Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
J Vis Exp ; (206)2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38738885

ABSTRACT

Adeno-associated viral vectors (AAVs) are a remarkable tool for investigating the central nervous system (CNS). Innovative capsids, such as AAV.PHP.eB, demonstrate extensive transduction of the CNS by intravenous injection in mice. To achieve comparable transduction, a 100-fold higher titer (minimally 1 x 1011 genome copies/mouse) is needed compared to direct injection in the CNS parenchyma. In our group, AAV production, including AAV.PHP.eB relies on adherent HEK293T cells and the triple transfection method. Achieving high yields of AAV with adherent cells entails a labor- and material-intensive process. This constraint prompted the development of a protocol for suspension-based cell culture in conical tubes. AAVs generated in adherent cells were compared to the suspension production method. Culture in suspension using transfection reagents Polyethylenimine or TransIt were compared. AAV vectors were purified by iodixanol gradient ultracentrifugation followed by buffer exchange and concentration using a centrifugal filter. With the adherent method, we achieved an average of 2.6 x 1012 genome copies (GC) total, whereas the suspension method and Polyethylenimine yielded 7.7 x 1012 GC in total, and TransIt yielded 2.4 x 1013 GC in total. There is no difference in in vivo transduction efficiency between vectors produced with adherent compared to the suspension cell system. In summary, a suspension HEK293 cell based AAV production protocol is introduced, resulting in a reduced amount of time and labor needed for vector production while achieving 3 to 9 times higher yields using components available from commercial vendors for research purposes.


Subject(s)
Dependovirus , Genetic Vectors , Humans , HEK293 Cells , Genetic Vectors/genetics , Dependovirus/genetics , Transfection/methods , Mice , Animals
2.
Alzheimers Res Ther ; 13(1): 45, 2021 02 17.
Article in English | MEDLINE | ID: mdl-33597019

ABSTRACT

BACKGROUND: Dysregulation of ceramide and sphingomyelin levels have been suggested to contribute to the pathogenesis of Alzheimer's disease (AD). Ceramide transfer proteins (CERTs) are ceramide carriers which are crucial for ceramide and sphingomyelin balance in cells. Extracellular forms of CERTs co-localize with amyloid-ß (Aß) plaques in AD brains. To date, the significance of these observations for the pathophysiology of AD remains uncertain. METHODS: A plasmid expressing CERTL, the long isoform of CERTs, was used to study the interaction of CERTL with amyloid precursor protein (APP) by co-immunoprecipitation and immunofluorescence in HEK cells. The recombinant CERTL protein was employed to study interaction of CERTL with amyloid-ß (Aß), Aß aggregation process in presence of CERTL, and the resulting changes in Aß toxicity in neuroblastoma cells. CERTL was overexpressed in neurons by adeno-associated virus (AAV) in a mouse model of familial AD (5xFAD). Ten weeks after transduction, animals were challenged with behavior tests for memory, anxiety, and locomotion. At week 12, brains were investigated for sphingolipid levels by mass spectrometry, plaques, and neuroinflammation by immunohistochemistry, gene expression, and/or immunoassay. RESULTS: Here, we report that CERTL binds to APP, modifies Aß aggregation, and reduces Aß neurotoxicity in vitro. Furthermore, we show that intracortical injection of AAV, mediating the expression of CERTL, decreases levels of ceramide d18:1/16:0 and increases sphingomyelin levels in the brain of male 5xFAD mice. CERTL in vivo over-expression has a mild effect on animal locomotion, decreases Aß formation, and modulates microglia by decreasing their pro-inflammatory phenotype. CONCLUSION: Our results demonstrate a crucial role of CERTL in regulating ceramide levels in the brain, in amyloid plaque formation and neuroinflammation, thereby opening research avenues for therapeutic targets of AD and other neurodegenerative diseases.


Subject(s)
Alzheimer Disease , Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/metabolism , Ceramides , Disease Models, Animal , Inflammation , Male , Mice , Mice, Transgenic , Plaque, Amyloid
3.
Gene Ther ; 28(1-2): 56-74, 2021 02.
Article in English | MEDLINE | ID: mdl-32576975

ABSTRACT

Adeno-associated viral vectors are widely used as vehicles for gene transfer to the nervous system. The promoter and viral vector serotype are two key factors that determine the expression dynamics of the transgene. A previous comparative study has demonstrated that AAV1 displays efficient transduction of layer V corticospinal neurons, but the optimal promoter for transgene expression in corticospinal neurons has not been determined yet. In this paper, we report a side-by-side comparison between four commonly used promoters: the short CMV early enhancer/chicken ß actin (sCAG), human cytomegalovirus (hCMV), mouse phosphoglycerate kinase (mPGK) and human synapsin (hSYN) promoter. Reporter constructs with each of these promoters were packaged in AAV1, and were injected in the sensorimotor cortex of rats and mice in order to transduce the corticospinal tract. Transgene expression levels and the cellular transduction profile were examined after 6 weeks. The AAV1 vectors harbouring the hCMV and sCAG promoters resulted in transgene expression in neurons, astrocytes and oligodendrocytes. The mPGK and hSYN promoters directed the strongest transgene expression. The mPGK promoter did drive expression in cortical neurons and oligodendrocytes, while transduction with AAV harbouring the hSYN promoter resulted in neuron-specific expression, including perineuronal net expressing interneurons and layer V corticospinal neurons. This promoter comparison study contributes to improve transgene delivery into the brain and spinal cord. The optimized transduction of the corticospinal tract will be beneficial for spinal cord injury research.


Subject(s)
Dependovirus , Pyramidal Tracts , Animals , Dependovirus/genetics , Genetic Vectors/genetics , Mice , Promoter Regions, Genetic , Rats , Transduction, Genetic , Transgenes
4.
Methods Mol Biol ; 1715: 3-17, 2018.
Article in English | MEDLINE | ID: mdl-29188502

ABSTRACT

Adeno-associated viral vectors have numerous applications in neuroscience, including the study of gene function in health and disease, targeting of light-sensitive proteins to anatomically distinct sets of neurons to manipulate neuronal activity (optogenetics), and the delivery of fluorescent protein to study anatomical connectivity in the brain. Moreover several phase I/II clinical trials for gene therapy of eye and brain diseases with adeno-associated viral vectors have shown that these vectors are well tolerated by human patients. In this chapter we describe a detailed protocol for the small scale production of recombinant adeno-associated viral vectors. This protocol can be executed by investigators with experience in cell culture and molecular biological techniques in any well-equipped molecular neurobiology laboratory. With this protocol we typically obtain research batches of 100-200 µL that range in titer from 5 × 1012 to 2 × 1013 genomic copies/mL.


Subject(s)
Brain Diseases/therapy , Dependovirus/genetics , Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors , Brain Diseases/genetics , Eye Diseases/genetics , Eye Diseases/therapy , HEK293 Cells , Humans , Injections, Intraocular/methods , Nervous System/metabolism , Plasmids
5.
J Neurosci ; 37(39): 9361-9379, 2017 09 27.
Article in English | MEDLINE | ID: mdl-28842419

ABSTRACT

Repulsive guidance molecule member a (RGMa) is a membrane-associated or released guidance molecule that is involved in axon guidance, cell patterning, and cell survival. In our previous work, we showed that RGMa is significantly upregulated in the substantia nigra of patients with Parkinson's disease. Here we demonstrate the expression of RGMa in midbrain human dopaminergic (DA) neurons. To investigate whether RGMa might model aspects of the neuropathology of Parkinson's disease in mouse, we targeted RGMa to adult midbrain dopaminergic neurons using adeno-associated viral vectors. Overexpression of RGMa resulted in a progressive movement disorder, including motor coordination and imbalance, which is typical for a loss of DA release in the striatum. In line with this, RGMa induced selective degeneration of dopaminergic neurons in the substantia nigra (SN) and affected the integrity of the nigrostriatal system. The degeneration of dopaminergic neurons was accompanied by a strong microglia and astrocyte activation. The behavioral, molecular, and anatomical changes induced by RGMa in mice are remarkably similar to the clinical and neuropathological hallmarks of Parkinson's disease. Our data indicate that dysregulation of RGMa plays an important role in the pathology of Parkinson's disease, and antibody-mediated functional interference with RGMa may be a disease modifying treatment option.SIGNIFICANCE STATEMENT Parkinson's disease (PD) is a neurodegenerative disease characterized by severe motor dysfunction due to progressive degeneration of mesencephalic dopaminergic (DA) neurons in the substantia nigra. To date, there is no regenerative treatment available. We previously showed that repulsive guidance molecule member a (RGMa) is upregulated in the substantia nigra of PD patients. Adeno-associated virus-mediated targeting of RGMa to mouse DA neurons showed that overexpression of this repulsive axon guidance and cell patterning cue models the behavioral and neuropathological characteristics of PD in a remarkable way. These findings have implications for therapy development as interfering with the function of this specific axon guidance cue may be beneficial to the survival of DA neurons.


Subject(s)
Nerve Tissue Proteins/genetics , Parkinson Disease/metabolism , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Female , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Microglia/metabolism , Microglia/pathology , Middle Aged , Nerve Tissue Proteins/metabolism , Parkinson Disease/genetics , Parkinson Disease/pathology , Parkinson Disease/physiopathology , Substantia Nigra/metabolism , Substantia Nigra/pathology
6.
PLoS One ; 12(1): e0170314, 2017.
Article in English | MEDLINE | ID: mdl-28103314

ABSTRACT

Terminal Schwann cells (TSCs) are specialized cells that envelop the motor nerve terminal, and play a role in the maintenance and regeneration of neuromuscular junctions (NMJs). The chemorepulsive protein semaphorin 3A (SEMA3A) is selectively up-regulated in TSCs on fast-fatigable muscle fibers following experimental denervation of the muscle (BotoxA-induced paralysis or crush injury to the sciatic nerve) or in the motor neuron disease amyotrophic lateral sclerosis (ALS). Re-expression of SEMA3A in this subset of TSCs is thought to play a role in the selective plasticity of nerve terminals as observed in ALS and following BotoxA-induced paralysis. Using a mouse model expressing a mutant SEMA3A with diminished signaling capacity, we studied the influence of SEMA3A signaling at the NMJ with two denervation paradigms; a motor neuron disease model (the G93A-hSOD1 ALS mouse line) and an injury model (BotoxA-induced paralysis). ALS mice that either expressed 1 or 2 mutant SEMA3A alleles demonstrated no difference in ALS-induced decline in motor behavior. We also investigated the effects of BotoxA-induced paralysis on the sprouting capacity of NMJs in the K108N-SEMA3A mutant mouse, and observed no change in the differential neuronal plasticity found at NMJs on fast-fatigable or slow muscle fibers due to the presence of the SEMA3A mutant protein. Our data may be explained by the residual repulsive activity of the mutant SEMA3A, or it may imply that SEMA3A alone is not a key component of the molecular signature affecting NMJ plasticity in ALS or BotoxA-induced paralysis. Interestingly, we did observe a sex difference in motor neuron sprouting behavior after BotoxA-induced paralysis in WT mice which we speculate may be an important factor in the sex dimorphic differences seen in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Neuromuscular Junction/genetics , Paralysis/genetics , Semaphorin-3A/genetics , Semaphorin-3A/metabolism , Amino Acid Substitution , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Botulinum Toxins, Type A/toxicity , Denervation , Disease Models, Animal , Female , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Transgenic , Motor Neurons/drug effects , Motor Neurons/pathology , Motor Neurons/physiology , Mutant Proteins/genetics , Mutant Proteins/metabolism , Neuromuscular Junction/drug effects , Neuromuscular Junction/physiology , Neuronal Plasticity/drug effects , Neuronal Plasticity/genetics , Neuronal Plasticity/physiology , Paralysis/chemically induced , Paralysis/physiopathology , Schwann Cells/physiology , Signal Transduction
7.
PLoS Biol ; 12(3): e1001808, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24618750

ABSTRACT

Neuropilin-1 (Nrp1) guides the development of the nervous and vascular systems, but its role in the mature brain remains to be explored. Here we report that the expression of the 65 kDa isoform of Sema3A, the ligand of Nrp1, by adult vascular endothelial cells, is regulated during the ovarian cycle and promotes axonal sprouting in hypothalamic neurons secreting gonadotropin-releasing hormone (GnRH), the neuropeptide controlling reproduction. Both the inhibition of Sema3A/Nrp1 signaling and the conditional deletion of Nrp1 in GnRH neurons counteract Sema3A-induced axonal sprouting. Furthermore, the localized intracerebral infusion of Nrp1- or Sema3A-neutralizing antibodies in vivo disrupts the ovarian cycle. Finally, the selective neutralization of endothelial-cell Sema3A signaling in adult Sema3aloxP/loxP mice by the intravenous injection of the recombinant TAT-Cre protein alters the amplitude of the preovulatory luteinizing hormone surge, likely by perturbing GnRH release into the hypothalamo-hypophyseal portal system. Our results identify a previously unknown function for 65 kDa Sema3A-Nrp1 signaling in the induction of axonal growth, and raise the possibility that endothelial cells actively participate in synaptic plasticity in specific functional domains of the adult central nervous system, thus controlling key physiological functions such as reproduction.


Subject(s)
Brain/metabolism , Endothelial Cells/metabolism , Fertility/physiology , Neuropilin-1/physiology , Semaphorin-3A/metabolism , Animals , Axons/metabolism , Axons/ultrastructure , Estrous Cycle/metabolism , Gonadotropin-Releasing Hormone/metabolism , Gonadotropin-Releasing Hormone/physiology , Ligands , Luteinizing Hormone/metabolism , Mice , Mice, Inbred C57BL , Neuropilin-1/metabolism , Rats , Rats, Sprague-Dawley , Semaphorin-3A/genetics , Semaphorin-3A/physiology , Signal Transduction
8.
Neurochem Int ; 61(5): 739-48, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22797007

ABSTRACT

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 Alzheimer's disease (AD). A role for the UPS has been suggested in the generation of amyloid ß (Aß) 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 Aß levels at 3, 6, 9 and 11 months of age. Surprisingly, we found a significant decrease in Aß deposition in amyloid plaques and levels of soluble Aß(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 Aß 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 Aß reduction. The molecular mechanism underlying this decrease in Aß 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).


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Disease Models, Animal , Mutation , Plaque, Amyloid/metabolism , Ubiquitin/genetics , Alzheimer Disease/etiology , Alzheimer Disease/genetics , Amyloid beta-Peptides/genetics , Animals , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Plaque, Amyloid/genetics , Plaque, Amyloid/pathology , Ubiquitin/physiology
9.
Physiol Behav ; 102(2): 193-200, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21059367

ABSTRACT

UBB(+1) is a mutant ubiquitin which accumulates in the hallmarks of tauopathies, including Alzheimer's 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.


Subject(s)
Memory Disorders/genetics , Mutation/genetics , Spatial Behavior/physiology , Ubiquitin/genetics , Age Factors , Analysis of Variance , Animals , Disease Models, Animal , Gene Expression Regulation/genetics , Humans , Male , Maze Learning/physiology , Memory Disorders/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/genetics , Neurologic Examination , Prosencephalon/metabolism , Proteasome Endopeptidase Complex/metabolism , Psychomotor Performance/physiology , Rotarod Performance Test , Ubiquitin/metabolism
10.
J Neurosci Res ; 88(11): 2325-37, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20336771

ABSTRACT

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 Alzheimer's 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.


Subject(s)
Proteasome Endopeptidase Complex/metabolism , Ubiquitin/genetics , Ubiquitin/metabolism , Acetylcysteine/analogs & derivatives , Acetylcysteine/pharmacology , Aging/physiology , Animals , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Cell Line , Immunohistochemistry , Mice , Mice, Transgenic , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Radioimmunoassay , Reverse Transcriptase Polymerase Chain Reaction , Serine Proteinase Inhibitors/pharmacology
11.
Mol Cell Neurosci ; 43(3): 281-6, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20005957

ABSTRACT

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.


Subject(s)
Disease Models, Animal , Huntington Disease/pathology , Proteasome Inhibitors , Ubiquitin/metabolism , Animals , Cell Death , Huntingtin Protein , Huntington Disease/metabolism , Male , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Peptides/toxicity , Ubiquitin/genetics
12.
Neurobiol Aging ; 30(6): 847-63, 2009 Jun.
Article in English | MEDLINE | ID: mdl-18760506

ABSTRACT

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 Alzheimer's disease (AD) and polyglutamine diseases such as Huntington's 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.


Subject(s)
Aging/metabolism , Alzheimer Disease/metabolism , Brain/metabolism , Disease Models, Animal , Proteasome Endopeptidase Complex/metabolism , Proteome/metabolism , Ubiquitin/metabolism , Animals , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Ubiquitin/genetics
13.
Proteomics ; 8(6): 1221-36, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18283660

ABSTRACT

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 alpha and beta resulted in a distinct nuclear and/or cytoplasmic staining of the entorhinal-hippocampal area and the temporal cortex of Alzheimer's 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 (Pick's 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.


Subject(s)
Proteasome Endopeptidase Complex/metabolism , Proteomics/methods , Synucleins/metabolism , Tauopathies/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Antibodies/immunology , Gene Expression , Humans , Immunohistochemistry , Lewy Body Disease/metabolism , Lewy Body Disease/pathology , Models, Biological , Multiple System Atrophy/metabolism , Multiple System Atrophy/pathology , Pick Disease of the Brain/metabolism , Pick Disease of the Brain/pathology , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/immunology , Protein Subunits/genetics , Protein Subunits/immunology , Protein Subunits/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tauopathies/pathology , Temporal Lobe/metabolism , Temporal Lobe/pathology
14.
Neurobiol Aging ; 27(10): 1445-50, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16214267

ABSTRACT

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.


Subject(s)
Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/pathology , Blood Proteins/cerebrospinal fluid , Blood Proteins/genetics , Brain/pathology , Disease Models, Animal , Neurons/metabolism , Poly(A)-Binding Proteins/cerebrospinal fluid , Poly(A)-Binding Proteins/genetics , Animals , Frameshift Mutation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Tissue Distribution
15.
Hum Mol Genet ; 13(16): 1803-13, 2004 Aug 15.
Article in English | MEDLINE | ID: mdl-15198995

ABSTRACT

Polyglutamine diseases are characterized by neuronal intranuclear inclusions (NIIs) of expanded polyglutamine proteins, indicating the failure of protein degradation. UBB(+1), an aberrant form of ubiquitin, is a substrate and inhibitor of the proteasome, and was previously reported to accumulate in Alzheimer disease and other tauopathies. Here, we show accumulation of UBB(+1) in the NIIs and the cytoplasm of neurons in Huntington disease and spinocerebellar ataxia type-3, indicating inhibition of the proteasome by polyglutamine proteins in human brain. We found that UBB(+1) not only increased aggregate formation of expanded polyglutamines in neuronally differentiated cell lines, but also had a synergistic effect on apoptotic cell death due to expanded polyglutamine proteins. These findings implicate UBB(+1) as an aggravating factor in polyglutamine-induced neurodegeneration, and clearly identify an important role for the ubiquitin-proteasome system in polyglutamine diseases.


Subject(s)
Apoptosis/physiology , Brain/metabolism , Heredodegenerative Disorders, Nervous System/metabolism , Inclusion Bodies/metabolism , Peptides/metabolism , Ubiquitin/metabolism , Blotting, Western , Cell Survival , Cloning, Molecular , DNA, Complementary/genetics , Fluorescent Antibody Technique , Heredodegenerative Disorders, Nervous System/physiopathology , Humans , Immunohistochemistry , Plasmids/genetics , Transfection , Tumor Cells, Cultured
16.
FASEB J ; 17(14): 2014-24, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14597671

ABSTRACT

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.


Subject(s)
Brain/enzymology , Cysteine Endopeptidases/metabolism , Multienzyme Complexes/metabolism , Neurodegenerative Diseases/enzymology , Ubiquitin/metabolism , Ubiquitins/metabolism , Antibody Specificity , Biomarkers/analysis , Brain/metabolism , Hippocampus/enzymology , Humans , Lewy Body Disease/metabolism , Lewy Body Disease/pathology , Multiple System Atrophy/metabolism , Multiple System Atrophy/pathology , Mutation , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Neurons/pathology , Proteasome Endopeptidase Complex , RNA, Messenger/genetics , Sequence Deletion , Tauopathies/genetics , Tauopathies/metabolism , Tauopathies/pathology , Ubiquitin/genetics , Ubiquitin/immunology , Ubiquitins/genetics , Ubiquitins/immunology
17.
J Biol Chem ; 278(41): 39637-43, 2003 Oct 10.
Article in English | MEDLINE | ID: mdl-12900421

ABSTRACT

Molecular misreading of the beta-amyloid precursor protein (APP) gene generates mRNA with dinucleotide deletions in GAGAG motifs. The resulting truncated and partly frameshifted APP protein (APP+1) accumulates in the dystrophic neurites and the neurofibrillary tangles in the cortex and hippocampus of Alzheimer patients. In contrast, we show here that neuronal cells transfected with APP+1 proficiently secreted APP+1. Because various secretory APP isoforms are present in cerebrospinal fluid (CSF), this study aimed to determine whether APP+1 is also a secretory protein that can be detected in CSF. Post-mortem CSF was obtained at autopsy from 50 non-demented controls and 122 Alzheimer patients; all subjects were staged for neuropathology (Braak score). Unexpectedly, we found that the APP+1 level in the CSF of non-demented controls was much higher (1.75 ng/ml) than in the CSF of Alzheimer patients (0.51 ng/ml) (p < 0.001), and the level of APP+1 in CSF was inversely correlated with the severity of the neuropathology. Moreover the earliest neuropathological changes are already reflected in a significant decrease of the APP+1 level in CSF. These data show that APP+1 is normally secreted by neurons, preventing intra-neuronal accumulation of APP+1 in brains of non-demented controls without neurofibrillary pathology.


Subject(s)
Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/cerebrospinal fluid , Amyloid beta-Protein Precursor/genetics , Frameshift Mutation , Aged , Alzheimer Disease/pathology , Amino Acid Sequence , Amyloid beta-Protein Precursor/metabolism , Blotting, Western , Brain/pathology , Case-Control Studies , Cell Line , Female , Humans , In Vitro Techniques , Male , Middle Aged , Molecular Sequence Data , Neurons/metabolism , Radioimmunoassay , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...