the severity of αS-IR pathology do not alter the inter-observer agreement, contrary to other assessment strategies. To reach these high agreement rates an entity of amygdala-predominant category was incorporated. In conclusion, here we report a protocol for assessing αS pathology that can achieve a high inter-observer agreement for both the assignment to brainstem, limbic, neocortical and amygdala-predominant categories of synucleinopathy and the Braak stages.


Antioxidants & Redox Signaling | 2011

Amyloid Beta Induces Oxidative Stress-Mediated Blood-Brain Barrier Changes in Capillary Amyloid Angiopathy

Anna Carrano; Jeroen J.M. Hoozemans; Saskia M. van der Vies; Annemieke Rozemuller; Jack van Horssen; Helga E. de Vries

Cerebral amyloid angiopathy (CAA) is frequently observed in Alzheimers disease (AD) and is characterized by deposition of amyloid beta (Aβ) in leptomeningeal and cortical brain vasculature. In 40% of AD cases, Aβ mainly accumulates in cortical capillaries, a phenomenon referred to as capillary CAA (capCAA). The aim of this study was to investigate blood-brain barrier (BBB) alterations in CAA-affected capillaries with the emphasis on tight junction (TJ) changes. First, capCAA brain tissue was analyzed for the distribution of TJs. Here, we show for the first time a dramatic loss of occludin, claudin-5, and ZO-1 in Aβ-laden capillaries surrounded by NADPH oxidase-2 (NOX-2)-positive activated microglia. Importantly, we observed abundant vascular expression of the Aβ transporter receptor for advanced glycation endproducts (RAGE). To unravel the underlying mechanism, a human brain endothelial cell line was stimulated with Aβ1-42 to analyze the effects of Aβ. We observed a dose-dependent cytotoxicity and increased ROS generation, which interestingly was reversed by administration of exogenous antioxidants, NOX-2 inhibitors, and by blocking RAGE. Taken together, our data evidently show that Aβ is toxic to brain endothelial cells via binding to RAGE and induction of ROS production, which ultimately leads to disruption of TJs and loss of BBB integrity.

Collaboration


Dive into the Annemieke Rozemuller's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Philip Scheltens

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Robert Veerhuis

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Giorgio Giaccone

Carlo Besta Neurological Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John C. van Swieten

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge