Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 179(6): 1246-1248, 2019 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-31778650

RESUMO

Cells are protected from endoplasmic reticulum stress through the unfolded protein response (UPR). In this issue of Cell, Schinzel, Higuchi-Sanabria, Shalem et al., identify a mechanism that helps cells cope with ER stress but is independent of canonical UPR activation, instead involving the extracellular matrix hyaluronidase, TMEM2, as a signaling mediator.


Assuntos
Hialuronoglucosaminidase , Longevidade , Retículo Endoplasmático , Estresse do Retículo Endoplasmático , Homeostase , Resposta a Proteínas não Dobradas
2.
EMBO J ; 40(21): e107568, 2021 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-34617299

RESUMO

While aggregation-prone proteins are known to accelerate aging and cause age-related diseases, the cellular mechanisms that drive their cytotoxicity remain unresolved. The orthologous proteins MOAG-4, SERF1A, and SERF2 have recently been identified as cellular modifiers of such proteotoxicity. Using a peptide array screening approach on human amyloidogenic proteins, we found that SERF2 interacted with protein segments enriched in negatively charged and hydrophobic, aromatic amino acids. The absence of such segments, or the neutralization of the positive charge in SERF2, prevented these interactions and abolished the amyloid-promoting activity of SERF2. In protein aggregation models in the nematode worm Caenorhabditis elegans, protein aggregation and toxicity were suppressed by mutating the endogenous locus of MOAG-4 to neutralize charge. Our data indicate that MOAG-4 and SERF2 drive protein aggregation and toxicity by interactions with negatively charged segments in aggregation-prone proteins. Such charge interactions might accelerate primary nucleation of amyloid by initiating structural changes and by decreasing colloidal stability. Our study points at charge interactions between cellular modifiers and amyloidogenic proteins as potential targets for interventions to reduce age-related protein toxicity.


Assuntos
Amiloide/química , Proteínas Amiloidogênicas/química , Proteínas de Caenorhabditis elegans/química , Caenorhabditis elegans/genética , Peptídeos e Proteínas de Sinalização Intracelular/química , Proteínas do Tecido Nervoso/química , alfa-Sinucleína/química , Sequência de Aminoácidos , Amiloide/genética , Amiloide/metabolismo , Proteínas Amiloidogênicas/genética , Proteínas Amiloidogênicas/metabolismo , Animais , Sítios de Ligação , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Regulação da Expressão Gênica , Células HEK293 , Humanos , Interações Hidrofóbicas e Hidrofílicas , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Peptídeos/genética , Peptídeos/metabolismo , Agregados Proteicos , Análise Serial de Proteínas , Ligação Proteica , Transdução de Sinais , Eletricidade Estática , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
3.
Cell ; 142(4): 601-12, 2010 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-20723760

RESUMO

Fibrillar protein aggregates are the major pathological hallmark of several incurable, age-related, neurodegenerative disorders. These aggregates typically contain aggregation-prone pathogenic proteins, such as amyloid-beta in Alzheimer's disease and alpha-synuclein in Parkinson's disease. It is, however, poorly understood how these aggregates are formed during cellular aging. Here we identify an evolutionarily highly conserved modifier of aggregation, MOAG-4, as a positive regulator of aggregate formation in C. elegans models for polyglutamine diseases. Inactivation of MOAG-4 suppresses the formation of compact polyglutamine aggregation intermediates that are required for aggregate formation. The role of MOAG-4 in driving aggregation extends to amyloid-beta and alpha-synuclein and is evolutionarily conserved in its human orthologs SERF1A and SERF2. MOAG-4/SERF appears to act independently from HSF-1-induced molecular chaperones, proteasomal degradation, and autophagy. Our results suggest that MOAG-4/SERF regulates age-related proteotoxicity through a previously unexplored pathway, which will open up new avenues for research on age-related, neurodegenerative diseases.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Senescência Celular , Proteínas do Tecido Nervoso/metabolismo , Doenças Neurodegenerativas/metabolismo , Proteínas/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Proteínas de Caenorhabditis elegans/química , Linhagem Celular , Linhagem Celular Tumoral , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Proteínas do Tecido Nervoso/química , Peptídeos/metabolismo , Proteínas/química , alfa-Sinucleína/metabolismo
4.
Mol Cell ; 65(6): 1096-1108.e6, 2017 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-28306505

RESUMO

Protein aggregation is associated with age-related neurodegenerative disorders, such as Alzheimer's and polyglutamine diseases. As a causal relationship between protein aggregation and neurodegeneration remains elusive, understanding the cellular mechanisms regulating protein aggregation will help develop future treatments. To identify such mechanisms, we conducted a forward genetic screen in a C. elegans model of polyglutamine aggregation and identified the protein MOAG-2/LIR-3 as a driver of protein aggregation. In the absence of polyglutamine, MOAG-2/LIR-3 regulates the RNA polymerase III-associated transcription of small non-coding RNAs. This regulation is lost in the presence of polyglutamine, which mislocalizes MOAG-2/LIR-3 from the nucleus to the cytosol. We then show biochemically that MOAG-2/LIR-3 can also catalyze the aggregation of polyglutamine-expanded huntingtin. These results suggest that polyglutamine can induce an aggregation-promoting activity of MOAG-2/LIR-3 in the cytosol. The concept that certain aggregation-prone proteins can convert other endogenous proteins into drivers of aggregation and toxicity adds to the understanding of how cellular homeostasis can be deteriorated in protein misfolding diseases.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/enzimologia , Doenças Neurodegenerativas/enzimologia , Peptídeos/metabolismo , Agregados Proteicos , Agregação Patológica de Proteínas , RNA Polimerase III/metabolismo , Fatores de Transcrição/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Animais Geneticamente Modificados , Sítios de Ligação , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Núcleo Celular/enzimologia , Citosol/enzimologia , Modelos Animais de Doenças , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/patologia , Regiões Promotoras Genéticas , Ligação Proteica , Interferência de RNA , RNA Polimerase III/genética , Pequeno RNA não Traduzido/genética , Pequeno RNA não Traduzido/metabolismo , Fatores de Transcrição/genética , Transcrição Gênica
5.
Proc Natl Acad Sci U S A ; 114(6): E1009-E1017, 2017 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-28096355

RESUMO

The self-assembly of α-synuclein is closely associated with Parkinson's disease and related syndromes. We show that squalamine, a natural product with known anticancer and antiviral activity, dramatically affects α-synuclein aggregation in vitro and in vivo. We elucidate the mechanism of action of squalamine by investigating its interaction with lipid vesicles, which are known to stimulate nucleation, and find that this compound displaces α-synuclein from the surfaces of such vesicles, thereby blocking the first steps in its aggregation process. We also show that squalamine almost completely suppresses the toxicity of α-synuclein oligomers in human neuroblastoma cells by inhibiting their interactions with lipid membranes. We further examine the effects of squalamine in a Caenorhabditis elegans strain overexpressing α-synuclein, observing a dramatic reduction of α-synuclein aggregation and an almost complete elimination of muscle paralysis. These findings suggest that squalamine could be a means of therapeutic intervention in Parkinson's disease and related conditions.


Assuntos
Agregados Proteicos/efeitos dos fármacos , Agregação Patológica de Proteínas/prevenção & controle , alfa-Sinucleína/química , Algoritmos , Sequência de Aminoácidos , Animais , Animais Geneticamente Modificados , Produtos Biológicos/química , Produtos Biológicos/farmacologia , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Linhagem Celular Tumoral , Colestanóis/química , Colestanóis/farmacologia , Humanos , Lipídeos de Membrana/química , Lipídeos de Membrana/metabolismo , Estrutura Molecular , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Paresia/genética , Paresia/metabolismo , Paresia/prevenção & controle , Doença de Parkinson/metabolismo , Ligação Proteica/efeitos dos fármacos , Multimerização Proteica/efeitos dos fármacos , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
6.
J Neurochem ; 151(5): 656-668, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31376341

RESUMO

The kynurenine (Kyn) pathway, which regulates neuroinflammation and N-methyl-d-aspartate receptor activation, is implicated in Parkinson's disease (PD) and Alzheimer's disease (AD). Age-related changes in Kyn metabolism and altered cerebral Kyn uptake along large neutral amino acid transporters, could contribute to these diseases. To gain further insight into the role and prognostic potential of the Kyn pathway in PD and AD, we investigated systemic and cerebral Kyn metabolite production and estimations of their transporter-mediated uptake in the brain. Kyn metabolites and large neutral amino acids were retrospectively measured in serum and cerebrospinal fluid (CSF) of clinically well-characterized PD patients (n = 33), AD patients (n = 33), and age-matched controls (n = 39) using solid-phase extraction-liquid chromatographic-tandem mass spectrometry. Aging was disease independently associated with increased Kyn, kynurenic acid and quinolinic acid in serum and CSF. Concentrations of kynurenic acid were reduced in CSF of PD and AD patients (p = 0.001; p = 0.002) but estimations of Kyn brain uptake did not differ between diseased and controls. Furthermore, serum Kyn and quinolinic acid levels strongly correlated with their respective content in CSF and Kyn in serum negatively correlated with AD disease severity (p = 0.002). Kyn metabolites accumulated with aging in serum and CSF similarly in PD patients, AD patients, and control subjects. In contrast, kynurenic acid was strongly reduced in CSF of PD and AD patients. Differential transporter-mediated Kyn uptake is unlikely to majorly contribute to these cerebral Kyn pathway disturbances. We hypothesize that the combination of age- and disease-specific changes in cerebral Kyn pathway activity could contribute to reduced neurogenesis and increased excitotoxicity in neurodegenerative disease.


Assuntos
Doença de Alzheimer/metabolismo , Cinurenina/metabolismo , Doença de Parkinson/metabolismo , Idoso , Envelhecimento/metabolismo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
7.
J Biol Chem ; 292(20): 8269-8278, 2017 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-28336532

RESUMO

Aberrant protein aggregation underlies a variety of age-related neurodegenerative disorders, including Alzheimer's and Parkinson's diseases. Little is known, however, about the molecular mechanisms that modulate the aggregation process in the cellular environment. Recently, MOAG-4/SERF has been identified as a class of evolutionarily conserved proteins that positively regulates aggregate formation. Here, by using nuclear magnetic resonance (NMR) spectroscopy, we examine the mechanism of action of MOAG-4 by characterizing its interaction with α-synuclein (α-Syn). NMR chemical shift perturbations demonstrate that a positively charged segment of MOAG-4 forms a transiently populated α-helix that interacts with the negatively charged C terminus of α-Syn. This process interferes with the intramolecular interactions between the N- and C-terminal regions of α-Syn, resulting in the protein populating less compact forms and aggregating more readily. These results provide a compelling example of the complex competition between molecular and cellular factors that protect against protein aggregation and those that promote it.


Assuntos
Proteínas de Caenorhabditis elegans/química , Caenorhabditis elegans/química , Proteínas do Tecido Nervoso/química , Agregados Proteicos , alfa-Sinucleína/química , Doença de Alzheimer , Animais , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Humanos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Doença de Parkinson , Eletricidade Estática , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
9.
Cell Mol Life Sci ; 72(21): 4027-47, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26190021

RESUMO

Protein homeostasis is fundamental for cell function and survival, because proteins are involved in all aspects of cellular function, ranging from cell metabolism and cell division to the cell's response to environmental challenges. Protein homeostasis is tightly regulated by the synthesis, folding, trafficking and clearance of proteins, all of which act in an orchestrated manner to ensure proteome stability. The protein quality control system is enhanced by stress response pathways, which take action whenever the proteome is challenged by environmental or physiological stress. Aging, however, damages the proteome, and such proteome damage is thought to be associated with aging-related diseases. In this review, we discuss the different cellular processes that define the protein quality control system and focus on their role in protein conformational diseases. We highlight the power of using small organisms to model neurodegenerative diseases and how these models can be exploited to discover genetic modulators of protein aggregation and toxicity. We also link findings from small model organisms to the situation in higher organisms and describe how some of the genetic modifiers discovered in organisms such as worms are functionally conserved throughout evolution. Finally, we demonstrate that the non-coding genome also plays a role in maintaining protein homeostasis. In all, this review highlights the importance of protein and RNA homeostasis in neurodegenerative diseases.


Assuntos
Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/metabolismo , Proteínas/metabolismo , RNA não Traduzido/metabolismo , Doença de Alzheimer/fisiopatologia , Animais , Caenorhabditis elegans/fisiologia , Modelos Animais de Doenças , Drosophila , Homeostase/genética , Humanos , Doenças Neurodegenerativas/fisiopatologia , Doença de Parkinson/fisiopatologia , Conformação Proteica , Dobramento de Proteína , Proteínas/química , Proteínas/genética , Saccharomyces cerevisiae/fisiologia
10.
Biochim Biophys Acta ; 1842(10): 1951-1959, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24525026

RESUMO

Caenorhabditis elegans comprises unique features that make it an attractive model organism in diverse fields of biology. Genetic screens are powerful to identify genes and C. elegans can be customized to forward or reverse genetic screens and to establish gene function. These genetic screens can be applied to "humanized" models of C. elegans for neurodegenerative diseases, enabling for example the identification of genes involved in protein aggregation, one of the hallmarks of these diseases. In this review, we will describe the genetic screens employed in C. elegans and how these can be used to understand molecular processes involved in neurodegenerative and other human diseases. This article is part of a Special Issue entitled: From Genome to Function.

11.
Proc Natl Acad Sci U S A ; 109(37): 14912-7, 2012 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-22927396

RESUMO

Toxicity of aggregation-prone proteins is thought to play an important role in aging and age-related neurological diseases like Parkinson and Alzheimer's diseases. Here, we identify tryptophan 2,3-dioxygenase (tdo-2), the first enzyme in the kynurenine pathway of tryptophan degradation, as a metabolic regulator of age-related α-synuclein toxicity in a Caenorhabditis elegans model. Depletion of tdo-2 also suppresses toxicity of other heterologous aggregation-prone proteins, including amyloid-ß and polyglutamine proteins, and endogenous metastable proteins that are sensors of normal protein homeostasis. This finding suggests that tdo-2 functions as a general regulator of protein homeostasis. Analysis of metabolite levels in C. elegans strains with mutations in enzymes that act downstream of tdo-2 indicates that this suppression of toxicity is independent of downstream metabolites in the kynurenine pathway. Depletion of tdo-2 increases tryptophan levels, and feeding worms with extra L-tryptophan also suppresses toxicity, suggesting that tdo-2 regulates proteotoxicity through tryptophan. Depletion of tdo-2 extends lifespan in these worms. Together, these results implicate tdo-2 as a metabolic switch of age-related protein homeostasis and lifespan. With TDO and Indoleamine 2,3-dioxygenase as evolutionarily conserved human orthologs of TDO-2, intervening with tryptophan metabolism may offer avenues to reducing proteotoxicity in aging and age-related diseases.


Assuntos
Envelhecimento/fisiologia , Homeostase/fisiologia , Triptofano Oxigenase/metabolismo , Triptofano/metabolismo , alfa-Sinucleína/toxicidade , Envelhecimento/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Animais Geneticamente Modificados , Caenorhabditis elegans , Cromatografia Líquida , Biologia Computacional , Primers do DNA/genética , Fertilidade/genética , Immunoblotting , Longevidade/genética , Peptídeos/metabolismo , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Espectrometria de Massas em Tandem , Triptofano/química , Triptofano Oxigenase/antagonistas & inibidores
12.
Cell Rep ; 43(5): 114204, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38748878

RESUMO

Amyotrophic lateral sclerosis can be caused by abnormal accumulation of TAR DNA-binding protein 43 (TDP-43) in the cytoplasm of neurons. Here, we use a C. elegans model for TDP-43-induced toxicity to identify the biological mechanisms that lead to disease-related phenotypes. By applying deep behavioral phenotyping and subsequent dissection of the neuromuscular circuit, we show that TDP-43 worms have profound defects in GABA neurons. Moreover, acetylcholine neurons appear functionally silenced. Enhancing functional output of repressed acetylcholine neurons at the level of, among others, G-protein-coupled receptors restores neurotransmission, but inefficiently rescues locomotion. Rebalancing the excitatory-to-inhibitory ratio in the neuromuscular system by simultaneous stimulation of the affected GABA- and acetylcholine neurons, however, not only synergizes the effects of boosting individual neurotransmitter systems, but instantaneously improves movement. Our results suggest that interventions accounting for the altered connectome may be more efficient in restoring motor function than those solely focusing on diseased neuron populations.


Assuntos
Caenorhabditis elegans , Proteínas de Ligação a DNA , Modelos Animais de Doenças , Animais , Caenorhabditis elegans/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/genética , Neurônios GABAérgicos/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Neurônios Motores/metabolismo , Locomoção , Transmissão Sináptica , Movimento , Neurônios Colinérgicos/metabolismo
13.
MicroPubl Biol ; 20232023.
Artigo em Inglês | MEDLINE | ID: mdl-37151215

RESUMO

Cytoplasmic inclusions consisting of transactive response DNA-binding protein 43 (TDP-43) are a key hallmark of TDP-43 proteinopathies like amyotrophic lateral sclerosis (ALS). Caenorhabditis elegans is considered a useful model for studying the molecular mechanisms underlying TDP-43 toxicity in vivo . Here, we assessed different neuronal systems through established behavioral assays and extended the phenotypic characterisation of a C. elegans model expressing wildtype human TDP-43 ( hTDP-43 ) pan-neuronally. Our data show that neuronal expression of hTDP-43 in C. elegans disrupts chemotaxis and decreases fecundity. The basal slowing response, on the other hand, appears to be preserved in the presence of hTDP-43.

14.
MicroPubl Biol ; 20232023.
Artigo em Inglês | MEDLINE | ID: mdl-37159576

RESUMO

Transactive response DNA binding-protein 43 (TDP-43) is a conserved RNA/DNA-binding protein with a role in RNA metabolism and homeostasis. Aberrant TDP-43 functioning has been considered a major culprit in amyotrophic lateral sclerosis (ALS). Caenorhabditis elegans can be used to phenocopy ALS in vivo . Since disrupted locomotion is a strong readout of toxicity, we examined multiple motor phenotypes of a C. elegans model expressing human wild-type TDP-43 ( hTDP-43 ) pan-neuronally. Our data reveal that impaired locomotion includes more than the common deficits in crawling capacity and the presence of early-onset paralysis. We show that reduced thrashing, abnormal coiling, and decreased pharyngeal pumping are also observed, in a temperature-dependent fashion.

15.
MicroPubl Biol ; 20232023.
Artigo em Inglês | MEDLINE | ID: mdl-37159575

RESUMO

Inclusions consisting of transactive response DNA-binding protein 43 (TDP-43) are a characteristic feature of amyotrophic lateral sclerosis (ALS). Caenorhabditis elegans has been instrumental in studying the underlying mechanisms of TDP-43 pathology. Here, we extend the possibilities of previous studies by examining a C. elegans model expressing human wild-type TDP-43 ( hTDP-43 ) pan-neuronally. We show that disease-related (hyper)phosphorylation and cytosolic localisation of hTDP-43 are present in hTDP-43 worms and that these features can be enhanced by adjusting the environmental temperature.

16.
Nat Cell Biol ; 4(10): 826-31, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12360295

RESUMO

Protein aggregation and the formation of inclusion bodies are hallmarks of the cytopathology of neurodegenerative diseases, including Huntington's disease, Amyotropic lateral sclerosis, Parkinson's disease and Alzheimer's disease. The cellular toxicity associated with protein aggregates has been suggested to result from the sequestration of essential proteins that are involved in key cellular events, such as transcription, maintenance of cell shape and motility, protein folding and protein degradation. Here, we use fluorescence imaging of living cells to show that polyglutamine protein aggregates are dynamic structures in which glutamine-rich proteins are tightly associated, but which exhibit distinct biophysical interactions. In contrast, the interaction between wild-type, but not mutant, Hsp70 exhibits rapid kinetics of association and dissociation similar to interactions between Hsp70 and thermally unfolded substrates. These studies provide new insights into the composite organization and formation of protein aggregates and show that molecular chaperones are not sequestered into aggregates, but are instead transiently associated.


Assuntos
Células Eucarióticas/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Corpos de Inclusão/metabolismo , Doenças Neurodegenerativas/metabolismo , Peptídeos/metabolismo , Animais , Proteínas de Bactérias , Compartimento Celular/fisiologia , Recuperação de Fluorescência Após Fotodegradação , Proteínas de Fluorescência Verde , Proteínas de Choque Térmico HSP70/genética , Células HeLa , Humanos , Corpos de Inclusão/genética , Proteínas Luminescentes , Substâncias Macromoleculares , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Doenças Neurodegenerativas/genética , Peptídeos/genética , Dobramento de Proteína , Proteínas Recombinantes de Fusão , Proteína de Ligação a TATA-Box/genética , Proteína de Ligação a TATA-Box/metabolismo
17.
Chemphyschem ; 12(3): 673-680, 2011 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-21308945

RESUMO

Misfolding and aggregation of amyloidogenic polypeptides lie at the root of many neurodegenerative diseases. Whilst protein aggregation can be readily studied in vitro by established biophysical techniques, direct observation of the nature and kinetics of aggregation processes taking place in vivo is much more challenging. We describe here, however, a Förster resonance energy transfer sensor that permits the aggregation kinetics of amyloidogenic proteins to be quantified in living systems by exploiting our observation that amyloid assemblies can act as energy acceptors for variants of fluorescent proteins. The observed lifetime reduction can be attributed to fluorescence energy transfer to intrinsic energy states associated with the growing amyloid species. Indeed, for a-synuclein, a protein whose aggregation is linked to Parkinson's disease, we have used this sensor to follow the kinetics of the self-association reactions taking place in vitro and in vivo and to reveal the nature of the ensuing aggregated species. Experiments were conducted in vitro, in cells in culture and in living Caenorhabditis elegans. For the latter the readout correlates directly with the appearance of a toxic phenotype. The ability to measure the appearance and development of pathogenic amyloid species in a living animal and the ability to relate such data to similar processes observed in vitro provides a powerful new tool in the study of the pathology of the family of misfolding disorders. Our study confirms the importance of the molecular environment in which aggregation reactions take place, highlighting similarities as well as differences between the processes occurring in vitro and in vivo, and their significance for defining the molecular physiology of the diseases with which they are associated.


Assuntos
Amiloide/química , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Caenorhabditis elegans/metabolismo , Linhagem Celular Tumoral , Transferência Ressonante de Energia de Fluorescência , Humanos , Cinética , Proteínas Luminescentes/química , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Microscopia de Fluorescência , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
18.
PLoS Genet ; 4(3): e1000027, 2008 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-18369446

RESUMO

Inclusions in the brain containing alpha-synuclein are the pathological hallmark of Parkinson's disease, but how these inclusions are formed and how this links to disease is poorly understood. We have developed a C. elegans model that makes it possible to monitor, in living animals, the formation of alpha-synuclein inclusions. In worms of old age, inclusions contain aggregated alpha- synuclein, resembling a critical pathological feature. We used genome-wide RNA interference to identify processes involved in inclusion formation, and identified 80 genes that, when knocked down, resulted in a premature increase in the number of inclusions. Quality control and vesicle-trafficking genes expressed in the ER/Golgi complex and vesicular compartments were overrepresented, indicating a specific role for these processes in alpha-synuclein inclusion formation. Suppressors include aging-associated genes, such as sir-2.1/SIRT1 and lagr-1/LASS2. Altogether, our data suggest a link between alpha-synuclein inclusion formation and cellular aging, likely through an endomembrane-related mechanism. The processes and genes identified here present a framework for further study of the disease mechanism and provide candidate susceptibility genes and drug targets for Parkinson's disease and other alpha-synuclein related disorders.


Assuntos
Envelhecimento/genética , Envelhecimento/metabolismo , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Corpos de Inclusão/metabolismo , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Animais , Animais Geneticamente Modificados , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sequência de Bases , Encéfalo/metabolismo , Primers do DNA/genética , Recuperação de Fluorescência Após Fotodegradação , Quinases de Receptores Acoplados a Proteína G/antagonistas & inibidores , Quinases de Receptores Acoplados a Proteína G/genética , Quinases de Receptores Acoplados a Proteína G/metabolismo , Deleção de Genes , Genes de Helmintos , Humanos , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Modelos Genéticos , Doença de Parkinson/etiologia , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , Interferência de RNA , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , alfa-Sinucleína/antagonistas & inibidores
19.
Front Cell Dev Biol ; 9: 637084, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33748125

RESUMO

Proteome damage plays a major role in aging and age-related neurodegenerative diseases. Under healthy conditions, molecular quality control mechanisms prevent toxic protein misfolding and aggregation. These mechanisms include molecular chaperones for protein folding, spatial compartmentalization for sequestration, and degradation pathways for the removal of harmful proteins. These mechanisms decline with age, resulting in the accumulation of aggregation-prone proteins that are harmful to cells. In the past decades, a variety of fast- and slow-aging model organisms have been used to investigate the biological mechanisms that accelerate or prevent such protein toxicity. In this review, we describe the most important mechanisms that are required for maintaining a healthy proteome. We describe how these mechanisms decline during aging and lead to toxic protein misassembly, aggregation, and amyloid formation. In addition, we discuss how optimized protein homeostasis mechanisms in long-living animals contribute to prolonging their lifespan. This knowledge might help us to develop interventions in the protein homeostasis network that delay aging and age-related pathologies.

20.
J Mol Cell Biol ; 13(4): 282-294, 2021 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-33386842

RESUMO

Misfolded α-synuclein is a major component of Lewy bodies, which are a hallmark of Parkinson's disease (PD). A large body of evidence shows that α-synuclein can aggregate into amyloid fibrils, but the relationship between α-synuclein self-assembly and Lewy body formation remains unclear. Here, we show, both in vitro and in a Caenorhabditis elegans model of PD, that α-synuclein undergoes liquid‒liquid phase separation by forming a liquid droplet state, which converts into an amyloid-rich hydrogel with Lewy-body-like properties. This maturation process towards the amyloid state is delayed in the presence of model synaptic vesicles in vitro. Taken together, these results suggest that the formation of Lewy bodies may be linked to the arrested maturation of α-synuclein condensates in the presence of lipids and other cellular components.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Corpos de Lewy/metabolismo , Doença de Parkinson/patologia , alfa-Sinucleína/metabolismo , Animais , Animais Geneticamente Modificados , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/genética , Modelos Animais de Doenças , Humanos , Corpos de Lewy/patologia , alfa-Sinucleína/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA