Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 12 de 12
2.
Mov Disord ; 38(8): 1428-1442, 2023 08.
Article En | MEDLINE | ID: mdl-37278528

BACKGROUND: Spinocerebellar ataxia type 1 (SCA1) is a neurodegenerative disease caused by a polyglutamine expansion in the ataxin-1 protein resulting in neuropathology including mutant ataxin-1 protein aggregation, aberrant neurodevelopment, and mitochondrial dysfunction. OBJECTIVES: Identify SCA1-relevant phenotypes in patient-specific fibroblasts and SCA1 induced pluripotent stem cells (iPSCs) neuronal cultures. METHODS: SCA1 iPSCs were generated and differentiated into neuronal cultures. Protein aggregation and neuronal morphology were evaluated using fluorescent microscopy. Mitochondrial respiration was measured using the Seahorse Analyzer. The multi-electrode array (MEA) was used to identify network activity. Finally, gene expression changes were studied using RNA-seq to identify disease-specific mechanisms. RESULTS: Bioenergetics deficits in patient-derived fibroblasts and SCA1 neuronal cultures showed altered oxygen consumption rate, suggesting involvement of mitochondrial dysfunction in SCA1. In SCA1 hiPSC-derived neuronal cells, nuclear and cytoplasmic aggregates were identified similar in localization as aggregates in SCA1 postmortem brain tissue. SCA1 hiPSC-derived neuronal cells showed reduced dendrite length and number of branching points while MEA recordings identified delayed development in network activity in SCA1 hiPSC-derived neuronal cells. Transcriptome analysis identified 1050 differentially expressed genes in SCA1 hiPSC-derived neuronal cells associated with synapse organization and neuron projection guidance, where a subgroup of 151 genes was highly associated with SCA1 phenotypes and linked to SCA1 relevant signaling pathways. CONCLUSIONS: Patient-derived cells recapitulate key pathological features of SCA1 pathogenesis providing a valuable tool for the identification of novel disease-specific processes. This model can be used for high throughput screenings to identify compounds, which may prevent or rescue neurodegeneration in this devastating disease. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Induced Pluripotent Stem Cells , Spinocerebellar Ataxias , Mice , Animals , Ataxins/metabolism , Protein Aggregates , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Mice, Transgenic , Purkinje Cells/metabolism , Purkinje Cells/pathology , Spinocerebellar Ataxias/metabolism , Fibroblasts/metabolism
4.
Front Oncol ; 13: 1218297, 2023.
Article En | MEDLINE | ID: mdl-38260852

Background: Only a small group of patients with glioblastoma multiforme (GBM) survives more than 36 months, so-called long-term survivors. Recent studies have shown that chromosomal instability (CIN) plays a prognostic and predictive role among different cancer types. Here, we compared histological (chromosome missegregation) and bioinformatic metrics (CIN signatures) of CIN in tumors of GBM typical survivors (≤36 months overall survival), GBM long-term survivors and isocitrate dehydrogenase (IDH)-mutant grade 4 astrocytomas. Methods: Tumor sections of all gliomas were examined for anaphases and chromosome missegregation. Further CIN signature activity analysis in the The Cancer Genome Atlas (TCGA)-GBM cohort was performed. Results: Our data show that chromosome missegregation is pervasive in high grade gliomas and is not different between the 3 groups. We find only limited evidence of altered CIN levels in tumors of GBM long-term survivors relative to the other groups, since a significant depletion in CIN signature 11 relative to GBM typical survivors was the only alteration detected. In contrast, within IDH-mutant grade 4 astrocytomas we detected a significant enrichment of CIN signature 5 and 10 activities and a depletion of CIN signature 1 activity relative to tumors of GBM typical survivors. Conclusions: Our data suggest that CIN is pervasive in high grade gliomas, however this is unlikely to be a major contributor to the phenomenon of long-term survivorship in GBM. Nevertheless, further evaluation of specific types of CIN (signatures) could have prognostic value in patients suffering from grade 4 gliomas.

