Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Biol Chem ; 300(7): 107402, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38782207

RESUMEN

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.


Asunto(s)
Enfermedad de Alexander , Proteína Ácida Fibrilar de la Glía , Ubiquitinación , Enfermedad de Alexander/metabolismo , Enfermedad de Alexander/genética , Enfermedad de Alexander/patología , Proteína Ácida Fibrilar de la Glía/metabolismo , Proteína Ácida Fibrilar de la Glía/genética , Humanos , Animales , Mutación , Ratones , Astrocitos/metabolismo , Astrocitos/patología , Encéfalo/metabolismo , Encéfalo/patología , Procesamiento Proteico-Postraduccional , Ratas , Masculino , Femenino , Agregación Patológica de Proteínas/metabolismo , Agregación Patológica de Proteínas/genética , Agregación Patológica de Proteínas/patología
2.
J Alzheimers Dis Rep ; 7(1): 51-76, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36777330

RESUMEN

Background: Alzheimer's disease (AD) is a multifactorial disorder characterized by cognitive decline. Current available therapeutics for AD have limited clinical benefit. Therefore, preventive therapies for interrupting the development of AD are critically needed. Molecules targeting multifunction to interact with various pathlogical components have been considered to improve the therapeutic efficiency of AD. In particular, herbal medicines with multiplicity of actions produce cognitive benefits on AD. Bugu-M is a multi-herbal extract composed of Ganoderma lucidum (Antler form), Nelumbo nucifera Gaertn., Ziziphus jujuba Mill., and Dimocarpus longan, with the ability of its various components to confer resilience to cognitive deficits. Objective: To evaluate the potential of Bugu-M on amyloid-ß (Aß) toxicity and its in vitro mechanisms and on in vivo cognitive function. Methods: We illustrated the effect of Bugu-M on Aß25-35-evoked toxicity as well as its possible mechanisms to diminish the pathogenesis of AD in rat cortical neurons. For cognitive function studies, 2-month-old female 3×Tg-AD mice were administered 400 mg/kg Bugu-M for 30 days. Behavioral tests were performed to assess the efficacy of Bugu-M on cognitive impairment. Results: In primary cortical neuronal cultures, Bugu-M mitigated Aß-evoked toxicity by reducing cytoskeletal aberrations and axonal disruption, restoring presynaptic and postsynaptic protein expression, suppressing mitochondrial damage and apoptotic signaling, and reserving neurogenic and neurotrophic factors. Importantly, 30-day administration of Bugu-M effectively prevented development of cognitive impairment in 3-month-old female 3×Tg-AD mice. Conclusion: Bugu-M might be beneficial in delaying the progression of AD, and thus warrants consideration for its preventive potential for AD.

3.
Medicina (Kaunas) ; 58(12)2022 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-36556950

RESUMEN

Background and Objectives: The aim of this study was to investigate the relationships between obesity-related factors including body mass index (BMI), diabetes or prediabetes, hyperlipidemia, fasting plasma glucose, fasting plasma insulin, homeostasis model assessment-estimated insulin resistance (HOMA-IR), highly sensitive C-reactive protein (hs-CRP) and Graves' orbitopathy (GO). Materials and Methods: Eighty-four patients with Graves' disease (GD) (42 without GO and 42 with GO) were enrolled in this cross-sectional cohort study. Gender, age, GD treatment history, height, body weight, waist circumference, smoking status, co-morbidities, levels of free thyroxin, thyroid-stimulating hormone, thyroid-stimulating hormone receptor (TSHR) antibodies, fasting plasma glucose and insulin, and hs-CRP were recorded. The eye condition was evaluated using the consensus statement of the European Group of Graves' Orbitopathy (EUGOGO) and the NOSPECS classification. Results: In this study, multivariate regression analysis showed that BMI, fasting plasma insulin, and HOMA-IR were associated with the presence of GO after adjusting the age, gender, smoking, TSHR antibodies, and steroid usage (adjusted odd's ratio (aOR) 1.182, 95% confidence interval (95% CI), 1.003-1.393, p = 0.046; aOR 1.165, 95% CI, 1.001-1.355, p = 0.048; and aOR 1.985, 95% CI, 1.046-3.764, p = 0.036, respectively). In addition, BMI, fasting plasma glucose, fasting plasma insulin, HOMA-IR, and hs-CRP levels were positively correlated with the severity of GO. Conclusions: The findings of this study suggest that obesity-related factors, especially fasting plasma insulin and HOMA-IR, are related to GO. Our study highlighted the importance of obesity-related factors in GO. Obesity-related factors may cause the development of GO or occur simultaneously with GO.


Asunto(s)
Enfermedad de Graves , Oftalmopatía de Graves , Resistencia a la Insulina , Humanos , Oftalmopatía de Graves/complicaciones , Proyectos Piloto , Proteína C-Reactiva/metabolismo , Glucemia , Estudios Transversales , Insulina , Obesidad/complicaciones
4.
Mol Biol Cell ; 33(8): ar69, 2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35511821

RESUMEN

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.


Asunto(s)
Enfermedad de Alexander , Proteína Ácida Fibrilar de la Glía/metabolismo , Enfermedad de Alexander/genética , Enfermedad de Alexander/metabolismo , Astrocitos/metabolismo , Proteína Ácida Fibrilar de la Glía/genética , Humanos , Filamentos Intermedios/metabolismo , Mutación/genética
5.
Eur J Hum Genet ; 30(6): 687-694, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35246666

RESUMEN

Alexander disease (AxD) is a neurodegenerative astrogliopathy caused by mutation in the glial fibrillary acidic protein (GFAP) gene. A 42-year-old Korean man presented with temporary gait disturbance and psychiatric regression after a minor head trauma in the absence of bulbar symptoms and signs. Magnetic resonance images of the brain and spinal cord showed significant atrophy of the medulla oblongata and the entire spinal cord as well as contrast-enhanced T2 hypointensity in the basal ganglia. DNA sequencing revealed a novel 33-bp in-frame deletion mutation (p.Glu138_Leu148del) within the 1B rod domain of GFAP, which was predicted to be deleterious by PROVEAN analysis. To test whether the deletion mutant is disease-causing, we performed in vitro GFAP assembly and sedimentation assays, and GFAP aggregation assays in human adrenal carcinoma SW13 (Vim-) cells and rat primary astrocytes. All the assays revealed that GFAP p.Glu138_Leu148del is aggregation prone. Based on these findings, we diagnosed the patient with Type II AxD. This is a report that demonstrates the pathogenicity of InDel mutation of GFAP through functional studies. This patient's atypical presentation as well as the discrepancy between clinical symptoms and radiologic findings may extend the scope of AxD.


Asunto(s)
Enfermedad de Alexander , Enfermedad de Alexander/diagnóstico , Enfermedad de Alexander/genética , Enfermedad de Alexander/patología , Animales , Encéfalo/metabolismo , Proteína Ácida Fibrilar de la Glía/genética , Humanos , Mutación , Fenotipo , Ratas
6.
Sci Transl Med ; 13(620): eabg4711, 2021 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-34788075

RESUMEN

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.


Asunto(s)
Enfermedad de Alexander , Proteína Ácida Fibrilar de la Glía , Trastornos Motores , Sustancia Blanca , Enfermedad de Alexander/genética , Enfermedad de Alexander/metabolismo , Enfermedad de Alexander/patología , Animales , Astrocitos/metabolismo , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Gliosis/patología , Trastornos Motores/metabolismo , Trastornos Motores/patología , Mutación/genética , Ratas , Sustancia Blanca/patología
7.
FASEB J ; 35(5): e21614, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33908669

RESUMEN

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.


Asunto(s)
Enfermedad de Alexander/patología , Astrocitos/patología , Proteína Ácida Fibrilar de la Glía/química , Proteína Ácida Fibrilar de la Glía/metabolismo , Ácido Glutámico/metabolismo , Mutación , Agregado de Proteínas , Enfermedad de Alexander/metabolismo , Animales , Astrocitos/metabolismo , Humanos , Ratones , Ratones Transgénicos , Conformación Proteica , Isoformas de Proteínas
8.
Mov Disord ; 35(9): 1662-1667, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32374915

RESUMEN

BACKGROUND: Alexander disease (AxD) is an autosomal-dominant leukodystrophy caused by heterozygous mutations in the glial fibrillary acidic protein (GFAP) gene. OBJECTIVES: The objective of this report is to characterize the clinical phenotype and identify the genetic mutation associated with adult-onset AxD. METHODS: A man presented with progressive unsteadiness since age 16. Magnetic resonance imaging findings revealed characteristic features of AxD. The GFAP gene was screened, and a candidate variant was functionally tested to evaluate causality. RESULTS: A homozygous c.197G > A (p.Arg66Gln) mutation was found in the proband, and his asymptomatic parents were heterozygous for the same mutation. This mutation affected GFAP solubility and promoted filament aggregation. The presence of the wild-type protein rescued mutational effects, consistent with the recessive nature of this mutation. CONCLUSIONS: This study is the first report of AxD caused by a homozygous mutation in GFAP. The clinical implication is while examining patients with characteristic features on suspicion of AxD, GFAP screening is recommended even without a supportive family history. © 2020 International Parkinson and Movement Disorder Society.


Asunto(s)
Enfermedad de Alexander , Adolescente , Adulto , Enfermedad de Alexander/diagnóstico por imagen , Enfermedad de Alexander/genética , Proteína Ácida Fibrilar de la Glía/genética , Homocigoto , Humanos , Masculino , Mutación/genética , Fenotipo
9.
Oxid Med Cell Longev ; 2020: 7353618, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32047579

RESUMEN

Cisplatin chemotherapy causes myelosuppression and often limits treatment duration and dose escalation in patients. Novel approaches to circumvent or lessen myelotoxicity may improve clinical outcome and quality of life in these patients. Chlorella sorokiniana (CS) is a freshwater unicellular green alga and exhibits encouraging efficacy in immunomodulation and anticancer in preclinical studies. However, the efficacy of CS on chemoprotection remains unclear. We report here, for the first time, that CS extract (CSE) could protect normal myeloid cells and PBMCs from cisplatin toxicity. Also, cisplatin-induced apoptosis in HL-60 cells was rescued through reservation of mitochondrial function, inhibition of cytochrome c release to cytosol, and suppression of caspase and PARP activation. Intriguingly, cotreatment of CSE attenuated cisplatin-evoked hypocellularity of bone marrow in mice. Furthermore, we observed the enhancement of CSF-GM activity in bone marrow and spleen in mice administered CSE and cisplatin, along with increased CD11b levels in spleen. In conclusion, we uncovered a novel mechanism of CSE on myeloprotection, whereby potentially supports the use of CSE as a chemoprotector against cisplatin-induced bone marrow toxicity. Further clinical investigation of CSE in combination with cisplatin is warranted.


Asunto(s)
Antineoplásicos/efectos adversos , Células de la Médula Ósea/efectos de los fármacos , Cisplatino/efectos adversos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/tratamiento farmacológico , Mitocondrias/metabolismo , Células Mieloides/efectos de los fármacos , Extractos Vegetales/uso terapéutico , Células de la Médula Ósea/patología , Antígeno CD11b/metabolismo , Chlorella , Cisplatino/uso terapéutico , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Células HL-60 , Humanos , Inmunomodulación , Terapia de Inmunosupresión , Células Mieloides/patología
10.
Elife ; 82019 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-31682229

RESUMEN

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.


Asunto(s)
Enfermedad de Alexander/metabolismo , Biomarcadores/metabolismo , Caspasas/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Adulto , Enfermedad de Alexander/diagnóstico , Enfermedad de Alexander/genética , Astrocitos/metabolismo , Sitios de Unión/genética , Encéfalo/metabolismo , Encéfalo/patología , Línea Celular , Proteína Ácida Fibrilar de la Glía/genética , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Lactante , Filamentos Intermedios/metabolismo , Mutación , Fosforilación , Proteolisis , Índice de Severidad de la Enfermedad
11.
Oxid Med Cell Longev ; 2017: 5414297, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29181126

RESUMEN

Neuroinflammation plays a central role in the pathophysiology of Alzheimer's disease (AD). Compounds that suppress neuroinflammation have been identified as potential therapeutic targets for AD. Rhinacanthin C (RC), a naphthoquinone ester found in Rhinacanthus nasutus Kurz (Acanthaceae), is currently proposed as an effective molecule against inflammation. However, the exact role of RC on neuroinflammation remains to be elucidated. In the present study, we investigated RC effect on modulating lipopolysaccharides (LPS), amyloid-ß peptide (Aß), or interferon-γ- (IFN-γ-) evoked pathological events in neurons and glia. Our findings demonstrated that RC prevented Aß-induced toxicity in rat hippocampal neurons and attenuated LPS-activated nitric oxide (NO) production, inducible nitric oxide synthase (iNOS) expression, and NF-κB signaling in rat glia. Likewise, RC suppressed LPS-induced neuroinflammation by reducing NO production and iNOS, IL-1ß, CCL-2, and CCL-5 mRNA levels in rat microglia. Further studies using BV-2 microglia revealed that RC inhibited LPS-, Aß-, and IFN-γ-stimulated IL-6 and TNF-α secretion. Of note, NF-κB and ERK activation was abrogated by RC in BV-2 cell response to Aß or IFN-γ. Moreover, RC protected neurons from Aß-stimulated microglial conditioned media-dependent toxicity. Collectively, these data highlight the beneficial effects of RC on neuroprotection and support the therapeutic implications of RC to neuroinflammation-mediated conditions.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Inflamación/tratamiento farmacológico , Lipopolisacáridos/metabolismo , Naftoquinonas/uso terapéutico , Neuronas/metabolismo , Animales , Interferón gamma/metabolismo , Ratones , Naftoquinonas/farmacología , Neuroglía/metabolismo , Ratas , Ratas Sprague-Dawley
12.
BMC Neurol ; 17(1): 175, 2017 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-28882119

RESUMEN

BACKGROUND: Alexander disease (AxD) is an astrogliopathy that predominantly affects the white matter of the central nervous system (CNS), and is caused by a mutation in the gene encoding the glial fibrillary acidic protein (GFAP), an intermediate filament primarily expressed in astrocytes and ependymal cells. The main pathologic feature of AxD is the presence of Rosenthal fibers (RFs), homogeneous eosinophilic inclusions found in astrocytes. Because of difficulties in procuring patient' CNS tissues and the presence of RFs in other pathologic conditions, there is a need to develop an in vivo assay that can determine whether a mutation in the GFAP results in aggregation and is thus disease-causing. METHODS: We found a GFAP mutation (c.382G > A, p.Asp128Asn) in a 68-year-old man with slowly progressive gait disturbance with tendency to fall. The patient was tentatively diagnosed with AxD based on clinical and radiological findings. To develop a vertebrate model to assess the aggregation tendency of GFAP, we expressed several previously reported mutant GFAPs and p.Asp128Asn GFAP in zebrafish embryos. RESULTS: The most common GFAP mutations in AxD, p.Arg79Cys, p.Arg79His, p.Arg239Cys and p.Arg239His, and p.Asp128Asn induced a significantly higher number of GFAP aggregates in zebrafish embryos than wild-type GFAP. CONCLUSIONS: The p.Asp128Asn GFAP mutation is likely to be a disease-causing mutation. Although it needs to be tested more extensively in larger case series, the zebrafish assay system presented here would help clinicians determine whether GFAP mutations identified in putative AxD patients are disease-causing.


Asunto(s)
Enfermedad de Alexander/genética , Proteína Ácida Fibrilar de la Glía/genética , Anciano , Animales , Astrocitos , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Masculino , Mutación , Pez Cebra
13.
PLoS One ; 12(7): e0180694, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28700643

RESUMEN

Alexander disease (AxD) is a neurodegenerative disease caused by heterozygous mutations in the GFAP gene, which encodes the major intermediate filament protein of astrocytes. This disease is characterized by the accumulation of cytoplasmic protein aggregates, known as Rosenthal fibers. Antibodies specific to GFAP could provide invaluable tools to facilitate studies of the normal biology of GFAP and to elucidate the pathologic role of this IF protein in disease. While a large number of antibodies to GFAP are available, few if any of them have defined epitopes. Here we described the characterization of a panel of commonly used anti-GFAP antibodies, which recognized epitopes at regions extending across the rod domain of GFAP. We show that all of the antibodies are useful for immunoblotting and immunostaining, and identify a subset that preferentially recognized human GFAP. Using these antibodies, we demonstrate the presence of biochemically modified forms of GFAP in brains of human AxD patients and mouse AxD models. These data suggest that this panel of anti-GFAP antibodies will be useful for studies of animal and cell-based models of AxD and related diseases in which cytoskeletal defects associated with GFAP modifications occur.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Epítopos/inmunología , Proteína Ácida Fibrilar de la Glía/inmunología , Adolescente , Adulto , Enfermedad de Alexander/genética , Enfermedad de Alexander/patología , Animales , Especificidad de Anticuerpos/inmunología , Encéfalo/metabolismo , Encéfalo/patología , Línea Celular Tumoral , Niño , Modelos Animales de Enfermedad , Mapeo Epitopo , Femenino , Humanos , Lactante , Masculino , Ratones Transgénicos , Persona de Mediana Edad , Dominios Proteicos , Proteolisis , Solubilidad , Ubiquitinación , Regulación hacia Arriba , Adulto Joven
14.
Mol Biol Cell ; 27(25): 3980-3990, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27798231

RESUMEN

Alexander disease (AxD) is a primary genetic disorder of astrocytes caused by dominant mutations in the gene encoding the intermediate filament (IF) protein GFAP. This disease is characterized by excessive accumulation of GFAP, known as Rosenthal fibers, within astrocytes. Abnormal GFAP aggregation also occurs in giant axon neuropathy (GAN), which is caused by recessive mutations in the gene encoding gigaxonin. Given that one of the functions of gigaxonin is to facilitate proteasomal degradation of several IF proteins, we sought to determine whether gigaxonin is involved in the degradation of GFAP. Using a lentiviral transduction system, we demonstrated that gigaxonin levels influence the degradation of GFAP in primary astrocytes and in cell lines that express this IF protein. Gigaxonin was similarly involved in the degradation of some but not all AxD-associated GFAP mutants. In addition, gigaxonin directly bound to GFAP, and inhibition of proteasome reversed the clearance of GFAP in cells achieved by overexpressing gigaxonin. These studies identify gigaxonin as an important factor that targets GFAP for degradation through the proteasome pathway. Our findings provide a critical foundation for future studies aimed at reducing or reversing pathological accumulation of GFAP as a potential therapeutic strategy for AxD and related diseases.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Enfermedad de Alexander/metabolismo , Astrocitos/metabolismo , Astrocitos/fisiología , Células Cultivadas , Proteínas del Citoesqueleto/genética , Neuropatía Axonal Gigante/genética , Neuropatía Axonal Gigante/metabolismo , Proteína Ácida Fibrilar de la Glía/genética , Humanos , Mutación , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis
15.
Methods Enzymol ; 569: 155-75, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26778558

RESUMEN

Intermediate filament (IF) scaffolds facilitate small heat shock protein (sHSP) function, while IF function is sHSP dependent. sHSPs interact with IFs and the importance of this interaction is to maintain the individuality of the IFs and to modulate interfilament interactions both in networks and in assembly intermediates. Mutations in both sHSPs and their interacting IF proteins phenocopy each other in the human diseases they cause. This establishes a key functional relationship between these two very distinct protein families, and it also evidences the role of this cytoskeleton-chaperone complex in the cellular stress response. In this chapter, we describe the detailed experimental protocols for the preparation of purified IF proteins and sHSPs to facilitate the study in vitro of their functional interactions. In addition, we describe the detailed biochemical procedures to assess the effect of sHSP on the assembly of IFs, the binding to IFs, and the prevention of noncovalent filament-filament interactions using in vitro cosedimentation, electron microscopy, and viscosity assays. These assays are valuable research tools to study and manipulate sHSP-IF complexes in vitro and therefore to determine the structure-function detail of this complex, and how it contributes to cellular, tissue, and organismal homeostasis and the in vivo stress response.


Asunto(s)
Proteínas del Citoesqueleto/química , Proteínas de Choque Térmico/aislamiento & purificación , Filamentos Intermedios/química , Cromatografía en Gel , Cromatografía por Intercambio Iónico , Proteínas del Citoesqueleto/aislamiento & purificación , Escherichia coli , Proteínas de Choque Térmico/química , Humanos , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación
16.
Eur J Hum Genet ; 23(1): 72-8, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24755947

RESUMEN

Alexander disease (AxD) is an astrogliopathy that primarily affects the white matter of the central nervous system (CNS). AxD is caused by mutations in a gene encoding GFAP (glial fibrillary acidic protein). The GFAP mutations in AxD have been reported to act in a gain-of-function manner partly because the identified mutations generate practically full-length GFAP. We found a novel nonsense mutation (c.1000 G>T, p.(Glu312Ter); also termed p.(E312*)) within a rod domain of GFAP in a 67-year-old Korean man with a history of memory impairment and leukoencephalopathy. This mutation, GFAP p.(E312*), removes part of the 2B rod domain and the whole tail domain from the GFAP. We characterized GFAP p.(E312*) using western blotting, in vitro assembly and sedimentation assay, and transient transfection of human adrenal cortex carcinoma SW13 (Vim(+)) cells with plasmids encoding GFAP p.(E312*). The GFAP p.(E312*) protein, either alone or in combination with wild-type GFAP, elicited self-aggregation. In addition, the assembled GFAP p.(E312*) aggregated into paracrystal-like structures, and GFAP p.(E312*) elicited more GFAP aggregation than wild-type GFAP in the human adrenal cortex carcinoma SW13 (Vim(+)) cells. Our findings are the first report, to the best of our knowledge, on this novel nonsense mutation of GFAP that is associated with AxD and paracrystal formation.


Asunto(s)
Enfermedad de Alexander/diagnóstico , Enfermedad de Alexander/genética , Codón sin Sentido , Proteína Ácida Fibrilar de la Glía/genética , Dominios y Motivos de Interacción de Proteínas/genética , Anciano , Encéfalo/patología , Línea Celular , Análisis Mutacional de ADN , Expresión Génica , Proteína Ácida Fibrilar de la Glía/química , Células HEK293 , Humanos , Imagen por Resonancia Magnética , Masculino , Fenotipo , Agregación Patológica de Proteínas
17.
PLoS One ; 9(12): e116229, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25551560

RESUMEN

BACKGROUND: Chronic pancreatitis (CP) is a necroinflammatory process resulting in extensive pancreatic fibrosis. Granulocyte colony-stimulating factor (G-CSF), a hematopoietic stem cell mobilizer, has been shown to exert an anti-fibrotic effect partly through the enrichment of bone marrow (BM) cells in fibrotic organ. We aimed to test the effect of G-CSF on fibrosis in a mouse model of CP. METHODS: CP was induced in C57Bl/6J mice by consecutive cerulein injection (50 µg/kg/day, 2 days a week) for 6 weeks. Mice were then treated with G-CSF (200 µg/kg/day, 5 day a week) or normal saline for 1 week, and sacrificed at week 7 or week 9 after first cerulein injection. Pancreatic histology, pancreatic matrix metallopeptidase 9 (MMP-9), MMP-13 and collagen expression were examined. Pancreatic myofibroblasts were isolated and cultured with G-CSF. Collagen, MMP-9 and MMP-13 expression by myofibroblasts was examined. The BM-mismatched mice model was used to examine the change of BM-derived myofibroblasts and non-myofibroblastic BM cells by G-CSF in the pancreas. RESULTS: The pancreatic collagen expression were significantly decreased in the G-CSF-treated group sacrificed at week 9. While collagen produced from myofibroblasts was not affected by G-CSF, the increase of MMP13 expression was observed in vitro. There were no effect of G-CSF in the number of myofibroblasts and BM-derived myofibroblasts. However, the number of non-myofibroblastic BM cells and macrophages were significantly increased in the pancreata of cerulein- and G-CSF-treated mice, suggesting a potential anti-fibrotic role of non-myofibroblastic BM cells and macrophages stimulated by G-CSF. CONCLUSIONS: Our data indicated that G-CSF contributed to the regression of pancreatic fibrosis. The anti-fibrotic effects were possibly through the stimulation of MMP-13 from myofibroblasts, and the enhanced accumulation of non-myofibroblastic BM cells and macrophages in the pancreas.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos/farmacología , Pancreatitis Crónica/tratamiento farmacológico , Pancreatitis Crónica/patología , Animales , Células de la Médula Ósea/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Colágeno/metabolismo , Modelos Animales de Enfermedad , Femenino , Fibrosis/tratamiento farmacológico , Fibrosis/patología , Proteínas Fluorescentes Verdes/genética , Macrófagos/efectos de los fármacos , Masculino , Metaloproteinasa 13 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Miofibroblastos/efectos de los fármacos , Miofibroblastos/metabolismo , Páncreas/efectos de los fármacos , Páncreas/metabolismo , Pancreatitis Crónica/metabolismo
18.
ASN Neuro ; 5(5): e00125, 2013 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-24102621

RESUMEN

IF (intermediate filament) proteins can be cleaved by caspases to generate proapoptotic fragments as shown for desmin. These fragments can also cause filament aggregation. The hypothesis is that disease-causing mutations in IF proteins and their subsequent characteristic histopathological aggregates could involve caspases. GFAP (glial fibrillary acidic protein), a closely related IF protein expressed mainly in astrocytes, is also a putative caspase substrate. Mutations in GFAP cause AxD (Alexander disease). The overexpression of wild-type or mutant GFAP promotes cytoplasmic aggregate formation, with caspase activation and GFAP proteolysis. In this study, we report that GFAP is cleaved specifically by caspase 6 at VELD²²5 in its L12 linker domain in vitro. Caspase cleavage of GFAP at Asp²²5 produces two major cleavage products. While the C-GFAP (C-terminal GFAP) is unable to assemble into filaments, the N-GFAP (N-terminal GFAP) forms filamentous structures that are variable in width and prone to aggregation. The effect of N-GFAP is dominant, thus affecting normal filament assembly in a way that promotes filament aggregation. Transient transfection of N-GFAP into a human astrocytoma cell line induces the formation of cytoplasmic aggregates, which also disrupt the endogenous GFAP networks. In addition, we generated a neo-epitope antibody that recognizes caspase-cleaved but not the intact GFAP. Using this antibody, we demonstrate the presence of the caspase-generated GFAP fragment in transfected cells expressing a disease-causing mutant GFAP and in two mouse models of AxD. These findings suggest that caspase-mediated GFAP proteolysis may be a common event in the context of both the GFAP mutation and excess.


Asunto(s)
Caspasa 6/farmacología , Citoesqueleto/metabolismo , Proteína Ácida Fibrilar de la Glía/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/metabolismo , Proteolisis/efectos de los fármacos , Enfermedad de Alexander/genética , Enfermedad de Alexander/metabolismo , Animales , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/patología , Línea Celular Tumoral , Citoesqueleto/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica/genética , Proteína Ácida Fibrilar de la Glía/genética , Humanos , Ratones , Ratones Transgénicos , Mutagénesis Sitio-Dirigida , Mutación/genética , Péptidos/farmacología , Unión Proteica/efectos de los fármacos , Unión Proteica/genética
19.
Exp Cell Res ; 317(16): 2252-66, 2011 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-21756903

RESUMEN

Alexander disease is a primary genetic disorder of astrocyte caused by dominant mutations in the astrocyte-specific intermediate filament glial fibrillary acidic protein (GFAP). While most of the disease-causing mutations described to date have been found in the conserved α-helical rod domain, some mutations are found in the C-terminal non-α-helical tail domain. Here, we compare five different mutations (N386I, S393I, S398F, S398Y and D417M14X) located in the C-terminal domain of GFAP on filament assembly properties in vitro and in transiently transfected cultured cells. All the mutations disrupted in vitro filament assembly. The mutations also affected the solubility and promoted filament aggregation of GFAP in transiently transfected MCF7, SW13 and U343MG cells. This correlated with the activation of the p38 stress-activated protein kinase and an increased association with the small heat shock protein (sHSP) chaperone, αB-crystallin. Of the mutants studied, D417M14X GFAP caused the most significant effects both upon filament assembly in vitro and in transiently transfected cells. This mutant also caused extensive filament aggregation coinciding with the sequestration of αB-crystallin and HSP27 as well as inhibition of the proteosome and activation of p38 kinase. Associated with these changes were an activation of caspase 3 and a significant decrease in astrocyte viability. We conclude that some mutations in the C-terminus of GFAP correlate with caspase 3 cleavage and the loss of cell viability, suggesting that these could be contributory factors in the development of Alexander disease.


Asunto(s)
Enfermedad de Alexander/genética , Caspasa 3/metabolismo , Supervivencia Celular/genética , Proteína Ácida Fibrilar de la Glía/genética , Filamentos Intermedios/metabolismo , Mutación/fisiología , Enfermedad de Alexander/etiología , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Astrocitoma/patología , Línea Celular Tumoral , Centrifugación , Ciclina D1/metabolismo , Epítopos/inmunología , Mutación del Sistema de Lectura/fisiología , Proteína Ácida Fibrilar de la Glía/inmunología , Proteína Ácida Fibrilar de la Glía/metabolismo , Proteínas de Choque Térmico HSP27/metabolismo , Proteínas de Choque Térmico , Humanos , Filamentos Intermedios/patología , Filamentos Intermedios/ultraestructura , Microscopía Electrónica de Transmisión , Chaperonas Moleculares , Mutagénesis Sitio-Dirigida , Mutación Missense/fisiología , Fosforilación , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica/fisiología , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Solubilidad , Transfección , Ubiquitina/metabolismo , Vimentina/metabolismo , Cadena B de alfa-Cristalina/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
20.
FEBS Lett ; 583(8): 1267-73, 2009 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-19265695

RESUMEN

The apparently stationary phase of mitochondrial motion was investigated in epithelial cells by spinning disk confocal light microscopy combined with image correlation based single particle tracking using custom software producing sub-pixel accuracy measurements (approximately 5 nm) at 10-12 Hz frame-rates. The analysis of these data suggests that the previously described stationary, or anchored phase, in mitochondrial movement actually comprise Brownian diffusion, interspersed with frequent and brief motor-driven events whose duration are stochastically determined. We have therefore discovered a new aspect of mitochondrial behavior, which we call stochastically determined, directed movement.


Asunto(s)
Mitocondrias/fisiología , Procesos Estocásticos , Animales , Línea Celular , Dipodomys , Humanos , Microscopía Confocal , Técnicas de Cultivo de Tejidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...