Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 284
Filtrar
1.
Nat Commun ; 15(1): 1451, 2024 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-38365903

RESUMO

Mutations in rhodopsin can cause it to misfold and lead to retinal degeneration. A distinguishing feature of these mutants in vitro is that they mislocalize and aggregate. It is unclear whether or not these features contribute to retinal degeneration observed in vivo. The effect of P23H and G188R misfolding mutations were examined in a heterologous expression system and knockin mouse models, including a mouse model generated here expressing the G188R rhodopsin mutant. In vitro characterizations demonstrate that both mutants aggregate, with the G188R mutant exhibiting a more severe aggregation profile compared to the P23H mutant. The potential for rhodopsin mutants to aggregate in vivo was assessed by PROTEOSTAT, a dye that labels aggregated proteins. Both mutants mislocalize in photoreceptor cells and PROTEOSTAT staining was detected surrounding the nuclei of photoreceptor cells. The G188R mutant promotes a more severe retinal degeneration phenotype and greater PROTEOSTAT staining compared to that promoted by the P23H mutant. Here, we show that the level of PROTEOSTAT positive cells mirrors the progression and level of photoreceptor cell death, which suggests a potential role for rhodopsin aggregation in retinal degeneration.


Assuntos
Degeneração Retiniana , Retinite Pigmentosa , Rodopsina , Animais , Camundongos , Modelos Animais de Doenças , Mutação , Células Fotorreceptoras/metabolismo , Degeneração Retiniana/genética , Retinite Pigmentosa/genética , Retinite Pigmentosa/metabolismo , Rodopsina/genética , Rodopsina/metabolismo , Agregados Proteicos/genética
2.
PLoS Genet ; 20(2): e1011194, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38422160

RESUMO

Misfolded proteins are usually refolded to their functional conformations or degraded by quality control mechanisms. When misfolded proteins evade quality control, they can be sequestered to specific sites within cells to prevent the potential dysfunction and toxicity that arises from protein aggregation. Btn2 and Hsp42 are compartment-specific sequestrases that play key roles in the assembly of these deposition sites. Their exact intracellular functions and substrates are not well defined, particularly since heat stress sensitivity is not observed in deletion mutants. We show here that Btn2 and Hsp42 are required for tolerance to oxidative stress conditions induced by exposure to hydrogen peroxide. Btn2 and Hsp42 act to sequester oxidized proteins into defined PQC sites following ROS exposure and their absence leads to an accumulation of protein aggregates. The toxicity of protein aggregate accumulation causes oxidant sensitivity in btn2 hsp42 sequestrase mutants since overexpression of the Hsp104 disaggregase rescues oxidant tolerance. We have identified the Sup35 translation termination factor as an in vivo sequestrase substrate and show that Btn2 and Hsp42 act to suppress oxidant-induced formation of the yeast [PSI+] prion, which is the amyloid form of Sup35. [PSI+] prion formation in sequestrase mutants does not require IPOD (insoluble protein deposit) localization which is the site where amyloids are thought to undergo fragmentation and seeding to propagate their heritable prion form. Instead, both amorphous and amyloid Sup35 aggregates are increased in btn2 hsp42 mutants consistent with the idea that prion formation occurs at multiple intracellular sites during oxidative stress conditions in the absence of sequestrase activity. Taken together, our data identify protein sequestration as a key antioxidant defence mechanism that functions to mitigate the damaging consequences of protein oxidation-induced aggregation.


Assuntos
Príons , Proteínas de Saccharomyces cerevisiae , Agregados Proteicos/genética , Príons/genética , Príons/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Estresse Oxidativo/genética , Amiloide/metabolismo , Oxidantes/farmacologia , Oxidantes/metabolismo , Fatores de Terminação de Peptídeos/genética , Fatores de Terminação de Peptídeos/metabolismo
3.
Nucleic Acids Res ; 51(21): 11466-11478, 2023 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-37870427

RESUMO

Nucleic acids can act as potent modulators of protein aggregation, and RNA has the ability to either hinder or facilitate protein assembly, depending on the molecular context. In this study, we utilized a computational approach to characterize the physico-chemical properties of regions involved in amyloid aggregation. In various experimental datasets, we observed that while the core is hydrophobic and highly ordered, external regions, which are more disordered, display a distinct tendency to interact with nucleic acids. To validate our predictions, we performed aggregation assays with alpha-synuclein (aS140), a non-nucleic acid-binding amyloidogenic protein, and a mutant truncated at the acidic C-terminus (aS103), which is predicted to have a higher tendency to interact with RNA. For both aS140 and aS103, we observed an acceleration of aggregation upon RNA addition, with a significantly stronger effect for aS103. Due to favorable electrostatics, we noted an enhanced nucleic acid sequestration ability for the aggregated aS103, allowing it to entrap a larger amount of RNA compared to the aggregated wild-type counterpart. Overall, our research suggests that RNA sequestration might be a common phenomenon linked to protein aggregation, constituting a gain-of-function mechanism that warrants further investigation.


Assuntos
Agregados Proteicos , alfa-Sinucleína , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Agregados Proteicos/genética , RNA/genética , Amiloide/genética , Amiloide/química , Proteínas Amiloidogênicas
4.
J Mol Biol ; 435(22): 168281, 2023 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-37734431

RESUMO

Amyloid aggregation is a key process in amyloidoses and neurodegenerative diseases. Hydrophobicity is one of the major driving forces for this type of aggregation, as an increase in hydrophobicity generally correlates with aggregation susceptibility and rate. However, most experimental systems in vitro and prediction tools in silico neglect the contribution of protective osmolytes present in the cellular environment. Here, we assessed the role of hydrophobic mutations in amyloid aggregation in the presence of osmolytes. To achieve this goal, we used the model protein human muscle acylphosphatase (mAcP) and mutations to leucine that increased its hydrophobicity without affecting its thermodynamic stability. Osmolytes significantly slowed down the aggregation kinetics of the hydrophobic mutants, with an effect larger than that observed on the wild-type protein. The effect increased as the mutation site was closer to the middle of the protein sequence. We propose that the preferential exclusion of osmolytes from mutation-introduced hydrophobic side-chains quenches the aggregation potential of the ensemble of partially unfolded states of the protein by inducing its compaction and inhibiting its self-assembly with other proteins. Our results suggest that including the effect of the cellular environment in experimental setups and predictive softwares, for both mechanistic studies and drug design, is essential in order to obtain a more complete combination of the driving forces of amyloid aggregation.


Assuntos
Amiloide , Agregados Proteicos , Humanos , Sequência de Aminoácidos , Amiloide/química , Amiloide/genética , Leucina/química , Leucina/genética , Dobramento de Proteína , Agregados Proteicos/genética , /genética , Interações Hidrofóbicas e Hidrofílicas , Solubilidade , Pressão Osmótica , Ureia/química
5.
Mol Biol Rep ; 50(7): 5547-5556, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37155014

RESUMO

BACKGROUND: α-synuclein aggregation is the hallmark feature of Parkinson's disease. Both familial and sporadic forms of the disease exhibit this feature. Several mutations have been identified in patients and are associated with the disease pathology. METHODS AND RESULTS: We have used site-directed mutagenesis to generate α-synuclein mutant variants tagged with GFP. Fluorescence microscopy, flow cytometry, western blotting, cell viability and oxidative stress analysis were performed to investigate the effect of two less studied α-synuclein variants. In this study we characterized two less studied α-synuclein mutations, A18T and A29S, in the well-established yeast model. Our data shows variable expression, distribution and toxicity of the protein in the mutant variants A18T, A29S, A53T and WT. The cells expressing the double mutant variant A18T/A53T showed the most increase in the aggregation phenotype and also depicted reduced viability suggesting a more substantial effect of this variant. CONCLUSION: The outcome of our study highlights the variable localization, aggregation phenotype and toxicity of the studied α-synuclein variants. This underscores the importance of in-depth analysis of every disease-associated mutation which may result in variable cellular phenotype.


Assuntos
Agregados Proteicos , alfa-Sinucleína , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Sobrevivência Celular/genética , Mutação/genética , Estresse Oxidativo/genética , Agregados Proteicos/genética , Saccharomyces cerevisiae
6.
J Biol Chem ; 299(5): 104631, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36963494

RESUMO

For decades, sarcomeric myosin heavy chain proteins were assumed to be restricted to striated muscle where they function as molecular motors that contract muscle. However, MYH7b, an evolutionarily ancient member of this myosin family, has been detected in mammalian nonmuscle tissues, and mutations in MYH7b are linked to hereditary hearing loss in compound heterozygous patients. These mutations are the first associated with hearing loss rather than a muscle pathology, and because there are no homologous mutations in other myosin isoforms, their functional effects were unknown. We generated recombinant human MYH7b harboring the D515N or R1651Q hearing loss-associated mutation and studied their effects on motor activity and structural and assembly properties, respectively. The D515N mutation had no effect on steady-state actin-activated ATPase rate or load-dependent detachment kinetics but increased actin sliding velocity because of an increased displacement during the myosin working stroke. Furthermore, we found that the D515N mutation caused an increase in the proportion of myosin heads that occupy the disordered-relaxed state, meaning more myosin heads are available to interact with actin. Although we found no impact of the R1651Q mutation on myosin rod secondary structure or solubility, we observed a striking aggregation phenotype when this mutation was introduced into nonmuscle cells. Our results suggest that each mutation independently affects MYH7b function and structure. Together, these results provide the foundation for further study of a role for MYH7b outside the sarcomere.


Assuntos
Perda Auditiva , Cadeias Pesadas de Miosina , Animais , Humanos , Camundongos , Actinas/metabolismo , Linhagem Celular , Chlorocebus aethiops , Células COS , Perda Auditiva/genética , Perda Auditiva/fisiopatologia , Cinética , Mutação , Cadeias Pesadas de Miosina/genética , Cadeias Pesadas de Miosina/metabolismo , Agregados Proteicos/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
7.
FASEB J ; 37(1): e22702, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36520044

RESUMO

Neurodegenerative diseases result from the interplay of abnormal gene expression and various pathological factors. Therefore, a disease-specific integrative genetic approach is required to understand the complexities and causes of target diseases. Recent studies have identified the correlation between genes encoding several transmembrane proteins, such as the cluster of differentiation (CD) and Alzheimer's disease (AD) pathogenesis. In this study, CD48 and CD40 gene expression in AD, a neurodegenerative disease, was analyzed to infer this link. Total RNA sequencing was performed using an Alzheimer's disease mouse model brain and blood, and gene expression was determined using a genome-wide association study (GWAS). We observed a marked elevation of CD48 and CD40 genes in Alzheimer's disease. Indeed, the upregulation of both CD48 and CD40 genes was significantly increased in the severe Alzheimer's disease group. With the elevation of CD48 and CD40 genes in Alzheimer's disease, associations of protein levels were also markedly increased in tissues. In addition, overexpression of CD48 and CD40 genes triggered tau aggregation, and co-expression of these genes accelerated aggregation. The nuclear factor kappa B (NF-ĸB) signaling pathway was enriched by CD48 and CD40 gene expression: it was also associated with tau pathology. Our data suggested that the CD48 and CD40 genes are novel AD-related genes, and this approach may be useful as a diagnostic or therapeutic target for the disease.


Assuntos
Doença de Alzheimer , Antígenos CD40 , Antígeno CD48 , Agregados Proteicos , Proteínas tau , Animais , Camundongos , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Antígenos CD40/genética , Antígenos CD40/metabolismo , Antígeno CD48/genética , Antígeno CD48/metabolismo , Expressão Gênica , Estudo de Associação Genômica Ampla , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/metabolismo , Agregados Proteicos/genética , Agregados Proteicos/fisiologia , Proteínas tau/genética , Proteínas tau/metabolismo
8.
Int J Mol Sci ; 23(22)2022 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-36430707

RESUMO

Sorsby fundus dystrophy (SFD) is an autosomal dominant macular disorder caused by mutations in tissue Inhibitor of the metalloproteinase-3 (TIMP3) gene with the onset of symptoms including choroidal neovascularization as early as the second decade of life. We have previously reported that wild-type TIMP3 is an endogenous angiogenesis inhibitor that inhibits Vascular Endothelial Growth Factor (VEGF)-mediated signaling in endothelial cells. In contrast, SFD-related S179C-TIMP3 when expressed in endothelial cells, does not have angiogenesis-inhibitory properties. To evaluate if this is a common feature of TIMP3 mutants associated with SFD, we examined and compared endothelial cells expressing S179C, Y191C and S204C TIMP3 mutants for their angiogenesis-inhibitory function. Western blot analysis, zymography and reverse zymography and migration assays were utilized to evaluate TIMP3 protein, Matrix Metalloproteinase (MMP) and MMP inhibitory activity, VEGF signaling and in vitro migration in endothelial cells expressing (VEGF receptor-2 (VEGFR-2) and wild-type TIMP3 or mutant-TIMP3. We demonstrate that mutant S179C, Y191C- and S204C-TIMP3 all show increased glycosylation and multimerization/aggregation of the TIMP3 protein. In addition, endothelial cells expressing TIMP3 mutations show increased angiogenic activities and elevated VEGFR-2. Removal of N-glycosylation by mutation of Asn184, the only potential N-glycosylation site in mutant TIMP3, resulted in increased aggregation of TIMP3, further upregulation of VEGFR-2, VEGF-induced phosphorylation of VEGFR2 and VEGF-mediated migration concomitant with reduced MMP inhibitory activity. These results suggest that even though mutant TIMP3 proteins are more glycosylated, post-translational deglycosylation may play a critical role in the aggregation of mutant TIMP3 and contribute to the pathogenesis of SFD. The identification of factors that might contribute to changes in the glycome of patients with SFD will be useful. Future studies will evaluate whether variations in the glycosylation of mutant TIMP3 proteins are contributing to the severity of the disease.


Assuntos
Degeneração Macular , Inibidor Tecidual de Metaloproteinase-3 , Receptor 2 de Fatores de Crescimento do Endotélio Vascular , Humanos , Células Endoteliais/metabolismo , Metaloproteinases da Matriz/metabolismo , Inibidor Tecidual de Metaloproteinase-3/genética , Inibidor Tecidual de Metaloproteinase-3/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Fatores de Crescimento do Endotélio Vascular/metabolismo , Glicosilação , Degeneração Macular/genética , Degeneração Macular/metabolismo , Agregados Proteicos/genética , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo
9.
J Mol Biol ; 434(23): 167859, 2022 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-36270580

RESUMO

Fibrillar aggregates of the α-synuclein (αS) protein are the hallmark of Parkinson's Disease and related neurodegenerative disorders. Characterization of the effects of mutations and post-translational modifications (PTMs) on the αS aggregation rate can provide insight into the mechanism of fibril formation, which remains elusive in spite of intense study. A comprehensive collection (375 examples) of mutant and PTM aggregation rate data measured using the fluorescent probe thioflavin T is presented, as well as a summary of the effects of fluorescent labeling on αS aggregation (20 examples). A curated set of 131 single mutant de novo aggregation experiments are normalized to wild type controls and analyzed in terms of structural data for the monomer and fibrillar forms of αS. These tabulated data serve as a resource to the community to help in interpretation of aggregation experiments and to potentially be used as inputs for computational models of aggregation.


Assuntos
Agregados Proteicos , Processamento de Proteína Pós-Traducional , alfa-Sinucleína , Humanos , alfa-Sinucleína/química , alfa-Sinucleína/genética , Amiloide/metabolismo , Mutação , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , Agregados Proteicos/genética
10.
Mol Metab ; 63: 101536, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35772643

RESUMO

OBJECTIVE: Mutations in the copper-zinc superoxide dismutase (SOD1) gene cause familial amyotrophic lateral sclerosis (ALS), a progressive fatal neuromuscular disease characterized by motor neurons death and severe skeletal muscle degeneration. However, there is no effective treatment for this debilitating disease, since the underlying cause for the pathogenesis remains poorly understood. Here, we investigated a role of acyl-CoA:lysocardiolipin acyltransferase 1 (ALCAT1), an acyltransferase that promotes mitochondrial dysfunction in age-related diseases by catalyzing pathological remodeling of cardiolipin, in promoting the development of ALS in the SOD1G93A transgenic mice. METHODS: Using SOD1G93A transgenic mice with targeted deletion of the ALCAT1 gene and treated with Dafaglitapin (Dafa), a very potent and highly selective ALCAT1 inhibitor, we determined whether ablation or pharmaceutical inhibition of ALCAT1 by Dafa would mitigate ALS and the underlying pathogenesis by preventing pathological remodeling of cardiolipin, oxidative stress, and mitochondrial dysfunction by multiple approaches, including lifespan analysis, behavioral tests, morphological and functional analysis of skeletal muscle, electron microscopic and Seahorse analysis of mitochondrial morphology and respiration, western blot analysis of the SOD1G93A protein aggregation, and lipidomic analysis of cardiolipin content and acyl composition in mice spinal cord. RESULTS: ALCAT1 protein expression is potently upregulated in the skeletal muscle of the SOD1G93A mice. Consequently, ablation or pharmacological inhibition of ALCAT1 by Dafa attenuates motor neuron dysfunction, neuronal inflammation, and skeletal muscle atrophy in SOD1G93A mice by preventing SOD1G93A protein aggregation, mitochondrial dysfunction, and pathological CL remodeling, leading to moderate extension of lifespan in the SOD1G93A transgenic mice. CONCLUSIONS: ALCAT1 promotes the development of ALS by linking SOD1G93A protein aggregation to mitochondrial dysfunction, implicating Dafa as a potential treatment for this debilitating disorder.


Assuntos
Aciltransferases , Esclerose Amiotrófica Lateral , Agregados Proteicos , Superóxido Dismutase-1 , Aciltransferases/genética , Aciltransferases/metabolismo , Esclerose Amiotrófica Lateral/tratamento farmacológico , Esclerose Amiotrófica Lateral/genética , Esclerose Amiotrófica Lateral/metabolismo , Animais , Cardiolipinas/uso terapêutico , Modelos Animais de Doenças , Camundongos , Camundongos Transgênicos , Agregados Proteicos/genética , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Superóxido Dismutase/uso terapêutico , Superóxido Dismutase-1/genética , Superóxido Dismutase-1/metabolismo , Superóxido Dismutase-1/uso terapêutico
11.
Gene ; 809: 146034, 2022 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-34688816

RESUMO

We previously reported the function of Rbs1 protein in RNA polymerase III complex assembly via interactions with both, proteins and mRNAs. Rbs1 is a poly(A)-binding protein. The R3H domain in Rbs1 is required for mRNA interactions. The present study utilized the results of a genome-wide analysis of RNA binding by Rbs1 to show a direct interaction between Rbs1 with the 5'-untranslated region (5'-UTR) in PCL5 mRNA. By examining Pcl5 protein levels, we found that Rbs1 overproduction inhibited the translation of PCL5 mRNA. Pcl5 is a cyclin that is associated with Pho85 kinase, which is involved in the degradation of Gcn4 transcription factor. Consequently, lower levels of Pcl5 that resulted from Rbs1 overproduction increased the Gcn4 response. The functional R3H domain in Rbs1 was required for the downregulation of Pcl5 translation and increase in the Gcn4 response, thus validating a regulatory mechanism that relies on the interaction between Rbs1 and the 5'-UTR in PCL5 mRNA. Rbs1 protein was further characterized by microscopy, which identified single Rbs1 assemblies in part of the cell population. The presence of Rbs1 aggregates was confirmed by the fractionation of cellular extracts. Altogether, our results suggest a more general role of Rbs1 in regulating cellular metabolism beyond the assembly of RNA polymerase III.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/fisiologia , Regiões 5' não Traduzidas , Fatores de Transcrição de Zíper de Leucina Básica/genética , Ciclinas/genética , Ciclinas/metabolismo , Regulação Fúngica da Expressão Gênica , Complexos Multiproteicos/metabolismo , Agregados Proteicos/genética , RNA Polimerase III/metabolismo , RNA Mensageiro/genética , Proteínas de Saccharomyces cerevisiae/genética
12.
Cells ; 10(12)2021 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-34943956

RESUMO

The lack of an animal model or an in vitro model limits experimental options for studying temporal molecular events in pseudoexfoliation syndrome (PXF), an age related fibrillopathy causing trabecular meshwork damage and glaucoma. Our goal was to create a workable in vitro model of PXF using primary human TM (HTM) cell lines simulating human disease. Primary HTM cells harvested from healthy donors (n = 3), were exposed to various concentrations (5 ng/mL, 10 ng/mL, 15 ng/mL) of transforming growth factor-beta1 (TGF-ß1) for different time points. Morphological change of epithelial-mesenchymal transition (EMT) was analyzed by direct microscopic visualization and immunoblotting for EMT markers. Expression of pro-fibrotic markers were analyzed by quantitative RT-PCR and immunoblotting. Cell viability and death in treated cells was analyzed using FACS and MTT assay. Protein complex and amyloid aggregate formation was analyzed by Immunofluorescence of oligomer11 and amyloid beta fibrils. Effect of these changes with pharmacological inhibitors of canonical and non-canonical TGF pathway was done to analyze the pathway involved. The expression of pro-fibrotic markers was markedly upregulated at 10 ng/mL of TGF-ß1 exposure at 48-72 h of exposure with associated EMT changes at the same time point. Protein aggregates were seen maximally at these time points that were found to be localized around the nucleus and in the extracellular matrix (ECM). EMT and pro-fibrotic expression was differentially regulated by different canonical and non-canonical pathways suggesting complex regulatory mechanisms. This in vitro model using HTM cells simulated the main characteristics of human disease in PXF like pro-fibrotic gene expression, EMT, and aggregate formation.


Assuntos
Síndrome de Exfoliação/genética , Agregados Proteicos/genética , Malha Trabecular/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Peptídeos beta-Amiloides/efeitos adversos , Peptídeos beta-Amiloides/genética , Sobrevivência Celular/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Síndrome de Exfoliação/complicações , Síndrome de Exfoliação/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Glaucoma/complicações , Glaucoma/genética , Glaucoma/patologia , Humanos , Cultura Primária de Células/métodos , Agregados Proteicos/efeitos dos fármacos , Malha Trabecular/patologia , Fator de Crescimento Transformador beta1/genética
13.
Biochemistry ; 60(48): 3671-3675, 2021 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-34788013

RESUMO

Amyotrophic lateral sclerosis (ALS) is an incurable neurodegenerative disease that often occurs concurrently with frontotemporal dementia (FTD), another disorder involving progressive neuronal loss. ALS and FTD form a neurodegenerative continuum and share pathological and genetic features. Mutations in a multitude of genes have been linked to ALS/FTD, including FUS. The FUS protein aggregates and forms inclusions within affected neurons. However, the precise mechanisms connecting protein aggregation to neurotoxicity remain under intense investigation. Recent evidence points to the contribution of epigenetics to ALS/FTD. A main epigenetic mechanism involves the post-translational modification (PTM) of histone proteins. We have previously characterized the histone PTM landscape in a FUS ALS/FTD yeast model, finding a decreased level of acetylation on lysine residues 14 and 56 of histone H3. Here, we describe the first report of amelioration of disease phenotypes by controlling histone acetylation on specific modification sites. We show that inhibiting histone deacetylases, via treatment with trichostatin A, suppresses the toxicity associated with FUS overexpression in yeast by preserving the levels of H3K56ac and H3K14ac without affecting the expression or aggregation of FUS. Our data raise the novel hypothesis that the toxic effect of protein aggregation in neurodegeneration is related to its association with altered histone marks. Altogether, we demonstrate the ability to counter the repercussions of protein aggregation on cell survival by preventing specific histone modification changes. Our findings launch a novel mechanistic framework that will enable alternative therapeutic approaches for ALS/FTD and other neurodegenerative diseases.


Assuntos
Esclerose Amiotrófica Lateral/genética , Demência Frontotemporal/genética , Histonas/metabolismo , Ácidos Hidroxâmicos/farmacologia , Proteína FUS de Ligação a RNA/genética , Acetilação/efeitos dos fármacos , Esclerose Amiotrófica Lateral/tratamento farmacológico , Epigênese Genética , Demência Frontotemporal/tratamento farmacológico , Código das Histonas/genética , Histonas/genética , Humanos , Mutação/genética , Neurônios/efeitos dos fármacos , Neurônios/patologia , Agregados Proteicos/genética , Agregação Patológica de Proteínas/genética , Saccharomyces cerevisiae/genética
14.
Int J Mol Sci ; 22(21)2021 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-34768745

RESUMO

Insoluble protein aggregates with fibrillar morphology called amyloids and ß-barrel proteins both share a ß-sheet-rich structure. Correctly folded ß-barrel proteins can not only function in monomeric (dimeric) form, but also tend to interact with one another-followed, in several cases, by formation of higher order oligomers or even aggregates. In recent years, findings proving that ß-barrel proteins can adopt cross-ß amyloid folds have emerged. Different ß-barrel proteins were shown to form amyloid fibrils in vitro. The formation of functional amyloids in vivo by ß-barrel proteins for which the amyloid state is native was also discovered. In particular, several prokaryotic and eukaryotic proteins with ß-barrel domains were demonstrated to form amyloids in vivo, where they participate in interspecies interactions and nutrient storage, respectively. According to recent observations, despite the variety of primary structures of amyloid-forming proteins, most of them can adopt a conformational state with the ß-barrel topology. This state can be intermediate on the pathway of fibrillogenesis ("on-pathway state"), or can be formed as a result of an alternative assembly of partially unfolded monomers ("off-pathway state"). The ß-barrel oligomers formed by amyloid proteins possess toxicity, and are likely to be involved in the development of amyloidoses, thus representing promising targets for potential therapy of these incurable diseases. Considering rapidly growing discoveries of the amyloid-forming ß-barrels, we may suggest that their real number and diversity of functions are significantly higher than identified to date, and represent only "the tip of the iceberg". Here, we summarize the data on the amyloid-forming ß-barrel proteins, their physicochemical properties, and their biological functions, and discuss probable means and consequences of the amyloidogenesis of these proteins, along with structural relationships between these two widespread types of ß-folds.


Assuntos
Amiloide/fisiologia , Agregados Proteicos/fisiologia , Conformação Proteica em Folha beta/fisiologia , Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Proteínas Amiloidogênicas/metabolismo , Amiloidose/metabolismo , Humanos , Simulação de Dinâmica Molecular , Agregados Proteicos/genética
15.
Biochem Biophys Res Commun ; 582: 8-15, 2021 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-34678594

RESUMO

Gap junction beta 1 (GJB1) is the pathogenic gene of X-linked Charcot-Marie-Tooth type 1 (CMTX1), a rare hereditary sensorimotor neuropathy. However, different mutations of GJB1 result in heterogeneous clinical manifestations with only some mutations leading to central nervous system involvement. We previously reported two GJB1 missense mutations: one novel mutation (c.212T > G) found in a CMTX1 family that only manifested as peripheral neuropathy, and another previously reported mutation GJB1(c.311A > C) leading to involvement of the peripheral nerves and cerebral white matter. However, the mechanism by which GJB1 mutations lead to CMTX1 has not been fully characterized. Here, we generated Schwann cells and primary cultured oligodendrocytes with these two mutations, resulting in the Cx32I71S (GJB1 c.212T > G) and Cx32K104T (GJB1 c.311A > C) mutants, to analyze the pathogenic mechanism using cytology, molecular biology, and electrophysiological methods. Both mutants showed abnormal endoplasmic reticulum aggregation, especially the Cx32K104T mutant, leading to an increase in endoplasmic reticulum stress, resulting in apoptosis. Furthermore, whole-cell patch clamp experiments in oligodendrocytes revealed that the Cx32K104T mutant reduced the cell membrane potential and inwardly rectifying potassium currents, which may be a vital element for central involvement. Therefore, our results may provide a new perspective for understanding the pathogenesis of CMTX1.


Assuntos
Doença de Charcot-Marie-Tooth/genética , Conexinas/genética , Mutação de Sentido Incorreto , Oligodendroglia/metabolismo , Potássio/metabolismo , Células de Schwann/metabolismo , Animais , Apoptose/genética , Linhagem Celular , Doença de Charcot-Marie-Tooth/metabolismo , Doença de Charcot-Marie-Tooth/patologia , Conexinas/deficiência , Retículo Endoplasmático/genética , Retículo Endoplasmático/metabolismo , Estresse do Retículo Endoplasmático/genética , Expressão Gênica , Humanos , Ativação do Canal Iônico , Potenciais da Membrana/fisiologia , Modelos Biológicos , Oligodendroglia/patologia , Técnicas de Patch-Clamp , Cultura Primária de Células , Agregados Proteicos/genética , Ratos , Ratos Sprague-Dawley , Células de Schwann/patologia
16.
Biomolecules ; 11(10)2021 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-34680054

RESUMO

Abnormal accumulation of aggregated α-synuclein (α-Syn) is seen in a variety of neurodegenerative diseases, including Parkinson's disease (PD), multiple system atrophy (MSA), dementia with Lewy body (DLB), Parkinson's disease dementia (PDD), and even subsets of Alzheimer's disease (AD) showing Lewy-body-like pathology. These synucleinopathies exhibit differences in their clinical and pathological representations, reminiscent of prion disorders. Emerging evidence suggests that α-Syn self-assembles and polymerizes into conformationally diverse polymorphs in vitro and in vivo, similar to prions. These α-Syn polymorphs arising from the same precursor protein may exhibit strain-specific biochemical properties and the ability to induce distinct pathological phenotypes upon their inoculation in animal models. In this review, we discuss clinical and pathological variability in synucleinopathies and several aspects of α-Syn fibril polymorphism, including the existence of high-resolution molecular structures and brain-derived strains. The current review sheds light on the recent advances in delineating the structure-pathogenic relationship of α-Syn and how diverse α-Syn molecular polymorphs contribute to the existing clinical heterogeneity in synucleinopathies.


Assuntos
Amiloide/genética , Encéfalo/metabolismo , Agregados Proteicos/genética , alfa-Sinucleína/genética , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Amiloide/ultraestrutura , Encéfalo/patologia , Humanos , Corpos de Lewy/genética , Corpos de Lewy/patologia , Atrofia de Múltiplos Sistemas/genética , Atrofia de Múltiplos Sistemas/patologia , Doença de Parkinson/genética , Doença de Parkinson/patologia , Doenças Priônicas/genética , Doenças Priônicas/patologia , alfa-Sinucleína/ultraestrutura
17.
Biomolecules ; 11(10)2021 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-34680141

RESUMO

Parkinson's Disease (PD) is a highly prevalent neurodegenerative disease among older adults. PD neuropathology is marked by the progressive loss of the dopaminergic neurons of the substantia nigra pars compacta and the widespread accumulation of misfolded intracellular α-synuclein (α-syn). Genetic mutations and post-translational modifications, such as α-syn phosphorylation, have been identified among the multiple factors supporting α-syn accrual during PD. A decline in the clearance capacity of the ubiquitin-proteasome and the autophagy-lysosomal systems, together with mitochondrial dysfunction, have been indicated as major pathophysiological mechanisms of PD neurodegeneration. The accrual of misfolded α-syn aggregates into soluble oligomers, and the generation of insoluble fibrils composing the core of intraneuronal Lewy bodies and Lewy neurites observed during PD neurodegeneration, are ignited by the overproduction of reactive oxygen species (ROS). The ROS activate the α-syn aggregation cascade and, together with the Lewy bodies, promote neurodegeneration. However, the molecular pathways underlying the dynamic evolution of PD remain undeciphered. These gaps in knowledge, together with the clinical heterogeneity of PD, have hampered the identification of the biomarkers that may be used to assist in diagnosis, treatment monitoring, and prognostication. Herein, we illustrate the main pathways involved in PD pathogenesis and discuss their possible exploitation for biomarker discovery.


Assuntos
Mitocôndrias/genética , Doença de Parkinson/genética , Deficiências na Proteostase/genética , alfa-Sinucleína/genética , Biomarcadores/metabolismo , Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/patologia , Humanos , Corpos de Lewy/genética , Corpos de Lewy/patologia , Mitocôndrias/patologia , Doenças Neuroinflamatórias/genética , Doenças Neuroinflamatórias/patologia , Doença de Parkinson/patologia , Agregados Proteicos/genética , Deficiências na Proteostase/patologia , Espécies Reativas de Oxigênio/metabolismo
18.
Biochemistry ; 60(43): 3213-3222, 2021 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-34648275

RESUMO

Glycine-rich regions feature prominently in intrinsically disordered regions (IDRs) of proteins that drive phase separation and the regulated formation of membraneless biomolecular condensates. Interestingly, the Gly-rich IDRs seldom feature poly-Gly tracts. The protein fused in sarcoma (FUS) is an exception. This protein includes two 10-residue poly-Gly tracts within the prion-like domain (PLD) and at the interface between the PLD and the RNA binding domain. Poly-Gly tracts are known to be highly insoluble, being potent drivers of self-assembly into solid-like fibrils. Given that the internal concentrations of FUS and FUS-like molecules cross the high micromolar and even millimolar range within condensates, we reasoned that the intrinsic insolubility of poly-Gly tracts might be germane to emergent fluid-to-solid transitions within condensates. To assess this possibility, we characterized the concentration-dependent self-assembly for three non-overlapping 25-residue Gly-rich peptides derived from FUS. Two of the three peptides feature 10-residue poly-Gly tracts. These peptides form either long fibrils based on twisted ribbon-like structures or self-supporting gels based on physical cross-links of fibrils. Conversely, the peptide with similar Gly contents but lacking a poly-Gly tract does not form fibrils or gels. Instead, it remains soluble across a wide range of concentrations. Our findings highlight the ability of poly-Gly tracts within IDRs that drive phase separation to undergo self-assembly. We propose that these tracts are likely to contribute to nucleation of fibrillar solids within dense condensates formed by FUS.


Assuntos
Glicina/metabolismo , Peptídeos/química , Proteína FUS de Ligação a RNA/metabolismo , Condensados Biomoleculares/metabolismo , Proteínas Intrinsicamente Desordenadas/metabolismo , Microscopia Eletrônica de Transmissão/métodos , Peptídeos/metabolismo , Agregados Proteicos/genética , Agregados Proteicos/fisiologia , Domínios Proteicos/fisiologia , Proteína FUS de Ligação a RNA/genética , Proteína FUS de Ligação a RNA/fisiologia , Espectroscopia de Infravermelho com Transformada de Fourier/métodos
19.
Nat Commun ; 12(1): 5338, 2021 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-34504072

RESUMO

Molecular chaperones, including Hsp70/J-domain protein (JDP) families, play central roles in binding substrates to prevent their aggregation. How JDPs select different conformations of substrates remains poorly understood. Here, we report an interaction between the JDP DnaJC7 and tau that efficiently suppresses tau aggregation in vitro and in cells. DnaJC7 binds preferentially to natively folded wild-type tau, but disease-associated mutants in tau reduce chaperone binding affinity. We identify that DnaJC7 uses a single TPR domain to recognize a ß-turn structural element in tau that contains the 275VQIINK280 amyloid motif. Wild-type tau, but not mutant, ß-turn structural elements can block full-length tau binding to DnaJC7. These data suggest DnaJC7 preferentially binds and stabilizes natively folded conformations of tau to prevent tau conversion into amyloids. Our work identifies a novel mechanism of tau aggregation regulation that can be exploited as both a diagnostic and a therapeutic intervention.


Assuntos
Amiloide/química , Proteínas de Choque Térmico/química , Chaperonas Moleculares/química , Agregados Proteicos/genética , Tauopatias/genética , Proteínas tau/química , Amiloide/antagonistas & inibidores , Amiloide/genética , Amiloide/metabolismo , Animais , Sítios de Ligação , Encéfalo/metabolismo , Encéfalo/patologia , Clonagem Molecular , Modelos Animais de Doenças , Expressão Gênica , Células HEK293 , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Camundongos , Modelos Moleculares , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Mutação , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Dobramento de Proteína , Domínios e Motivos de Interação entre Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidade por Substrato , Tauopatias/metabolismo , Tauopatias/patologia , Termodinâmica , Proteínas tau/genética , Proteínas tau/metabolismo
20.
Sci Rep ; 11(1): 17977, 2021 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-34504195

RESUMO

Huntington's disease (HD) is caused by a CAG trinucleotide repeat expansion in the first exon of the huntingtin (HTT) gene coding for the huntingtin (HTT) protein. The misfolding and consequential aggregation of CAG-expanded mutant HTT (mHTT) underpin HD pathology. Our interest in the life cycle of HTT led us to consider the development of high-affinity small-molecule binders of HTT oligomerized/amyloid-containing species that could serve as either cellular and in vivo imaging tools or potential therapeutic agents. We recently reported the development of PET tracers CHDI-180 and CHDI-626 as suitable for imaging mHTT aggregates, and here we present an in-depth pharmacological investigation of their binding characteristics. We have implemented an array of in vitro and ex vivo radiometric binding assays using recombinant HTT, brain homogenate-derived HTT aggregates, and brain sections from mouse HD models and humans post-mortem to investigate binding affinities and selectivity against other pathological proteins from indications such as Alzheimer's disease and spinocerebellar ataxia 1. Radioligand binding assays and autoradiography studies using brain homogenates and tissue sections from HD mouse models showed that CHDI-180 and CHDI-626 specifically bind mHTT aggregates that accumulate with age and disease progression. Finally, we characterized CHDI-180 and CHDI-626 regarding their off-target selectivity and binding affinity to beta amyloid plaques in brain sections and homogenates from Alzheimer's disease patients.


Assuntos
Proteína Huntingtina/metabolismo , Doença de Huntington/metabolismo , Tomografia por Emissão de Pósitrons/métodos , Agregados Proteicos/genética , Agregação Patológica de Proteínas/diagnóstico por imagem , Compostos Radiofarmacêuticos/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Autorradiografia/métodos , Encéfalo/metabolismo , Modelos Animais de Doenças , Humanos , Proteína Huntingtina/genética , Doença de Huntington/patologia , Imuno-Histoquímica/métodos , Camundongos , Camundongos Transgênicos , Radioisótopos de Nitrogênio/metabolismo , Traçadores Radioativos , Ensaio Radioligante/métodos , Proteínas Recombinantes/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...