5.
Brain Pathol ; 32(4): e13044, 2022 07.
Article En | MEDLINE | ID: mdl-34913212

Increasing evidence supports the role of neurotropic herpes simplex virus 1 (HSV-1) in the pathogenesis of Alzheimer's disease (AD). However, it is unclear whether previously reported findings in HSV-1 cell culture and animal models can be translated to humans. Here, we analyzed clinical specimens from latently HSV-1 infected individuals and individuals with lytic HSV infection of the brain (herpes simplex encephalitis; HSE). Latent HSV-1 DNA load and latency-associated transcript (LAT) expression were identical between trigeminal ganglia (TG) of AD patients and controls. Amyloid ß (Aß) and hyperphosphorylated tau (pTau) were not detected in latently HSV-infected TG neurons. Aging-related intraneuronal Aß accumulations, neurofibrillary tangles (NFT), and/or extracellular Aß plaques were observed in the brain of some HSE patients, but these were neither restricted to HSV-infected neurons nor brain regions containing virus-infected cells. Analysis of unique brain material from an AD patient with concurrent HSE showed that HSV-infected cells frequently localized close to Aß plaques and NFT, but were not associated with exacerbated AD-related pathology. HSE-associated neuroinflammation was not associated with specific Aß or pTau phenotypes. Collectively, we observed that neither latent nor lytic HSV infection of human neurons is directly associated with aberrant Aß or pTau protein expression in ganglia and brain.


Alzheimer Disease , Amyloid beta-Peptides/metabolism , Herpes Simplex , Herpesvirus 1, Human , tau Proteins/metabolism , Animals , Brain/metabolism , Herpes Simplex/metabolism , Herpesvirus 1, Human/metabolism , Humans , Neurons/metabolism , Plaque, Amyloid , Trigeminal Ganglion/metabolism
7.
J Vis Exp ; (171)2021 05 10.
Article En | MEDLINE | ID: mdl-34028453

The goal of head and neck oncological surgery is complete tumor resection with adequate resection margins while preserving acceptable function and appearance. For oral cavity squamous cell carcinoma (OCSCC), different studies showed that only 15%-26% of all resections are adequate. A major reason for the low number of adequate resections is the lack of information during surgery; the margin status is only available after the final histopathologic assessment, days after surgery. The surgeons and pathologists at the Erasmus MC University Medical Center in Rotterdam started the implementation of specimen-driven intraoperative assessment of resection margins (IOARM) in 2013, which became the standard of care in 2015. This method enables the surgeon to turn an inadequate resection into an adequate resection by performing an additional resection during the initial surgery. Intraoperative assessment is supported by a relocation method procedure that allows accurate identification of inadequate margins (found on the specimen) in the wound bed. The implementation of this protocol resulted in an improvement of adequate resections from 15%-40%. However, the specimen-driven IOARM is not widely adopted because grossing fresh tissue is counter-intuitive for pathologists. The fear exists that grossing fresh tissue will deteriorate the anatomical orientation, shape, and size of the specimen and therefore will affect the final histopathologic assessment. These possible negative effects are countered by the described protocol. Here, the protocol for specimen-driven IOARM is presented in detail, as performed at the institute.


Mouth Neoplasms , Carcinoma, Squamous Cell/surgery , Humans , Intraoperative Care , Margins of Excision , Mouth Neoplasms/surgery
8.
Dev Neurobiol ; 74(12): 1226-42, 2014 Dec.
Article En | MEDLINE | ID: mdl-24909416

The hippocampus is one of the two areas in the mammalian brain where adult neurogenesis occurs. Adult neurogenesis is well known to be involved in hippocampal physiological functions as well as pathophysiological conditions. Microtubules (MTs), providing intracellular transport, stability, and transmitting force, are indispensable for neurogenesis by facilitating cell division, migration, growth, and differentiation. Although there are several examples of MT-stabilizing proteins regulating different aspects of adult neurogenesis, relatively little is known about the function of MT-destabilizing proteins. Stathmin is such a MT-destabilizing protein largely restricted to the CNS, and in contrast to its developmental family members, stathmin is also expressed at significant levels in the adult brain, notably in areas involved in adult neurogenesis. Here, we show an important role for stathmin during adult neurogenesis in the subgranular zone of the mouse hippocampus. After carefully mapping stathmin expression in the adult dentate gyrus (DG), we investigated its role in hippocampal neurogenesis making use of stathmin knockout mice. Although hippocampus development appears normal in these animals, different aspects of adult neurogenesis are affected. First, the number of proliferating Ki-67+ cells is decreased in stathmin knockout mice, as well as the expression of the immature markers Nestin and PSA-NCAM. However, newborn cells that do survive express more frequently the adult marker NeuN and have a more mature morphology. Furthermore, our data suggest that migration in the DG might be affected. We propose a model in which stathmin controls the transition from neuronal precursors to early postmitotic neurons.


Hippocampus/physiology , Neural Stem Cells/physiology , Neurogenesis/physiology , Neurons/physiology , Stathmin/metabolism , Animals , Cell Movement/physiology , Cell Survival/physiology , DNA-Binding Proteins , Hippocampus/cytology , Ki-67 Antigen/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/metabolism , Nestin/metabolism , Neural Cell Adhesion Molecule L1/metabolism , Neural Stem Cells/cytology , Neurons/cytology , Nuclear Proteins/metabolism , Sialic Acids/metabolism , Stathmin/genetics
9.
Acta Neuropathol Commun ; 2: 38, 2014 Apr 07.
Article En | MEDLINE | ID: mdl-24708899

BACKGROUND: Fragmentation of stacked cisterns of the Golgi apparatus into dispersed smaller elements is a feature associated with degeneration of neurons in amyotrophic lateral sclerosis (ALS) and some other neurodegenerative disorders. However, the role of Golgi fragmentation in motor neuron degeneration is not well understood. RESULTS: Here we use a SOD1-ALS mouse model (low-copy Gurney G93A-SOD1 mouse) to show that motor neurons with Golgi fragmentation are retrogradely labeled by intramuscularly injected CTB (beta subunit of cholera toxin), indicating that Golgi fragmentation precedes neuromuscular denervation and axon retraction. We further show that Golgi fragmentation may occur in the absence of and precede two other pathological markers, i.e. somatodendritic SOD1 inclusions, and the induction of ATF3 expression. In addition, we show that Golgi fragmentation is associated with an altered dendritic organization of the Golgi apparatus, does not depend on intact apoptotic machinery, and is facilitated in transgenic mice with impaired retrograde dynein-dependent transport (BICD2-N mice). A connection to altered dynein-dependent transport also is suggested by reduced expression of endosomal markers in neurons with Golgi fragmentation, which also occurs in neurons with impaired dynein function. CONCLUSIONS: Together the data indicate that Golgi fragmentation is a very early event in the pathological cascade in ALS that is associated with altered organization of intracellular trafficking.


Amyotrophic Lateral Sclerosis/complications , Endosomes/pathology , Golgi Apparatus/pathology , Motor Neurons/ultrastructure , Neuromuscular Diseases/etiology , Neuromuscular Diseases/pathology , Activating Transcription Factor 3/metabolism , Amyotrophic Lateral Sclerosis/genetics , Animals , Calcitonin Gene-Related Peptide/metabolism , Carrier Proteins/metabolism , Cells, Cultured , Cholera Toxin/metabolism , Choline O-Acetyltransferase/metabolism , Disease Models, Animal , Humans , Intracellular Signaling Peptides and Proteins , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Motor Neurons/pathology , Nerve Tissue Proteins/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Vesicular Transport Proteins/metabolism
10.
Acta Neuropathol Commun ; 1: 24, 2013 Jun 12.
Article En | MEDLINE | ID: mdl-24252306

BACKGROUND: Protein aggregation and the formation of intracellular inclusions are a central feature of many neurodegenerative disorders, but precise knowledge about their pathogenic role is lacking in most instances. Here we have characterized inclusions formed in transgenic mice carrying the P56S mutant form of VAPB that causes various motor neuron syndromes including ALS8. RESULTS: Inclusions in motor neurons of VAPB-P56S transgenic mice are characterized by the presence of smooth ER-like tubular profiles, and are immunoreactive for factors that operate in the ER associated degradation (ERAD) pathway, including p97/VCP, Derlin-1, and the ER membrane chaperone BAP31. The presence of these inclusions does not correlate with signs of axonal and neuronal degeneration, and axotomy leads to their gradual disappearance, indicating that they represent reversible structures. Inhibition of the proteasome and knockdown of the ER membrane chaperone BAP31 increased the size of mutant VAPB inclusions in primary neuron cultures, while knockdown of TEB4, an ERAD ubiquitin-protein ligase, reduced their size. Mutant VAPB did not codistribute with mutant forms of seipin that are associated with an autosomal dominant motor neuron disease, and accumulate in a protective ER derived compartment termed ERPO (ER protective organelle) in neurons. CONCLUSIONS: The data indicate that the VAPB-P56S inclusions represent a novel reversible ER quality control compartment that is formed when the amount of mutant VAPB exceeds the capacity of the ERAD pathway and that isolates misfolded and aggregated VAPB from the rest of the ER. The presence of this quality control compartment reveals an additional level of flexibility of neurons to cope with misfolded protein stress in the ER.


Amyotrophic Lateral Sclerosis/physiopathology , Endoplasmic Reticulum/physiology , Inclusion Bodies/physiology , Motor Neurons/physiology , Vesicular Transport Proteins/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Axons/physiology , Axons/ultrastructure , Cells, Cultured , Disease Models, Animal , Endoplasmic Reticulum/ultrastructure , Endoplasmic Reticulum-Associated Degradation/physiology , Gene Knockdown Techniques , Hippocampus/physiopathology , Hippocampus/ultrastructure , Inclusion Bodies/ultrastructure , Mice, Transgenic , Motor Neurons/ultrastructure , Mutation , Rats , Sciatic Nerve/injuries , Sciatic Nerve/physiopathology , Sciatic Nerve/ultrastructure , Vesicular Transport Proteins/genetics
11.
J Neuropathol Exp Neurol ; 70(8): 662-77, 2011 Aug.
Article En | MEDLINE | ID: mdl-21760539

Motor neuron degeneration and skeletal muscle denervation are hallmarks of amyotrophic lateral sclerosis (ALS), but other neuron populations and glial cells are also involved in ALS pathogenesis. We examined changes in inhibitory interneurons in spinal cords of the ALS model low-copy Gurney G93A-SOD1 (G1del) mice and found reduced expression of markers of glycinergic and GABAergic neurons, that is, glycine transporter 2 (GlyT2) and glutamic acid decarboxylase (GAD65/67), specifically in the ventral horns of clinically affected mice. There was also loss of GlyT2 and GAD67 messenger RNA-labeled neurons in the intermediate zone. Ubiquitinated inclusions appeared in interneurons before 20 weeks of age, that is, after their development in motor neurons but before the onset of clinical signs and major motor neuron degeneration, which starts from 25 weeks of age. Because mutant superoxide dismutase 1 (SOD1) in glia might contribute to the pathogenesis, we also examined neuron-specific G93A-SOD1 mice; they also had loss of inhibitory interneuron markers in ventral horns and ubiquitinated interneuron inclusions. These data suggest that, in mutant SOD1-associated ALS, pathological changes may spread from motor neurons to interneuronsin a relatively early phase of the disease, independent of the presence of mutant SOD1 in glia. The degeneration of spinal inhibitory interneurons may in turn facilitate degeneration of motor neurons and contribute to disease progression.


Amyotrophic Lateral Sclerosis , Interneurons/pathology , Motor Neurons/pathology , Nerve Degeneration/etiology , Neuroglia/metabolism , Spinal Cord/pathology , Activating Transcription Factor 3/metabolism , Age Factors , Amyotrophic Lateral Sclerosis/complications , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Calbindins , Choline O-Acetyltransferase/metabolism , Disease Models, Animal , Galectin 3/metabolism , Gene Expression Regulation/genetics , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Glycine Plasma Membrane Transport Proteins/genetics , Glycine Plasma Membrane Transport Proteins/metabolism , Green Fluorescent Proteins/genetics , Humans , Interneurons/metabolism , Mice , Mice, Transgenic , Motor Neurons/metabolism , Mutation/genetics , Parvalbumins/metabolism , Proto-Oncogene Proteins c-jun/metabolism , RNA, Messenger/metabolism , S100 Calcium Binding Protein G/metabolism , Superoxide Dismutase/genetics , Ubiquitin/metabolism
12.
Hum Mol Genet ; 17(18): 2849-62, 2008 Sep 15.
Article En | MEDLINE | ID: mdl-18579581

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative condition characterized by progressive motor neuron degeneration and muscle paralysis. Genetic evidence from man and mouse has indicated that mutations in the dynein/dynactin motor complex are correlated with motor neuron degeneration. In this study, we have generated transgenic mice with neuron-specific expression of Bicaudal D2 N-terminus (BICD2-N) to chronically impair dynein/dynactin function. Motor neurons expressing BICD2-N showed accumulation of dynein and dynactin in the cell body, Golgi fragmentation and several signs of impaired retrograde trafficking: the appearance of giant neurofilament swellings in the proximal axon, reduced retrograde labelling by tracer injected in the muscle and delayed expression of the injury transcription factor ATF3 after axon transection. Despite these abnormalities, BICD2-N mice did not develop signs of motor neuron degeneration and motor abnormalities. Interestingly, the BICD2-N transgene increased lifespan in 'low copy' SOD1-G93A ALS transgenic mice. Our findings indicate that impaired dynein/dynactin function can explain several pathological features observed in ALS patients, but may be beneficial in some forms of ALS.


Amyotrophic Lateral Sclerosis/metabolism , Carrier Proteins/metabolism , Disease Models, Animal , Dyneins/metabolism , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Motor Neurons/metabolism , Superoxide Dismutase/metabolism , Amyotrophic Lateral Sclerosis/mortality , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Biological Transport , Carrier Proteins/genetics , Cells, Cultured , Dynactin Complex , Dyneins/genetics , Female , Gene Expression , Golgi Apparatus/metabolism , Humans , Life Expectancy , Male , Membrane Proteins/genetics , Mice , Mice, Transgenic , Microtubule-Associated Proteins/genetics , Motor Neurons/pathology , Motor Neurons/physiology , Rats , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , Survival
...