hosted by
publicationslist.org
    

Jinte Middeldorp


jintemiddeldorp@gmail.com

Journal articles

2011
J Middeldorp, E M Hol (2011)  GFAP in health and disease.   Prog Neurobiol 93: 3. 421-443 Mar  
Abstract: Glial fibrillary acidic protein (GFAP) is the main intermediate filament protein in mature astrocytes, but also an important component of the cytoskeleton in astrocytes during development. Major recent developments in astrocyte biology and the discovery of novel intermediate filament functions enticed the interest in the function of GFAP. The discovery of various GFAP splice variants gave an additional boost to explore this protein in more detail. The structural role of GFAP in astrocytes has been widely accepted for a long time, but over the years, GFAP has been shown to be involved in astrocyte functions, which are important during regeneration, synaptic plasticity and reactive gliosis. Moreover, different subpopulations of astrocytes have been identified, which are likely to have distinctive tasks in brain physiology and pathology, and which are not only classified by their spatial and temporal appearance, but also by their specific expression of intermediate filaments, including distinct GFAP isoforms. The presence of these isoforms enhances the complexity of the astrocyte cytoskeleton and is likely to underlie subtype specific functions. In this review we discuss the versatility of the GFAP cytoskeletal network from gene to function with a focus on astrocytes during human brain development, aging and disease.
Notes:
2010
Jinte Middeldorp, Karin Boer, Jacqueline A Sluijs, Lidia De Filippis, Férechté Encha-Razavi, Angelo L Vescovi, Dick F Swaab, Eleonora Aronica, Elly M Hol (2010)  GFAPdelta in radial glia and subventricular zone progenitors in the developing human cortex.   Development 137: 2. 313-321 Jan  
Abstract: A subpopulation of glial fibrillary acidic protein (GFAP)-expressing cells located along the length of the lateral ventricles in the subventricular zone (SVZ) have been identified as the multipotent neural stem cells of the adult mammalian brain. We have previously found that, in the adult human brain, a splice variant of GFAP, termed GFAPdelta, was expressed specifically in these cells. To investigate whether GFAPdelta is also present in the precursors of SVZ astrocytes during development and whether GFAPdelta could play a role in the developmental process, we analyzed GFAPdelta expression in the normal developing human cortex and in the cortex of foetuses with the migration disorder lissencephaly type II. We demonstrated for the first time that GFAPdelta is specifically expressed in radial glia and SVZ neural progenitors during human brain development. Expression of GFAPdelta in radial glia starts at around 13 weeks of pregnancy and disappears before birth. GFAPdelta is continuously expressed in the SVZ progenitors at later gestational ages and in the postnatal brain. Co-localization with Ki67 proved that these GFAPdelta-expressing cells are able to proliferate. Furthermore, we showed that the expression pattern of GFAPdelta was disturbed in lissencephaly type II. Overall, these results suggest that the adult SVZ is indeed a remnant of the foetal SVZ, which develops from radial glia. Furthermore, we provide evidence that GFAPdelta can distinguish resting astrocytes from proliferating SVZ progenitors.
Notes:
Simone A van den Berge, Jinte Middeldorp, C Eleana Zhang, Maurice A Curtis, Brian W Leonard, Diego Mastroeni, Pieter Voorn, Wilma D J van de Berg, Inge Huitinga, Elly M Hol (2010)  Longterm quiescent cells in the aged human subventricular neurogenic system specifically express GFAP-delta.   Aging Cell 9: 3. 313-326 Jun  
Abstract: A main neurogenic niche in the adult human brain is the subventricular zone (SVZ). Recent data suggest that the progenitors that are born in the human SVZ migrate via the rostral migratory stream (RMS) towards the olfactory bulb (OB), similar to what has been observed in other mammals. A subpopulation of astrocytes in the SVZ specifically expresses an assembly-compromised isoform of the intermediate filament protein glial fibrillary acidic protein (GFAP-delta). To further define the phenotype of these GFAP-delta expressing cells and to determine whether these cells are present throughout the human subventricular neurogenic system, we analysed SVZ, RMS and OB sections of 14 aged brain donors (ages 74-93). GFAP-delta was expressed in the SVZ along the ventricle, in the RMS and in the OB. The GFAP-delta cells in the SVZ co-expressed the neural stem cell (NSC) marker nestin and the cell proliferation markers proliferating cell nuclear antigen (PCNA) and Mcm2. Furthermore, BrdU retention was found in GFAP-delta positive cells in the SVZ. In the RMS, GFAP-delta was expressed in the glial net surrounding the neuroblasts. In the OB, GFAP-delta positive cells co-expressed PCNA. We also showed that GFAP-delta cells are present in neurosphere cultures that were derived from SVZ precursors, isolated postmortem from four brain donors (ages 63-91). Taken together, our findings show that GFAP-delta is expressed in an astrocytic subpopulation in the SVZ, the RMS and the OB. Importantly, we provide the first evidence that GFAP-delta is specifically expressed in longterm quiescent cells in the human SVZ, which are reminiscent of NSCs.
Notes:
K Boer, J Middeldorp, W G M Spliet, F Razavi, P C van Rijen, J C Baayen, E M Hol, E Aronica (2010)  Immunohistochemical characterization of the out-of frame splice variants GFAP Delta164/Deltaexon 6 in focal lesions associated with chronic epilepsy.   Epilepsy Res 90: 1-2. 99-109 Jun  
Abstract: GFAP Delta164/Deltaexon 6 are two out-of frame splice variants of GFAP. The aim of this study was to investigate the distribution of GFAP Delta164/Deltaexon 6 expressing cells, in focal lesions associated with chronic intractable epilepsy, in light of the increasing interest in the role of specific astrocyte subtypes in epilepsy. Immunocytochemical analysis, using an antibody against Delta164 and Deltaexon6 (GFAP+1), was performed in surgical specimens of patients with hippocampal sclerosis (HS), focal cortical dysplasia type IIB (FCD), cortical tubers of tuberous sclerosis complex (TSC), glioneuronal and glial tumors. Expression of GFAP+1 was also evaluated in developing and adult human control cortex and hippocampus. GFAP+1 immunoreactivity was undetectable in developing human brain. In control human hippocampus and cortex (from young controls) only occasional GFAP+1 positive cells were observed. In contrast, GFAP+1 immunoreactivity was consistently detected in the glial component of the epileptogenic lesions. Balloon cells in FCD and giant cells in TSC, only rarely express GFAP+1. GFAP+1 co-localized with GFAPalpha, but not with GFAPdelta. Co-localization with aquaporin 4 was observed around blood vessels. GFAP+1 immunoreactivity in epilepsy-associated pathologies reveals a specific subpopulation of astrocytes in regions of astrogliosis. Further studies on GFAP+1 positive astrocytes are important to understand whether the expression of this isoform may affect the cytoskeletal integrity and the shape and function of glial cells under pathological conditions. However, while the staining is increased in epilepsy-associated pathologies, GFAP+1 is expressed in a small percentage of astrocytes. Thus, the possible role of this subpopulation of astrocytes in epilepsy is likely minor, compared to astrocytes expressing other GFAP isoforms.
Notes:
2009
Jinte Middeldorp, Willem Kamphuis, Jacqueline A Sluijs, Dalila Achoui, Cathalijn H C Leenaars, Matthijs G P Feenstra, Paula van Tijn, David F Fischer, Celia Berkers, Huib Ovaa, Roy A Quinlan, Elly M Hol (2009)  Intermediate filament transcription in astrocytes is repressed by proteasome inhibition.   FASEB J 23: 8. 2710-2726 Aug  
Abstract: Increased expression of the astrocytic intermediate filament protein glial fibrillary acidic protein (GFAP) is a characteristic of astrogliosis. This process occurs in the brain during aging and neurodegeneration and coincides with impairment of the ubiquitin proteasome system. Inhibition of the proteasome impairs protein degradation; therefore, we hypothesized that the increase in GFAP may be the result of impaired proteasomal activity in astrocytes. We investigated the effect of proteasome inhibitors on GFAP expression and other intermediate filament proteins in human astrocytoma cells and in a rat brain model for astrogliosis. Extensive quantitative RT-PCR, immunocytochemistry, and Western blot analysis resulted unexpectedly in a strong decrease of GFAP mRNA to <4% of control levels [Control (DMSO) 100+/-19.2%; proteasome inhibitor (epoxomicin) 3.5+/-1.3%, n=8; P < or = 0.001] and a loss of GFAP protein in astrocytes in vitro. We show that the proteasome alters GFAP promoter activity, possibly mediated by transcription factors as demonstrated by a GFAP promoter-luciferase assay and RT(2) Profiler PCR array for human transcription factors. Most important, we demonstrate that proteasome inhibitors also reduce GFAP and vimentin expression in a rat model for induced astrogliosis in vivo. Therefore, proteasome inhibitors could serve as a potential therapy to modulate astrogliosis associated with CNS injuries and disease.
Notes:
Jinte Middeldorp, Simone A van den Berge, Eleonora Aronica, Dave Speijer, Elly M Hol (2009)  Specific human astrocyte subtype revealed by affinity purified GFAP antibody; unpurified serum cross-reacts with neurofilament-L in Alzheimer.   PLoS One 4: 11. 11  
Abstract: The human GFAP splice variants GFAPDelta164 and GFAPDeltaexon6 both result in a GFAP protein isoform with a unique out-of-frame carboxy-terminus that can be detected by the GFAP+1 antibody. We previously reported that GFAP+1 was expressed in astrocytes and in degenerating neurons in Alzheimer's disease brains. In this study we aimed at further investigating the neuronal GFAP+1 expression and we started by affinity purifying the GFAP+1 antibody. The purified antibody resulted in a loss of neuronal GFAP+1 signal, although other antibodies directed against the amino- and carboxy-terminus of GFAPalpha still revealed GFAP-immunopositive neurons, as described before. With an in-depth analysis of a western blot, followed by mass spectrometry we discovered that the previously detected neuronal GFAP+1 expression was due to cross-reactivity of the antibody with neurofilament-L (NF-L). This was confirmed by double-label fluorescent immunohistochemistry and western blotting with the unpurified GFAP+1 antibody and an antibody against NF-L. Our data imply that NF-L can accumulate in some tangle-like structures in Alzheimer brains. More importantly, the purified GFAP+1 antibody clearly revealed a specific subtype of astrocytes in the adult human brain. These large astrocytes are present throughout the brain, e.g., along the subventricular zone, in the hippocampus, in the striatum and in the spinal cord of controls, Alzheimer, and Parkinson patients. The presence of a specific GFAP-isoform suggests a specialized function of these astrocytes.
Notes:
L Martinian, K Boer, J Middeldorp, E M Hol, S M Sisodiya, W Squier, E Aronica, M Thom (2009)  Expression patterns of glial fibrillary acidic protein (GFAP)-delta in epilepsy-associated lesional pathologies.   Neuropathol Appl Neurobiol 35: 4. 394-405 Aug  
Abstract: Glial fibrillary acidic protein (GFAP)-delta is a novel isoform that differs in its C-terminal sequence from other GFAP isoforms. Previous studies suggest restriction of expression to the subpial layer, subventricular zone and the subgranular zone astrocytes, with an absence in pathological conditions causing reactive gliosis. GFAP-delta is speculated to have roles in regulation of astrocyte size and motility and a subpopulation of GFAP-delta-positive glia may be multipotent stem cells. The aim of this study was to investigate its expression in common causes of lesion-related refractory epilepsy.
Notes:
2008
Ming-Der Perng, Shu-Fang Wen, Terry Gibbon, Jinte Middeldorp, Jacqueline Sluijs, Elly M Hol, Roy A Quinlan (2008)  Glial fibrillary acidic protein filaments can tolerate the incorporation of assembly-compromised GFAP-delta, but with consequences for filament organization and alphaB-crystallin association.   Mol Biol Cell 19: 10. 4521-4533 Oct  
Abstract: The glial fibrillary acidic protein (GFAP) gene is alternatively spliced to give GFAP-alpha, the most abundant isoform, and seven other differentially expressed transcripts including GFAP-delta. GFAP-delta has an altered C-terminal domain that renders it incapable of self-assembly in vitro. When titrated with GFAP-alpha, assembly was restored providing GFAP-delta levels were kept low (approximately 10%). In a range of immortalized and transformed astrocyte derived cell lines and human spinal cord, we show that GFAP-delta is naturally part of the endogenous intermediate filaments, although levels were low (approximately 10%). This suggests that GFAP filaments can naturally accommodate a small proportion of assembly-compromised partners. Indeed, two other assembly-compromised GFAP constructs, namely enhanced green fluorescent protein (eGFP)-tagged GFAP and the Alexander disease-causing GFAP mutant, R416W GFAP both showed similar in vitro assembly characteristics to GFAP-delta and could also be incorporated into endogenous filament networks in transfected cells, providing expression levels were kept low. Another common feature was the increased association of alphaB-crystallin with the intermediate filament fraction of transfected cells. These studies suggest that the major physiological role of the assembly-compromised GFAP-delta splice variant is as a modulator of the GFAP filament surface, effecting changes in both protein- and filament-filament associations as well as Jnk phosphorylation.
Notes:
2006
C W Noorlander, P N E De Graan, J Middeldorp, J J B C Van Beers, G H A Visser (2006)  Ontogeny of hippocampal corticosteroid receptors: effects of antenatal glucocorticoids in human and mouse.   J Comp Neurol 499: 6. 924-932 Dec  
Abstract: Women at risk for preterm delivery are treated with synthetic glucocorticoids (GCs) to enhance fetal lung maturation. GCs can bind to two intracellular receptors, the glucocorticoid receptor (GR) and the mineralocorticoid receptor (MR), which function as transcription factors. Both are highly expressed in the hippocampus. Several studies have focused on adverse side effects of antenatal GC treatment. However, relatively little is known about the ontogeny of GR and MR, especially in human. Therefore, we studied the ontogeny of both receptors in the human and mouse hippocampus and investigated the effects of antenatal dexamethasone (dex) treatment, a synthetic glucocorticoid, on MR and GR mRNA levels during hippocampal development. The results demonstrate that MR mRNA was first expressed in mouse hippocampus at embryonic day (E)15.5, at the timepoint when dex was administered. In contrast, GR mRNA expression was first observed after birth at postnatal day (P)5. However, in the human hippocampus both receptors are expressed at 24 weeks of gestation, when antenatal GCs are administered in clinical practice. Quantitative in situ hybridization demonstrated that MR mRNA levels were reduced only shortly after dex treatment at E16, but were unaffected from E18 onwards. These findings indicate that a single antenatal dex administration at E15.5 transiently affects MR mRNA levels in the mouse hippocampus. No effect of antenatal dex treatment was found on the human hippocampus at the third trimester of pregnancy. These data on the prenatal ontogeny of both corticosteroid receptors in the human hippocampus is important for understanding the significance of fetal glucocorticoid or stress exposure and its potential effects on health and disease.
Notes:
Powered by PublicationsList.org.