Network


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

Hotspot


Dive into the research topics where Barbara Hobo is active.

Publication


Featured researches published by Barbara Hobo.


The FASEB Journal | 2003

Disease-specific accumulation of mutant ubiquitin as a marker for proteasomal dysfunction in the brain

David F. Fischer; Rob A.I. de Vos; Renske van Dijk; Femke M. S. De Vrij; Evelien A. Proper; Marc A. F. Sonnemans; Marian C. Verhage; Jacqueline A. Sluijs; Barbara Hobo; Mohamed Zouambia; Ernst N.H. Jansen Steur; Wouter Kamphorst; Elly M. Hol; Fred W. van Leeuwen

Molecular misreading of the ubiquitin‐B (UBB) gene results in a dinucleotide deletion in UBB mRNA. The resulting mutant protein, UBB+1, accumulates in the neuropathological hallmarks of Alzheimer disease. In vitro, UBB+1 inhibits proteasomal proteolysis, although it is also an ubiquitin fusion degradation substrate for the proteasome. Using the ligase chain reaction to detect dinucleotide deletions, we report here that UBB+1 transcripts are present in each neurodegenerative disease studied (tauo‐ and synucleinopathies) and even in control brain samples. In contrast to UBB+1 transcripts, UBB+1 protein accumulation in the ubiquitin‐containing neuropathological hallmarks is restricted to the tauopathies such as Pick disease, frontotemporal dementia, progressive supranuclear palsy, and argyrophilic grain disease. Remarkably, UBB+1 protein is not detected in the major forms of synucleinopathies (Lewy body disease and multiple system atrophy). The neurologically intact brain can cope with UBB+1 as lentivirally delivered UBB+1 protein is rapidly degraded in rat hippocampus, whereas the K29,48R mutant of UBB+1, which is not ubiquitinated, is abundantly expressed. The finding that UBB+1 protein only accumulates in tauopathies thus implies that the ubiquitin‐proteasome system is impaired specifically in this group of neurodegenerative diseases and not in synucleinopathies and that the presence of UBB+1 protein reports proteasomal dysfunction in the brain.—Fischer, D. F., de Vos, R. A. I., van Dijk, R., de Vrij, F. M. S., Proper, E. A., Sonnemans, M. A. F., Verhage, M. C., Sluijs, J. A., Hobo, B., Zouambia, M., Jansen Steur, E. N. H., Kamphorst, W., Hol, E. M., van Leeuwen, F. W. Disease‐specific accumulation of mutant ubiquitin as a marker for proteasomal dysfunction in the brain. FASEB J. 17, 2014–2024 (2003)


Neurobiology of Aging | 2009

Long-term proteasome dysfunction in the mouse brain by expression of aberrant ubiquitin

David F. Fischer; Renske van Dijk; Paula van Tijn; Barbara Hobo; Marian C. Verhage; Roel C. van der Schors; Ka Wan Li; Jan van Minnen; Elly M. Hol; Fred W. van Leeuwen

Many neurodegenerative diseases are characterized by deposits of ubiquitinated and aberrant proteins, suggesting a failure of the ubiquitin-proteasome system (UPS). The aberrant ubiquitin UBB(+1) is one of the ubiquitinated proteins accumulating in tauopathies such as Alzheimers disease (AD) and polyglutamine diseases such as Huntingtons disease. We have generated UBB(+1) transgenic mouse lines with post-natal neuronal expression of UBB(+1), resulting in increased levels of ubiquitinated proteins in the cortex. Moreover, by proteomic analysis, we identified expression changes in proteins involved in energy metabolism or organization of the cytoskeleton. These changes show a striking resemblance to the proteomic profiles of both AD brain and several AD mouse models. Moreover, UBB(+1) transgenic mice show a deficit in contextual memory in both water maze and fear conditioning paradigms. Although UBB(+1) partially inhibits the UPS in the cortex, these mice do not have an overt neurological phenotype. These mouse models do not replicate the full spectrum of AD-related changes, yet provide a tool to understand how the UPS is involved in AD pathological changes and in memory formation.


Neurology | 2006

Frameshift proteins in autosomal dominant forms of Alzheimer disease and other tauopathies

F. W. van Leeuwen; P van Tijn; Marc A. F. Sonnemans; Barbara Hobo; Derek A. Mann; C. Van Broeckhoven; Samir Kumar-Singh; Patrick Cras; Geneviève Leuba; Armand Savioz; Marion L.C. Maat-Schieman; Haruyasu Yamaguchi; Johan M. Kros; Wouter Kamphorst; Elly M. Hol; R.A.I. de Vos; David F. Fischer

Frameshift (+1) proteins such as APP+1 and UBB+1 accumulate in sporadic cases of Alzheimer disease (AD) and in older subjects with Down syndrome (DS). We investigated whether these proteins also accumulate at an early stage of neuropathogenesis in young DS individuals without neuropathology and in early-onset familial forms of AD (FAD), as well as in other tauopathies, such as Pick disease (PiD) or progressive supranuclear palsy (PSP). APP+1 is present in many neurons and beaded neurites in very young cases of DS, which suggests that it is axonally transported. In older DS patients (>37 years), a mixed pattern of APP+1 immunoreactivity was observed in healthy looking neurons and neurites, dystrophic neurites, in association with neuritic plaques, as well as neurofibrillary tangles. UBB+1 immunoreactivity was exclusively present in AD type of neuropathology. A similar pattern of APP+1 and UBB+1 immunoreactivity was also observed for FAD and much less explicit in nondemented controls after the age of 51 years. Furthermore, we observed accumulation of +1 proteins in other types of tauopathies, such as PiD, frontotemporal dementia, PSP and argyrophylic grain disease. These data suggest that accumulation of +1 proteins contributes to the early stages of dementia and plays a pathogenic role in a number of diseases that involve the accumulation of tau.


Proteomics | 2008

Proteasome subunit proteins and neuropathology in tauopathies and synucleinopathies: Consequences for proteomic analyses.

Mohamed Zouambia; David F. Fischer; Barbara Hobo; Rob A.I. de Vos; Elly M. Hol; Ian M. Varndell; Paul W. Sheppard; Fred W. van Leeuwen

Accumulation of proteins in inclusions in neurological disorders is partly due to dysfunction of the ubiquitin–proteasome system. Proteasomal dysfunction may be caused by misexpression of one or more of its subunits. A large number of antibodies reactive with proteasome subunits were screened on material from patients exhibiting tau‐ and synucleinopathies. Many antisera against proteasomal subunits (11S activator, 19S regulator ATPase/non‐ATPase, and 20S α and β resulted in a distinct nuclear and/or cytoplasmic staining of the entorhinal‐hippocampal area and the temporal cortex of Alzheimers disease (AD) patients. In particular an antibody directed against 19S regulator ATPase subunit 6b (S6b) specifically stained the neurofibrillary tangles and dystrophic neurites in AD, Down syndrome and aged nondemented controls. In other tauopathies (Picks disease, frontotemporal dementia, progressive supranuclear palsy and argyrophilic grain disease), neuronal and/or glial inclusions were also S6b immunoreactive. In contrast, in synucleinopathies (Lewy body disease (LBD) and multiple system atrophy) no S6b staining was seen. Real time quantitative PCR on the temporal cortex of AD patients revealed a significant increase in S6b subunit mRNA. This increase was not found in the gyrus cinguli anterior of patients with LBD. This differential expression of S6b most likely will result in different proteomic patterns. Here we present evidence to show that S6b coexists with a reporter for proteasomal dysfunction (ubiquitin+1), and we conclude that S6b transcript up‐regulation and the dysfunction in tauopathies may be functionally related.


Neurochemistry International | 2012

Mutant ubiquitin decreases amyloid β plaque formation in a transgenic mouse model of Alzheimer's disease.

Paula van Tijn; Frank J.A. Dennissen; Romina J.G. Gentier; Barbara Hobo; Denise J. H. P. Hermes; Harry W.M. Steinbusch; Fred W. van Leeuwen; David F. Fischer

The mutant ubiquitin UBB(+1) is a substrate as well as an inhibitor of the ubiquitin-proteasome system (UPS) and accumulates in the neuropathological hallmarks of Alzheimers disease (AD). A role for the UPS has been suggested in the generation of amyloid β (Aβ) plaques in AD. To investigate the effect of UBB(+1) expression on amyloid pathology in vivo, we crossed UBB(+1) transgenic mice with a transgenic line expressing AD-associated mutant amyloid precursor protein (APPSwe) and mutant presenilin 1 (PS1dE9), resulting in APPPS1/UBB(+1) triple transgenic mice. In these mice, we determined the Aβ levels at 3, 6, 9 and 11 months of age. Surprisingly, we found a significant decrease in Aβ deposition in amyloid plaques and levels of soluble Aβ(42) in APPPS1/UBB(+1) transgenic mice compared to APPPS1 mice at 6 months of age, without alterations in UBB(+1) protein levels or proteasomal chymotrypsin activity. These lowering effects of UBB(+1) on Aβ deposition were transient, as this relative decrease in plaque load was not significant in APPPS1/UBB(+1) mice at 9 and 11 months of age. We also show that APPPS1/UBB(+1) mice exhibit astrogliosis, indicating that they may not be improved functionally compared to APPPS1 mice despite the Aβ reduction. The molecular mechanism underlying this decrease in Aβ deposition in APPPS1/UBB(+1) mice is more complex than previously assumed because UBB(+1) is also ubiquitinated at K63 opening the possibility of additional effects of UBB(+1) (e.g. kinase activation).


Molecular and Cellular Neuroscience | 2010

Modest proteasomal inhibition by aberrant ubiquitin exacerbates aggregate formation in a Huntington disease mouse model

Remko de Pril; Barbara Hobo; Paula van Tijn; Raymund A.C. Roos; Fred W. van Leeuwen; David F. Fischer

UBB(+1), a mutant form of ubiquitin, is both a substrate and an inhibitor of the proteasome which accumulates in the neuropathological hallmarks of Huntington disease (HD). In vitro, expression of UBB(+1) and mutant huntingtin synergistically increase aggregate formation and polyglutamine induced cell death. We generated a UBB(+1) transgenic mouse line expressing UBB(+1) within the neurons of the striatum. In these mice lentiviral driven expression of expanded huntingtin constructs in the striatum results in a significant increase in neuronal inclusion formation. Although UBB(+1) transgenic mice show neither a decreased lifespan nor apparent neuronal loss, they appear to be more vulnerable to toxic insults like expanded polyglutamine proteins due to a modest proteasome inhibition. These findings underscore the relevance of an efficient ubiquitin-proteasome system in HD.


Journal of Neuroscience Research | 2010

Low levels of mutant ubiquitin are degraded by the proteasome in vivo

Paula van Tijn; Marian C. Verhage; Barbara Hobo; Fred W. van Leeuwen; David F. Fischer

The ubiquitin‐proteasome system fulfills a pivotal role in regulating intracellular protein turnover. Impairment of this system is implicated in the pathogenesis of neurodegenerative diseases characterized by ubiquitin‐ containing proteinaceous deposits. UBB+1, a mutant ubiquitin, is one of the proteins accumulating in the neuropathological hallmarks of tauopathies, including Alzheimers disease, and polyglutamine diseases. In vitro, UBB+1 properties shift from a proteasomal ubiquitin‐fusion degradation substrate at low expression levels to a proteasome inhibitor at high expression levels. Here we report on a novel transgenic mouse line (line 6663) expressing low levels of neuronal UBB+1. In these mice, UBB+1 protein is scarcely detectable in the neuronal cell population. Accumulation of UBB+1 commences only after intracranial infusion of the proteasome inhibitors lactacystin or MG262, showing that, at these low expression levels, the UBB+1 protein is a substrate for proteasomal degradation in vivo. In addition, accumulation of the protein serves as a reporter for proteasome inhibition. These findings strengthen our proposition that, in healthy brain, UBB+1 is continuously degraded and disease‐related UBB+1 accumulation serves as an endogenous marker for proteasomal dysfunction. This novel transgenic line can give more insight into the intrinsic properties of UBB+1 and its role in neurodegenerative disease.


Physiology & Behavior | 2011

Alzheimer-associated mutant ubiquitin impairs spatial reference memory.

Paula van Tijn; Barbara Hobo; Marian C. Verhage; Melly S. Oitzl; Fred W. van Leeuwen; David F. Fischer

UBB(+1) is a mutant ubiquitin which accumulates in the hallmarks of tauopathies, including Alzheimers disease. Transgenic mice expressing high levels of neuronal UBB(+1) exhibit moderately decreased proteasome activity and spatial reference memory deficits at 9months of age. In the present study, we characterized the behavioral phenotype of male UBB(+1) transgenic mice at different ages. We show that UBB(+1) transgenic mice displayed an age-related functional decline similar to wild-type littermates, without gross neurological abnormalities or alterations in procedural motor-learning and motor coordination. At 15months of age, a transgene-specific spatial learning deficit was dependent on the period of training in the Morris watermaze. This deficit could be eliminated after additional training. We conclude that the previously reported spatial reference memory deficits of UBB(+1) transgenic mice persist during aging. In addition, our results demonstrate that the subtle defect in spatial reference memory formation, caused by a decrease in forebrain proteasome activity, is a persistent defect and not a structural defect.


Journal of Neurochemistry | 2004

Frame-shifted amyloid precursor protein found in Alzheimer's disease and Down's syndrome increases levels of secreted amyloid β40

R. van Dijk; David F. Fischer; Jacqueline A. Sluijs; Marc A. F. Sonnemans; Barbara Hobo; Luc Mercken; D. M. A. Mann; Elly M. Hol; F. W. van Leeuwen

Frame‐shifted amyloid precursor protein (APP+1), which has a truncated out‐of‐frame C‐terminus, accumulates in the neuropathological hallmarks of patients with Alzheimers disease pathology. To study a possible involvement of APP+1 in the pathogenesis of Alzheimers disease, we expressed APP695 and APP+1 in the HEK293 cell‐line and studied whether the processing of APP695 was affected. APP+1 is a secretory protein, but high expression of APP695 and APP+1 results in the formation of intracellular aggregate‐like structures containing both proteins and Fe65, an adaptor protein that interacts with APP695. APP+1 is shown to interact with APP695, suggesting that these structures consist of functional protein complexes. Such an interaction can also be anticipated in post‐mortem brains of young Downs syndrome patients without any sign of neuropathology. Here we observed APP+1 immunoreactivity in beaded fibres. Additional support for functional consequences on the processing of APP695 comes from a 1.4‐fold increase in levels of secreted amyloid β40 in cells co‐expressing APP695 and APP+1, although APP+1 itself does not contain the amyloid β sequence. Taken together, these data show that co‐expression of APP695 and APP+1 affects the processing of APP695 in a pro‐amyloidogenic way and this could gradually contribute to Alzheimers disease pathology, as has been implicated in Downs syndrome patients.


Neurobiology of Aging | 2006

Alzheimer-associated APP+1 transgenic mice: Frameshift β-amyloid precursor protein is secreted in cerebrospinal fluid without inducing neuropathology

David F. Fischer; Elly M. Hol; Barbara Hobo; Fred W. van Leeuwen

Biomarkers present in the cerebrospinal fluid (CSF) of Alzheimer Disease patients could be instrumental in guiding diagnosis and monitoring of progression of the disease. We have previously reported on the secretion of a frameshifted form of amyloid-beta precursor protein, APP+1, into the CSF of Alzheimer patients and controls. APP+1 is secreted efficiently in controls, but during the progression of Alzheimer Disease, its secretion is reduced and APP+1 accumulates in tangle-bearing neurons. Here we describe the generation of a transgenic mouse line expressing APP+1 in the brain. These mice do not suffer from overt pathology or neurodegeneration, suggesting that APP+1 is not neurotoxic. To measure APP+1 levels in the CSF, we serially sampled CSF from the cisterna magna in the same mouse over a period of months. Indeed, APP+1 is secreted into the CSF of the transgenic mice, and APP+1 levels are stable over 1 year. This mouse model may guide the study of secretion deficits as found in Alzheimer Disease.

Collaboration


Dive into the Barbara Hobo's collaboration.

Top Co-Authors

Avatar

David F. Fischer

Netherlands Institute for Neuroscience

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Paula van Tijn

Royal Netherlands Academy of Arts and Sciences

View shared research outputs
Top Co-Authors

Avatar

Marian C. Verhage

Royal Netherlands Academy of Arts and Sciences

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jacqueline A. Sluijs

Royal Netherlands Academy of Arts and Sciences

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Marc A. F. Sonnemans

Netherlands Institute for Neuroscience

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge