Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
1.
J Biol Chem ; 300(7): 107402, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38782207

ABSTRACT

Here, we describe pathological events potentially involved in the disease pathogenesis of Alexander disease (AxD). This is a primary genetic disorder of astrocyte caused by dominant gain-of-function mutations in the gene coding for an intermediate filament protein glial fibrillary acidic protein (GFAP). Pathologically, this disease is characterized by the upregulation of GFAP and its accumulation as Rosenthal fibers. Although the genetic basis linking GFAP mutations with Alexander disease has been firmly established, the initiating events that promote GFAP accumulation and the role of Rosenthal fibers (RFs) in the disease process remain unknown. Here, we investigate the hypothesis that disease-associated mutations promote GFAP aggregation through aberrant posttranslational modifications. We found high molecular weight GFAP species in the RFs of AxD brains, indicating abnormal GFAP crosslinking as a prominent pathological feature of this disease. In vitro and cell-based studies demonstrate that cystine-generating mutations promote GFAP crosslinking by cysteine-dependent oxidation, resulting in defective GFAP assembly and decreased filament solubility. Moreover, we found GFAP was ubiquitinated in RFs of AxD patients and rodent models, supporting this modification as a critical factor linked to GFAP aggregation. Finally, we found that arginine could increase the solubility of aggregation-prone mutant GFAP by decreasing its ubiquitination and aggregation. Our study suggests a series of pathogenic events leading to AxD, involving interplay between GFAP aggregation and abnormal modifications by GFAP ubiquitination and oxidation. More important, our findings provide a basis for investigating new strategies to treat AxD by targeting abnormal GFAP modifications.


Subject(s)
Alexander Disease , Glial Fibrillary Acidic Protein , Ubiquitination , Alexander Disease/metabolism , Alexander Disease/genetics , Alexander Disease/pathology , Glial Fibrillary Acidic Protein/metabolism , Glial Fibrillary Acidic Protein/genetics , Humans , Animals , Mutation , Mice , Astrocytes/metabolism , Astrocytes/pathology , Brain/metabolism , Brain/pathology , Protein Processing, Post-Translational , Rats , Male , Female , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/pathology
2.
Brain ; 147(2): 698-716, 2024 02 01.
Article in English | MEDLINE | ID: mdl-37955589

ABSTRACT

Alexander disease (AxD) is an intractable neurodegenerative disorder caused by GFAP mutations. It is a primary astrocyte disease with a pathological hallmark of Rosenthal fibres within astrocytes. AxD astrocytes show several abnormal phenotypes. Our previous study showed that AxD astrocytes in model mice exhibit aberrant Ca2+ signals that induce AxD aetiology. Here, we show that microglia have unique phenotypes with morphological and functional alterations, which are related to the pathogenesis of AxD. Immunohistochemical studies of 60TM mice (AxD model) showed that AxD microglia exhibited highly ramified morphology. Functional changes in microglia were assessed by Ca2+ imaging using hippocampal brain slices from Iba1-GCaMP6-60TM mice and two-photon microscopy. We found that AxD microglia showed aberrant Ca2+ signals, with high frequency Ca2+ signals in both the processes and cell bodies. These microglial Ca2+ signals were inhibited by pharmacological blockade or genetic knockdown of P2Y12 receptors but not by tetrodotoxin, indicating that these signals are independent of neuronal activity but dependent on extracellular ATP from non-neuronal cells. Our single-cell RNA sequencing data showed that the expression level of Entpd2, an astrocyte-specific gene encoding the ATP-degrading enzyme NTPDase2, was lower in AxD astrocytes than in wild-type astrocytes. In situ ATP imaging using the adeno-associated virus vector GfaABC1D ATP1.0 showed that exogenously applied ATP was present longer in 60TM mice than in wild-type mice. Thus, the increased ATP level caused by the decrease in its metabolizing enzyme in astrocytes could be responsible for the enhancement of microglial Ca2+ signals. To determine whether these P2Y12 receptor-mediated Ca2+ signals in AxD microglia play a significant role in the pathological mechanism, a P2Y12 receptor antagonist, clopidogrel, was administered. Clopidogrel significantly exacerbated pathological markers in AxD model mice and attenuated the morphological features of microglia, suggesting that microglia play a protective role against AxD pathology via P2Y12 receptors. Taken together, we demonstrated that microglia sense AxD astrocyte dysfunction via P2Y12 receptors as an increase in extracellular ATP and alter their morphology and Ca2+ signalling, thereby protecting against AxD pathology. Although AxD is a primary astrocyte disease, our study may facilitate understanding of the role of microglia as a disease modifier, which may contribute to the clinical diversity of AxD.


Subject(s)
Alexander Disease , Mice , Animals , Alexander Disease/metabolism , Alexander Disease/pathology , Glial Fibrillary Acidic Protein/metabolism , Astrocytes/metabolism , Microglia/metabolism , Clopidogrel/metabolism , Calcium/metabolism , Disease Progression , Adenosine Triphosphate/metabolism
3.
Cells ; 12(7)2023 03 23.
Article in English | MEDLINE | ID: mdl-37048051

ABSTRACT

Alexander disease (AxD) is caused by mutations in the gene for glial fibrillary acidic protein (GFAP), an intermediate filament expressed by astrocytes in the central nervous system. AxD-associated mutations cause GFAP aggregation and astrogliosis, and GFAP is elevated with the astrocyte stress response, exacerbating mutant protein toxicity. Studies in mouse models suggest disease severity is tied to Gfap expression levels, and signal transducer and activator of transcription (STAT)-3 regulates Gfap during astrocyte development and in response to injury and is activated in astrocytes in rodent models of AxD. In this report, we show that STAT3 is also activated in the human disease. To determine whether STAT3 contributes to GFAP elevation, we used a combination of genetic approaches to knockout or reduce STAT3 activation in AxD mouse models. Conditional knockout of Stat3 in cells expressing Gfap reduced Gfap transactivation and prevented protein accumulation. Astrocyte-specific Stat3 knockout in adult mice with existing pathology reversed GFAP accumulation and aggregation. Preventing STAT3 activation reduced markers of reactive astrocytes, stress-related transcripts, and microglial activation, regardless of disease stage or genetic knockout approach. These results suggest that pharmacological inhibition of STAT3 could potentially reduce GFAP toxicity and provide a therapeutic benefit in patients with AxD.


Subject(s)
Alexander Disease , Glial Fibrillary Acidic Protein , STAT3 Transcription Factor , Animals , Humans , Mice , Alexander Disease/genetics , Alexander Disease/metabolism , Alexander Disease/pathology , Astrocytes/metabolism , Disease Models, Animal , Glial Fibrillary Acidic Protein/metabolism , Intermediate Filaments/metabolism , Mutation , STAT3 Transcription Factor/metabolism
4.
J Hist Neurosci ; 32(4): 399-422, 2023.
Article in English | MEDLINE | ID: mdl-37000960

ABSTRACT

In 1949, William Stewart Alexander (1919-2013), a young pathologist from New Zealand working in London, reported the neuropathological findings in a 15-month-old boy who had developed normally until the age of seven months, but thereafter had progressive enlargement of his head and severe developmental delay. The most striking neuropathological abnormality was the presence of numerous Rosenthal fibers in the brain. The distribution of these fibers suggested to Alexander that the primary pathological change involved astrocytes. In the next 15 years, five similar patients were reported, and in 1964 Friede recognized these cases reflected a single disease process and coined the eponym "Alexander's disease" to describe the disorder. In the 1960s, electron microscopy confirmed that Rosenthal fibers were localized to astrocytes. In 2001, it was shown that Alexander disease is caused by mutations in the gene encoding glial fibrillary acidic protein, the major intermediate filament protein in astrocytes. Although the clinical, imaging, and pathological manifestations of Alexander disease are now well known, few people are familiar with Alexander's career. Although he did not make a further contribution to the literature on Alexander disease, his observations and accurate interpretation of the neuropathology have justified the continued use of the eponym "Alexander disease."


Subject(s)
Alexander Disease , Male , Humans , Infant , Alexander Disease/genetics , Alexander Disease/metabolism , Alexander Disease/pathology , Eponyms , Brain/pathology , Mutation , Astrocytes/metabolism , Astrocytes/pathology
5.
Mol Biol Cell ; 33(8): ar69, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35511821

ABSTRACT

Alexander disease is a primary genetic disorder of astrocytes caused by dominant mutations in the gene encoding glial fibrillary acidic protein (GFAP). How single-amino-acid changes can lead to cytoskeletal catastrophe and brain degeneration remains poorly understood. In this study, we have analyzed 14 missense mutations located in the GFAP rod domain to investigate how these mutations affect in vitro filament assembly. Whereas the internal rod mutants assembled into filaments that were shorter than those of wild type, the rod end mutants formed structures with one or more of several atypical characteristics, including short filament length, irregular width, roughness of filament surface, and filament aggregation. When transduced into primary astrocytes, GFAP mutants with in vitro assembly defects usually formed cytoplasmic aggregates, which were more resistant to biochemical extraction. The resistance of GFAP to solubilization was also observed in brain tissues of patients with Alexander disease, in which a significant proportion of insoluble GFAP were accumulated in Rosenthal fiber fractions. These findings provide clinically relevant evidence that link GFAP assembly defects to disease pathology at the tissue level and suggest that altered filament assembly and properties as a result of GFAP mutation are critical initiating factors for the pathogenesis of Alexander disease.


Subject(s)
Alexander Disease , Glial Fibrillary Acidic Protein/metabolism , Alexander Disease/genetics , Alexander Disease/metabolism , Astrocytes/metabolism , Glial Fibrillary Acidic Protein/genetics , Humans , Intermediate Filaments/metabolism , Mutation/genetics
6.
Curr Opin Neurobiol ; 72: 140-147, 2022 02.
Article in English | MEDLINE | ID: mdl-34826654

ABSTRACT

Alexander disease is a primary disorder of astrocytes caused by gain-of-function mutations in the gene for glial fibrillary acidic protein (GFAP), which lead to protein aggregation and a reactive astrocyte response, with devastating effects on the central nervous system. Over the past two decades since the discovery of GFAP as the culprit, several cellular and animal models have been generated, and much has been learned about underlying mechanisms contributing to the disease. Despite these efforts, many aspects of Alexander disease have remained enigmatic, particularly the initiating events in GFAP accumulation and astrocyte pathology, the relation between astrocyte dysfunction and myelin deficits, and the variability in age of onset and disease severity. More recent work in both old and new models has begun to address these complex questions and identify new therapeutics that finally offer the promise of effective treatment.


Subject(s)
Alexander Disease , Alexander Disease/genetics , Alexander Disease/metabolism , Alexander Disease/pathology , Animals , Astrocytes/metabolism , Central Nervous System/pathology , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Humans , Mutation , Protein Aggregates
7.
Mol Cell Proteomics ; 21(1): 100180, 2022 01.
Article in English | MEDLINE | ID: mdl-34808356

ABSTRACT

Alexander disease (AxD) is a rare and fatal neurodegenerative disorder caused by mutations in the gene encoding glial fibrillary acidic protein (GFAP). In this report, a mouse model of AxD (GFAPTg;Gfap+/R236H) was analyzed that contains a heterozygous R236H point mutation in murine Gfap as well as a transgene with a GFAP promoter to overexpress human GFAP. Using label-free quantitative proteomic comparisons of brain tissue from GFAPTg;Gfap+/R236H versus wild-type mice confirmed upregulation of the glutathione metabolism pathway and indicated proteins were elevated in the peroxisome proliferator-activated receptor (PPAR) signaling pathway, which had not been reported previously in AxD. Relative protein-level differences were confirmed by a targeted proteomics assay, including proteins related to astrocytes and oligodendrocytes. Of particular interest was the decreased level of the oligodendrocyte protein, 2-hydroxyacylsphingosine 1-beta-galactosyltransferase (Ugt8), since Ugt8-deficient mice exhibit a phenotype similar to GFAPTg;Gfap+/R236H mice (e.g., tremors, ataxia, hind-limb paralysis). In addition, decreased levels of myelin-associated proteins were found in the GFAPTg;Gfap+/R236H mice, consistent with the role of Ugt8 in myelin synthesis. Fabp7 upregulation in GFAPTg;Gfap+/R236H mice was also selected for further investigation due to its uncharacterized association to AxD, critical function in astrocyte proliferation, and functional ability to inhibit the anti-inflammatory PPAR signaling pathway in models of amyotrophic lateral sclerosis (ALS). Within Gfap+ astrocytes, Fabp7 was markedly increased in the hippocampus, a brain region subjected to extensive pathology and chronic reactive gliosis in GFAPTg;Gfap+/R236H mice. Last, to determine whether the findings in GFAPTg;Gfap+/R236H mice are present in the human condition, AxD patient and control samples were analyzed by Western blot, which indicated that Type I AxD patients have a significant fourfold upregulation of FABP7. However, immunohistochemistry analysis showed that UGT8 accumulates in AxD patient subpial brain regions where abundant amounts of Rosenthal fibers are located, which was not observed in the GFAPTg;Gfap+/R236H mice.


Subject(s)
Alexander Disease , Alexander Disease/genetics , Alexander Disease/metabolism , Alexander Disease/pathology , Animals , Astrocytes/metabolism , Disease Models, Animal , Gliosis/metabolism , Gliosis/pathology , Humans , Mice , Mice, Transgenic , Mutation , Proteomics
8.
Sci Transl Med ; 13(620): eabg4711, 2021 11 17.
Article in English | MEDLINE | ID: mdl-34788075

ABSTRACT

Alexander disease (AxD) is a devastating leukodystrophy caused by gain-of-function mutations in GFAP, and the only available treatments are supportive. Recent advances in antisense oligonucleotide (ASO) therapy have demonstrated that transcript targeting can be a successful strategy for human neurodegenerative diseases amenable to this approach. We have previously used mouse models of AxD to show that Gfap-targeted ASO suppresses protein accumulation and reverses pathology; however, the mice have a mild phenotype with no apparent leukodystrophy or overt clinical features and are therefore limited for assessing functional outcomes. In this report, we introduce a rat model of AxD that exhibits hallmark pathology with GFAP aggregation in the form of Rosenthal fibers, widespread astrogliosis, and white matter deficits. These animals develop normally during the first postnatal weeks but fail to thrive after weaning and develop severe motor deficits as they mature, with about 14% dying of unknown cause between 6 and 12 weeks of age. In this model, a single treatment with Gfap-targeted ASO provides long-lasting suppression, reverses GFAP pathology, and, depending on age of treatment, prevents or mitigates white matter deficits and motor impairment. In this report, we characterize an improved animal model of AxD with myelin pathology and motor impairment, recapitulating prominent features of the human disease, and use this model to show that ASO therapy has the potential to not only prevent but also reverse many aspects of disease.


Subject(s)
Alexander Disease , Glial Fibrillary Acidic Protein , Motor Disorders , White Matter , Alexander Disease/genetics , Alexander Disease/metabolism , Alexander Disease/pathology , Animals , Astrocytes/metabolism , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Gliosis/pathology , Motor Disorders/metabolism , Motor Disorders/pathology , Mutation/genetics , Rats , White Matter/pathology
9.
Mol Med Rep ; 24(2)2021 Aug.
Article in English | MEDLINE | ID: mdl-34109421

ABSTRACT

Alexander disease (AxD) is a cerebral white matter disease affecting a wide range of ages, from infants to adults. In the present study, two cases of bulbospinal form AxD were reported, and a preliminary exploration of AxD was conducted thorough clinical, functional magnetic resonance imaging (fMRI) and functional analyses. In total, two de novo mutations in the glial fibrillary acidic protein (GFAP) gene (c.214G>A and c.1235C>T) were identified in unrelated patients (one in each patient). Both patients showed increased regional neural activity and functional connectivity in the cerebellum and posterior parietal cortex according to fMRI analysis. Notably, grey matter atrophy was discovered in the patient with c.214G>A variant. Functional experiments revealed aberrant accumulation of mutant GFAP and decreased solubility of c.1235C>T variant. Under pathological conditions, autophagic flux was activated for GFAP aggregate degradation. Moreover, transcriptional data of AxD and healthy human brain samples were obtained from the Gene Expression Omnibus database. Gene set enrichment analysis revealed an upregulation of immune­related responses and downregulation of ion transport, synaptic transmission and neurotransmitter homeostasis. Enrichment analysis of cell­specific differentially expressed genes also indicated a marked inflammatory environment in AxD. Overall, the clinical features of the two patients with bulbospinal form AxD were thoroughly described. To the best of our knowledge, the brain atrophy pattern and spontaneous brain functional network activity of patients with AxD were explored for the first time. Cytological experiments provided evidence of the pathogenicity of the identified variants. Furthermore, bioinformatics analysis found that inflammatory immune­related reactions may play a critical role in AxD, which may be conducive to the understanding of this disease.


Subject(s)
Alexander Disease/genetics , Alexander Disease/metabolism , Spinal Cord Diseases/genetics , Spinal Cord Diseases/metabolism , Adolescent , Adult , Alexander Disease/complications , Alexander Disease/diagnostic imaging , Brain/diagnostic imaging , Computational Biology , Female , Gait Disorders, Neurologic/complications , Glial Fibrillary Acidic Protein/chemistry , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Gray Matter/pathology , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Spinal Cord Diseases/complications , Young Adult
10.
Neurogenetics ; 22(3): 215-219, 2021 07.
Article in English | MEDLINE | ID: mdl-34046764

ABSTRACT

Alexander Disease (AxD) is a rare leukodystrophy caused by missense mutations of glial fibrillary acidic protein (GFAP). Primarily seen in infants and juveniles, it can present in adulthood. We report a family with inherited AxD in which the mother presented with symptoms many years after her daughter. We reviewed the age of onset in all published cases of familial AxD and found that 32 of 34 instances of parent-offspring pairs demonstrated an earlier age of onset in offspring compared to the parent. We suggest that genetic anticipation occurs in familial AxD and speculate that genetic mosaicism could explain this phenomenon.


Subject(s)
Alexander Disease/genetics , Anticipation, Genetic/genetics , Brain/pathology , Mutation/genetics , Alexander Disease/diagnosis , Alexander Disease/metabolism , Disease Progression , Female , Glial Fibrillary Acidic Protein/genetics , Humans , Middle Aged
11.
FASEB J ; 35(5): e21614, 2021 05.
Article in English | MEDLINE | ID: mdl-33908669

ABSTRACT

Alexander disease (AxD) caused by mutations in the coding region of GFAP is a neurodegenerative disease characterized by astrocyte dysfunction, GFAP aggregation, and Rosenthal fiber accumulation. Although how GFAP mutations cause disease is not fully understood, Rosenthal fibers could be induced by forced overexpression of human GFAP and this could be lethal in mice implicate that an increase in GFAP levels is central to AxD pathogenesis. Our recent studies demonstrated that intronic GFAP mutations cause disease by altering GFAP splicing, suggesting that an increase in GFAP isoform expression could lead to protein aggregation and astrocyte dysfunction that typify AxD. Here we test this hypothesis by establishing primary astrocyte cultures from transgenic mice overexpressing human GFAP. We found that GFAP-δ and GFAP-κ were disproportionately increased in transgenic astrocytes and both were enriched in Rosenthal fibers of human AxD brains. In vitro assembly studies showed that while the major isoform GFAP-α self-assembled into typical 10-nm filaments, minor isoforms including GFAP-δ, -κ, and -λ were assembly-compromised and aggregation prone. Lentiviral transduction showed that expression of these minor GFAP isoforms decreased filament solubility and increased GFAP stability, leading to the formation of Rosenthal fibers-like aggregates that also disrupted the endogenous intermediate filament networks. The aggregate-bearing astrocytes lost their normal morphology and glutamate buffering capacity, which had a toxic effect on neighboring neurons. In conclusion, our findings provide evidence that links elevated GFAP isoform expression with GFAP aggregation and impaired glutamate transport, and suggest a potential non-cell-autonomous mechanism underlying neurodegeneration through astrocyte dysfunction.


Subject(s)
Alexander Disease/pathology , Astrocytes/pathology , Glial Fibrillary Acidic Protein/chemistry , Glial Fibrillary Acidic Protein/metabolism , Glutamic Acid/metabolism , Mutation , Protein Aggregates , Alexander Disease/metabolism , Animals , Astrocytes/metabolism , Humans , Mice , Mice, Transgenic , Protein Conformation , Protein Isoforms
12.
J Neuroinflammation ; 18(1): 67, 2021 Mar 08.
Article in English | MEDLINE | ID: mdl-33685480

ABSTRACT

BACKGROUND: Alexander disease (AxD) is a rare neurodegenerative disorder that is caused by dominant mutations in the gene encoding glial fibrillary acidic protein (GFAP), an intermediate filament that is primarily expressed by astrocytes. In AxD, mutant GFAP in combination with increased GFAP expression result in astrocyte dysfunction and the accumulation of Rosenthal fibers. A neuroinflammatory environment consisting primarily of macrophage lineage cells has been observed in AxD patients and mouse models. METHODS: To examine if macrophage lineage cells could serve as a therapeutic target in AxD, GFAP knock-in mutant AxD model mice were treated with a colony-stimulating factor 1 receptor (CSF1R) inhibitor, pexidartinib. The effects of pexidartinib treatment on disease phenotypes were assessed. RESULTS: In AxD model mice, pexidartinib administration depleted macrophages in the CNS and caused elevation of GFAP transcript and protein levels with minimal impacts on other phenotypes including body weight, stress response activation, chemokine/cytokine expression, and T cell infiltration. CONCLUSIONS: Together, these results highlight the complicated role that macrophages can play in neurological diseases and do not support the use of pexidartinib as a therapy for AxD.


Subject(s)
Alexander Disease , Aminopyridines/pharmacology , Glial Fibrillary Acidic Protein/drug effects , Macrophages/drug effects , Pyrroles/pharmacology , Alexander Disease/metabolism , Alexander Disease/pathology , Animals , Disease Models, Animal , Glial Fibrillary Acidic Protein/metabolism , Mice , Mice, Inbred C57BL , Phenotype
13.
Int J Mol Sci ; 23(1)2021 Dec 23.
Article in English | MEDLINE | ID: mdl-35008573

ABSTRACT

In pathological brain conditions, glial cells become reactive and show a variety of responses. We examined Ca2+ signals in pathological brains and found that reactive astrocytes share abnormal Ca2+ signals, even in different types of diseases. In a neuropathic pain model, astrocytes in the primary sensory cortex became reactive and showed frequent Ca2+ signals, resulting in the production of synaptogenic molecules, which led to misconnections of tactile and pain networks in the sensory cortex, thus causing neuropathic pain. In an epileptogenic model, hippocampal astrocytes also became reactive and showed frequent Ca2+ signals. In an Alexander disease (AxD) model, hGFAP-R239H knock-in mice showed accumulation of Rosenthal fibers, a typical pathological marker of AxD, and excessively large Ca2+ signals. Because the abnormal astrocytic Ca2+ signals observed in the above three disease models are dependent on type II inositol 1,4,5-trisphosphate receptors (IP3RII), we reanalyzed these pathological events using IP3RII-deficient mice and found that all abnormal Ca2+ signals and pathologies were markedly reduced. These findings indicate that abnormal Ca2+ signaling is not only a consequence but may also be greatly involved in the cause of these diseases. Abnormal Ca2+ signals in reactive astrocytes may represent an underlying pathology common to multiple diseases.


Subject(s)
Alexander Disease , Astrocytes , Calcium Signaling , Calcium , Animals , Alexander Disease/metabolism , Astrocytes/metabolism , Calcium/metabolism , Calcium Signaling/physiology , Mice
14.
Genes (Basel) ; 11(12)2020 12 11.
Article in English | MEDLINE | ID: mdl-33322348

ABSTRACT

Alexander disease (AxD) is a rare astrogliopathy caused by heterozygous mutations, either inherited or arising de novo, on the glial fibrillary acid protein (GFAP) gene (17q21). Mutations in the GFAP gene make the protein prone to forming aggregates which, together with heat-shock protein 27 (HSP27), αB-crystallin, ubiquitin, and proteasome, contribute to form Rosenthal fibers causing a toxic effect on the cell. Unfortunately, no pharmacological treatment is available yet, except for symptom reduction therapies, and patients undergo a progressive worsening of the disease. The aim of this study was the production of a zebrafish model for AxD, to have a system suitable for drug screening more complex than cell cultures. To this aim, embryos expressing the human GFAP gene carrying the most severe p.R239C under the control of the zebrafish gfap gene promoter underwent functional validation to assess several features already observed in in vitro and other in vivo models of AxD, such as the localization of mutant GFAP inclusions, the ultrastructural analysis of cells expressing mutant GFAP, the effects of treatments with ceftriaxone, and the heat shock response. Our results confirm that zebrafish is a suitable model both to study the molecular pathogenesis of GFAP mutations and to perform pharmacological screenings, likely useful for the search of therapies for AxD.


Subject(s)
Alexander Disease , Animals, Genetically Modified , Ceftriaxone/pharmacology , Disease Models, Animal , Glial Fibrillary Acidic Protein , Mutation , Zebrafish , Alexander Disease/drug therapy , Alexander Disease/genetics , Alexander Disease/metabolism , Alexander Disease/pathology , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/metabolism , Drug Evaluation, Preclinical , Gene Expression , Glial Fibrillary Acidic Protein/biosynthesis , Glial Fibrillary Acidic Protein/genetics , Humans , Zebrafish/genetics , Zebrafish/metabolism
15.
Rinsho Shinkeigaku ; 60(10): 712-715, 2020 Oct 24.
Article in Japanese | MEDLINE | ID: mdl-32893243

ABSTRACT

A 50-year-old woman developed gait disturbances and dysarthria since the past 2 years. She also presented with dystonia and hypokinesia of her left lower limb, and orthostatic hypotension. The dopamine transporter SPECT with 123I ioflupane showed abnormal scans in bilateral striatum. Cerebral MRI revealed atrophy and signal changes in the medulla and spinal cord, from which Alexander disease (AxD) was suspected. Consequently, we checked the Glial fibrillary acidic protein (GFAP) gene. The analysis of the gene detected a heterozygous c.219G>T mutation, which was the first mutation reported in Japan, and finally she was diagnosed with AxD. Dystonia is relatively rare in AxD patients, but this case demonstrated that AxD should be listed in the differential diagnosis of extrapyramidal syndromes with abnormalities of the medulla and spinal cord on MRI.


Subject(s)
Alexander Disease/diagnosis , Dopamine Plasma Membrane Transport Proteins , Dopamine/metabolism , Dystonia/etiology , Lower Extremity , Tomography, Emission-Computed, Single-Photon , Alexander Disease/complications , Alexander Disease/diagnostic imaging , Alexander Disease/metabolism , Diagnosis, Differential , Dystonia/diagnostic imaging , Female , Glial Fibrillary Acidic Protein/genetics , Humans , Magnetic Resonance Imaging/methods , Middle Aged , Mutation
16.
ASN Neuro ; 12: 1759091420949680, 2020.
Article in English | MEDLINE | ID: mdl-32811163

ABSTRACT

Fifty years have passed since the discovery of glial fibrillary acidic protein (GFAP) by Lawrence Eng and colleagues. Now recognized as a member of the intermediate filament family of proteins, it has become a subject for study in fields as diverse as structural biology, cell biology, gene expression, basic neuroscience, clinical genetics and gene therapy. This review covers each of these areas, presenting an overview of current understanding and controversies regarding GFAP with the goal of stimulating continued study of this fascinating protein.


Subject(s)
Astrocytes/metabolism , Cloning, Molecular/methods , Glial Fibrillary Acidic Protein/biosynthesis , Glial Fibrillary Acidic Protein/genetics , Intermediate Filaments/genetics , Intermediate Filaments/metabolism , Alexander Disease/genetics , Alexander Disease/metabolism , Alexander Disease/pathology , Animals , Astrocytes/pathology , Humans , Intermediate Filaments/pathology , Time Factors
17.
Elife ; 82019 11 04.
Article in English | MEDLINE | ID: mdl-31682229

ABSTRACT

Alexander disease (AxD) is a fatal neurodegenerative disorder caused by mutations in glial fibrillary acidic protein (GFAP), which supports the structural integrity of astrocytes. Over 70 GFAP missense mutations cause AxD, but the mechanism linking different mutations to disease-relevant phenotypes remains unknown. We used AxD patient brain tissue and induced pluripotent stem cell (iPSC)-derived astrocytes to investigate the hypothesis that AxD-causing mutations perturb key post-translational modifications (PTMs) on GFAP. Our findings reveal selective phosphorylation of GFAP-Ser13 in patients who died young, independently of the mutation they carried. AxD iPSC-astrocytes accumulated pSer13-GFAP in cytoplasmic aggregates within deep nuclear invaginations, resembling the hallmark Rosenthal fibers observed in vivo. Ser13 phosphorylation facilitated GFAP aggregation and was associated with increased GFAP proteolysis by caspase-6. Furthermore, caspase-6 was selectively expressed in young AxD patients, and correlated with the presence of cleaved GFAP. We reveal a novel PTM signature linking different GFAP mutations in infantile AxD.


Subject(s)
Alexander Disease/metabolism , Biomarkers/metabolism , Caspases/metabolism , Glial Fibrillary Acidic Protein/metabolism , Adult , Alexander Disease/diagnosis , Alexander Disease/genetics , Astrocytes/metabolism , Binding Sites/genetics , Brain/metabolism , Brain/pathology , Cell Line , Glial Fibrillary Acidic Protein/genetics , Humans , Induced Pluripotent Stem Cells/metabolism , Infant , Intermediate Filaments/metabolism , Mutation , Phosphorylation , Proteolysis , Severity of Illness Index
18.
J Biol Chem ; 294(43): 15604-15612, 2019 10 25.
Article in English | MEDLINE | ID: mdl-31484723

ABSTRACT

Alexander disease (AxD) is an often fatal astrogliopathy caused by dominant gain-of-function missense mutations in the glial fibrillary acidic protein (GFAP) gene. The mechanism by which the mutations produce the AxD phenotype is not known. However, the observation that features of AxD are displayed by mice that express elevated levels of GFAP from a human WT GFAP transgene has contributed to the notion that the mutations produce AxD by increasing accumulation of total GFAP above some toxic threshold rather than the mutant GFAP being inherently toxic. A possible mechanism for accumulation of GFAP in AxD patients is that the mutated GFAP variants are more stable than the WT, an attribution abetted by observations that GFAP complexes containing GFAP variants are more resistant to solvent extraction. Here we tested this hypothesis by determining the relative levels of WT and mutant GFAP in three individuals with AxD, each of whom carried a common but different GFAP mutation (R79C, R239H, or R416W). Mass spectrometry analysis identified a peptide specific to the mutant or WT GFAP in each patient, and we quantified this peptide by comparing its signal to that of an added [15N]GFAP standard. In all three individuals, the level of mutant GFAP was less than that of the WT. This finding suggests that AxD onset is due to an intrinsic toxicity of the mutant GFAP instead of it acting indirectly by being more stable than WT GFAP and thereby increasing the total GFAP level.


Subject(s)
Alexander Disease/metabolism , Glial Fibrillary Acidic Protein/metabolism , Mutant Proteins/metabolism , Adolescent , Amino Acid Sequence , Child , Glial Fibrillary Acidic Protein/chemistry , Humans , Infant , Male , Mutant Proteins/chemistry , Peptides/chemistry , Peptides/metabolism , Protein Stability , Proteolysis , Reference Standards
19.
Cell Rep ; 25(4): 947-958.e4, 2018 10 23.
Article in English | MEDLINE | ID: mdl-30355500

ABSTRACT

How mutations in glial fibrillary acidic protein (GFAP) cause Alexander disease (AxD) remains elusive. We generated iPSCs from two AxD patients and corrected the GFAP mutations to examine the effects of mutant GFAP on human astrocytes. AxD astrocytes displayed GFAP aggregates, recapitulating the pathological hallmark of AxD. RNA sequencing implicated the endoplasmic reticulum, vesicle regulation, and cellular metabolism. Corroborating this analysis, we observed enlarged and heterogeneous morphology coupled with perinuclear localization of endoplasmic reticulum and lysosomes in AxD astrocytes. Functionally, AxD astrocytes showed impaired extracellular ATP release, which is responsible for attenuated calcium wave propagation. These results reveal that AxD-causing mutations in GFAP disrupt intracellular vesicle regulation and impair astrocyte secretion, resulting in astrocyte dysfunction and AxD pathogenesis.


Subject(s)
Astrocytes/metabolism , Glial Fibrillary Acidic Protein/genetics , Mutation/genetics , Organelles/metabolism , Adenosine Triphosphate/metabolism , Alexander Disease/metabolism , Alexander Disease/pathology , Animals , Astrocytes/cytology , Calcium Signaling , Cell Differentiation , Endoplasmic Reticulum/metabolism , Humans , Lysosomes/metabolism , Mice , Protein Aggregates , RNA, Messenger/genetics , RNA, Messenger/metabolism
20.
Neurología (Barc., Ed. impr.) ; 33(8): 526-533, oct. 2018. ilus
Article in Spanish | IBECS | ID: ibc-175967

ABSTRACT

Introducción: La enfermedad de Alexander (AxD) es una leucodistrofia. Su base patológica, junto a la pérdida de mielina, es la aparición de los cuerpos de Rosenthal, que son inclusiones citoplasmáticas en células astrocitarias. Mutaciones en el gen que codifica la GFAP se han identificado como una base genética para AxD. Sin embargo, no se conoce el mecanismo por el cual estas variantes producen la enfermedad. Desarrollo: La hipótesis más extendida es que AxD se desarrolla por un mecanismo por ganancia de función debido al incremento de GFAP. Sin embargo, este mecanismo no explica la pérdida mielínica, dado que los modelos experimentales que expresan GFAP normal o mutada no generan alteración mielínica. En la presente revisión se analizan otras posibilidades que permitan justificar dicha alteración, como son alteraciones epigenéticas, inflamatorias, la existencia de células NG2 (+)-GFAP (+) o cambios postraslacionales sobre la GFAP al margen de la mayor expresión. Conclusiones: Las diferentes hipótesis analizadas pueden explicar la alteración de la mielina que aparece en los pacientes y que pueden presentarse asociadas y abren la posibilidad de plantear terapéuticas basadas en estos mecanismos


Introduction: Alexander disease (AxD) is a type of leukodystrophy. Its pathological basis, along with myelin loss, is the appearance of Rosenthal bodies, which are cytoplasmic inclusions in astrocytes. Mutations in the gene coding for GFAP have been identified as a genetic basis for AxD. However, the mechanism by which these variants produce the disease is not understood. Development: The most widespread hypothesis is that AxD develops when a gain of function mutation causes an increase in GFAP. However, this mechanism does not explain myelin loss, given that experimental models in which GFAP expression is normal or mutated do not exhibit myelin disorders. This review analyses other possibilities that may explain this alteration, such as epigenetic or inflammatory alterations, presence of NG2 (+) - GFAP (+) cells, or post-translational modifications in GFAP that are unrelated to increased expression. Conclusions:The different hypotheses analysed here may explain the myelin alteration affecting these patients, and multiple mechanisms may coexist. These theories raise the possibility of designing therapies based on these mechanisms


Subject(s)
Humans , Animals , Myelin Sheath/metabolism , Alexander Disease/metabolism , Myelin Sheath/pathology , Alexander Disease/pathology , Glial Fibrillary Acidic Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL