Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 420
Filtrar
1.
eNeuro ; 11(4)2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38565295

RESUMO

The accumulation of amyloid-ß (Aß) and hyperphosphorylated-tau (hp-tau) are two classical histopathological biomarkers in Alzheimer's disease (AD). However, their detailed interactions with the electrophysiological changes at the meso- and macroscale are not yet fully understood. We developed a mechanistic multiscale model of AD progression, linking proteinopathy to its effects on neural activity and vice-versa. We integrated a heterodimer model of prion-like protein propagation and a brain network model of Jansen-Rit neural masses derived from human neuroimaging data whose parameters varied due to neurotoxicity. Results showed that changes in inhibition guided the electrophysiological alterations found in AD, and these changes were mainly attributed to Aß effects. Additionally, we found a causal disconnection between cellular hyperactivity and interregional hypersynchrony contrary to previous beliefs. Finally, we demonstrated that early Aß and hp-tau depositions' location determine the spatiotemporal profile of the proteinopathy. The presented model combines the molecular effects of both Aß and hp-tau together with a mechanistic protein propagation model and network effects within a closed-loop model. This holds the potential to enlighten the interplay between AD mechanisms on various scales, aiming to develop and test novel hypotheses on the contribution of different AD-related variables to the disease evolution.


Assuntos
Doença de Alzheimer , Deficiências na Proteostase , Humanos , Doença de Alzheimer/patologia , Encéfalo/metabolismo , Proteínas tau/metabolismo , Peptídeos beta-Amiloides/metabolismo , Neuroimagem/métodos , Deficiências na Proteostase/metabolismo , Deficiências na Proteostase/patologia , Progressão da Doença
2.
ACS Chem Neurosci ; 15(7): 1533-1547, 2024 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-38507813

RESUMO

Neuroinflammation plays a crucial role in the development of neurodegenerative protein misfolding disorders. This category of progressive diseases includes, but is not limited to, Alzheimer's disease, Parkinson's disease, and prion diseases. Shared pathogenesis involves the accumulation of misfolded proteins, chronic neuroinflammation, and synaptic dysfunction, ultimately leading to irreversible neuronal loss, measurable cognitive deficits, and death. Presently, there are few to no effective treatments to halt the advancement of neurodegenerative diseases. We hypothesized that directly targeting neuroinflammation by downregulating the transcription factor, NF-κB, and the inflammasome protein, NLRP3, would be neuroprotective. To achieve this, we used a cocktail of RNA targeting therapeutics (SB_NI_112) shown to be brain-penetrant, nontoxic, and effective inhibitors of both NF-κB and NLRP3. We utilized a mouse-adapted prion strain as a model for neurodegenerative diseases to assess the aggregation of misfolded proteins, glial inflammation, neuronal loss, cognitive deficits, and lifespan. Prion-diseased mice were treated either intraperitoneally or intranasally with SB_NI_112. Behavioral and cognitive deficits were significantly protected by this combination of NF-κB and NLRP3 downregulators. Treatment reduced glial inflammation, protected against neuronal loss, prevented spongiotic change, rescued cognitive deficits, and significantly lengthened the lifespan of prion-diseased mice. We have identified a nontoxic, systemic pharmacologic that downregulates NF-κB and NLRP3, prevents neuronal death, and slows the progression of neurodegenerative diseases. Though mouse models do not always predict human patient success and the study was limited due to sample size and number of dosing methods utilized, these findings serve as a proof of principle for continued translation of the therapeutic SB_NI_112 for prion disease and other neurodegenerative diseases. Based on the success in a murine prion model, we will continue testing SB_NI_112 in a variety of neurodegenerative disease models, including Alzheimer's disease and Parkinson's disease.


Assuntos
Doença de Alzheimer , Doenças Neurodegenerativas , Doença de Parkinson , Doenças Priônicas , Príons , Deficiências na Proteostase , Humanos , Camundongos , Animais , Doenças Neurodegenerativas/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , NF-kappa B/metabolismo , Doença de Alzheimer/metabolismo , Doenças Neuroinflamatórias , Regulação para Baixo , Doença de Parkinson/metabolismo , Neurônios/metabolismo , Doenças Priônicas/tratamento farmacológico , Doenças Priônicas/metabolismo , Príons/metabolismo , Inflamação/metabolismo , Deficiências na Proteostase/tratamento farmacológico , Deficiências na Proteostase/metabolismo
3.
ACS Chem Neurosci ; 15(5): 898-908, 2024 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-38407017

RESUMO

Protein misfolding has been extensively studied in the context of neurodegenerative disorders and systemic amyloidoses. Due to misfolding and aggregation of proteins being highly heterogeneous and generating a variety of structures, a growing body of evidence illustrates numerous ways how the aggregates contribute to progression of diseases such as Alzheimer's disease, Parkinson's disease, and prion disorders. Different misfolded species of the same protein, commonly referred to as strains, appear to play a significant role in shaping the disease clinical phenotype and clinical progression. The distinct toxicity profiles of various misfolded proteins underscore their importance. Current diagnostics struggle to differentiate among these strains early in the disease course. This review explores the potential of spectral fluorescence approaches to illuminate the complexities of protein misfolding pathology and discusses the applications of advanced spectral methods in the detection and characterization of protein misfolding disorders. By examining spectrally variable probes, current data analysis approaches, and important considerations for the use of these techniques, this review aims to provide an overview of the progress made in this field and highlights directions for future research.


Assuntos
Amiloidose , Doenças Neurodegenerativas , Doenças Priônicas , Deficiências na Proteostase , Humanos , Fluorescência , Deficiências na Proteostase/metabolismo , Amiloidose/metabolismo , Doenças Priônicas/metabolismo , Doenças Neurodegenerativas/metabolismo , Dobramento de Proteína
4.
Alzheimers Res Ther ; 15(1): 170, 2023 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-37821965

RESUMO

The glymphatic system is a crucial component in preserving brain homeostasis by facilitating waste clearance from the central nervous system (CNS). Aquaporin-4 (AQP4) water channels facilitate the continuous interchange between cerebrospinal fluid and brain interstitial fluid by convective flow movement. This flow is responsible for guiding proteins and metabolites away from the CNS. Proteinopathies are neurological conditions characterized by the accumulation of aggregated proteins or peptides in the brain. In Alzheimer's disease (AD), the deposition of amyloid-ß (Aß) peptides causes the formation of senile plaques. This accumulation has been hypothesized to be a result of the imbalance between Aß production and clearance. Recent studies have shown that an extended form of AQP4 increases Aß clearance from the brain. In this mini-review, we present a summary of these findings and explore the potential for future therapeutic strategies aiming to boost waste clearance in AD.


Assuntos
Doença de Alzheimer , Deficiências na Proteostase , Humanos , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Aquaporina 4/metabolismo , Encéfalo/metabolismo , Isoformas de Proteínas/metabolismo , Deficiências na Proteostase/metabolismo
5.
Int J Mol Sci ; 24(10)2023 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-37239937

RESUMO

The accumulation of protein aggregates is the hallmark of many neurodegenerative diseases. The dysregulation of protein homeostasis (or proteostasis) caused by acute proteotoxic stresses or chronic expression of mutant proteins can lead to protein aggregation. Protein aggregates can interfere with a variety of cellular biological processes and consume factors essential for maintaining proteostasis, leading to a further imbalance of proteostasis and further accumulation of protein aggregates, creating a vicious cycle that ultimately leads to aging and the progression of age-related neurodegenerative diseases. Over the long course of evolution, eukaryotic cells have evolved a variety of mechanisms to rescue or eliminate aggregated proteins. Here, we will briefly review the composition and causes of protein aggregation in mammalian cells, systematically summarize the role of protein aggregates in the organisms, and further highlight some of the clearance mechanisms of protein aggregates. Finally, we will discuss potential therapeutic strategies that target protein aggregates in the treatment of aging and age-related neurodegenerative diseases.


Assuntos
Doenças Neurodegenerativas , Deficiências na Proteostase , Animais , Humanos , Agregados Proteicos , Proteostase , Deficiências na Proteostase/metabolismo , Doenças Neurodegenerativas/metabolismo , Proteínas/genética , Proteínas/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Mamíferos/metabolismo
6.
Brain Dev ; 45(5): 251-259, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36870919

RESUMO

Chaperone therapy was introduced first as a new molecular therapeutic approach to lysosomal diseases. In a recent article, I reviewed the development of chaperone therapy mainly for lysosomal diseases. Then, more data have been collected particularly on non-lysosomal protein misfolding diseases. In this short review, I propose the concept of chaperone therapy to be classified into two different therapeutic approaches, for pH-dependent lysosomal, and pH-independent non-lysosomal protein misfolding diseases. The concept of lysosomal chaperone therapy is well established, but the non-lysosomal chaperone therapy is heterogeneous and to be investigated further for various individual diseases. As a whole, these two-types of new molecular therapeutic approaches will make an impact on the treatment of a wide range of pathological conditions caused by protein misfolding, not necessarily lysosomal but also many non-lysosomal diseases caused by gene mutations, metabolic diseases, malignancy, infectious diseases, and aging. The concept will open a completely new aspect of protein therapy in future.


Assuntos
Chaperonas Moleculares , Deficiências na Proteostase , Humanos , Chaperonas Moleculares/metabolismo , Deficiências na Proteostase/genética , Deficiências na Proteostase/terapia , Deficiências na Proteostase/metabolismo , Mutação , Lisossomos/metabolismo
7.
Nat Rev Neurol ; 19(4): 235-245, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36828943

RESUMO

The protein homeostasis (proteostasis) system encompasses the cellular processes that regulate protein synthesis, folding, concentration, trafficking and degradation. In the case of intracellular proteostasis, the identity and nature of these processes have been extensively studied and are relatively well known. By contrast, the mechanisms of extracellular proteostasis are yet to be fully elucidated, although evidence is accumulating that their age-related progressive impairment might contribute to neuronal death in neurodegenerative diseases. Constitutively secreted extracellular chaperones are emerging as key players in processes that operate to protect neurons and other brain cells by neutralizing the toxicity of extracellular protein aggregates and promoting their safe clearance and disposal. Growing evidence indicates that these extracellular chaperones exert multiple effects to promote cell viability and protect neurons against pathologies arising from the misfolding and aggregation of proteins in the synaptic space and interstitial fluid. In this Review, we outline the current knowledge of the mechanisms of extracellular proteostasis linked to neurodegenerative diseases, and we examine the latest understanding of key molecules and processes that protect the brain from the pathological consequences of extracellular protein aggregation and proteotoxicity. Finally, we contemplate possible therapeutic opportunities for neurodegenerative diseases on the basis of this emerging knowledge.


Assuntos
Doenças Neurodegenerativas , Deficiências na Proteostase , Humanos , Proteostase , Dobramento de Proteína , Doenças Neurodegenerativas/metabolismo , Chaperonas Moleculares/metabolismo , Chaperonas Moleculares/uso terapêutico , Homeostase , Deficiências na Proteostase/metabolismo , Deficiências na Proteostase/patologia
8.
Can J Neurol Sci ; 50(3): 329-345, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-35356856

RESUMO

Neurodegenerative diseases are a pathologically, clinically and genetically diverse group of disorders without effective disease-modifying therapies. Pathologically, these disorders are characterised by disease-specific protein aggregates in neurons and/or glia and referred to as proteinopathies. Many neurodegenerative diseases show pathological overlap with the same abnormally deposited protein occurring in anatomically distinct regions, which give rise to specific patterns of cognitive and motor clinical phenotypes. Sequential distribution patterns of protein inclusions throughout the brain have been described. Rather than occurring in isolation, it is increasingly recognised that combinations of one or more proteinopathies with or without cerebrovascular disease frequently occur in individuals with neurodegenerative diseases. In addition, complex constellations of ageing-related and incidental pathologies associated with tau, TDP-43, Aß, α-synuclein deposition have been commonly reported in longitudinal ageing studies. This review provides an overview of current classification of neurodegenerative and age-related pathologies and presents the spectrum and complexity of mixed pathologies in community-based, longitudinal ageing studies, in major proteinopathies, and genetic conditions. Mixed pathologies are commonly reported in individuals >65 years with and without cognitive impairment; however, they are increasingly recognised in younger individuals (<65 years). Mixed pathologies are thought to lower the threshold for developing cognitive impairment and dementia. Hereditary neurodegenerative diseases also show a diverse range of mixed pathologies beyond the proteinopathy primarily linked to the genetic abnormality. Cases with mixed pathologies might show a different clinical course, which has prognostic relevance and obvious implications for biomarker and therapy development, and stratifying patients for clinical trials.


Assuntos
Transtornos Cerebrovasculares , Doenças Neurodegenerativas , Deficiências na Proteostase , Humanos , Doenças Neurodegenerativas/patologia , Encéfalo/patologia , Estudos Longitudinais , Deficiências na Proteostase/metabolismo , Deficiências na Proteostase/patologia , Proteínas tau
9.
J Biol Chem ; 299(2): 102806, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36529289

RESUMO

Karyopherin-ß2 (Kapß2) is a nuclear-import receptor that recognizes proline-tyrosine nuclear localization signals of diverse cytoplasmic cargo for transport to the nucleus. Kapß2 cargo includes several disease-linked RNA-binding proteins with prion-like domains, such as FUS, TAF15, EWSR1, hnRNPA1, and hnRNPA2. These RNA-binding proteins with prion-like domains are linked via pathology and genetics to debilitating degenerative disorders, including amyotrophic lateral sclerosis, frontotemporal dementia, and multisystem proteinopathy. Remarkably, Kapß2 prevents and reverses aberrant phase transitions of these cargoes, which is cytoprotective. However, the molecular determinants of Kapß2 that enable these activities remain poorly understood, particularly from the standpoint of nuclear-import receptor architecture. Kapß2 is a super-helical protein comprised of 20 HEAT repeats. Here, we design truncated variants of Kapß2 and assess their ability to antagonize FUS aggregation and toxicity in yeast and FUS condensation at the pure protein level and in human cells. We find that HEAT repeats 8 to 20 of Kapß2 recapitulate all salient features of Kapß2 activity. By contrast, Kapß2 truncations lacking even a single cargo-binding HEAT repeat display reduced activity. Thus, we define a minimal Kapß2 construct for delivery in adeno-associated viruses as a potential therapeutic for amyotrophic lateral sclerosis/frontotemporal dementia, multisystem proteinopathy, and related disorders.


Assuntos
Chaperonas Moleculares , Fragmentos de Peptídeos , Príons , Proteína FUS de Ligação a RNA , beta Carioferinas , Humanos , Esclerose Amiotrófica Lateral/metabolismo , Esclerose Amiotrófica Lateral/terapia , beta Carioferinas/química , beta Carioferinas/genética , beta Carioferinas/metabolismo , Linhagem Celular , Dependovirus/metabolismo , Demência Frontotemporal/metabolismo , Demência Frontotemporal/terapia , Técnicas In Vitro , Chaperonas Moleculares/química , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Príons/química , Príons/metabolismo , Deficiências na Proteostase/metabolismo , Deficiências na Proteostase/terapia , Proteína FUS de Ligação a RNA/química , Proteína FUS de Ligação a RNA/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Ligação Proteica
10.
Anal Chim Acta ; 1238: 340163, 2023 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-36464456

RESUMO

Misfolding of superoxide dismutase-1 (SOD1) has been correlated with many neurodegenerative diseases, such as Amyotrophic lateral sclerosis's and Alzheimer's among others. However, it is unclear whether misfolded SOD1 plays a role in another neurodegenerative disease of white matter lesions (WMLs). In this study, a sensitive and specific method based on SERS technique was proposed for quantitative detection of misfolded SOD1 content in WMLs. To fabricate the double antibodysandwich substrates for SERS detection, gold nanostars modified with capture antibody were immobilized on glass substrates to prepare active SERS substrates, and then SERS probes conjugated with a Raman reporter and a specific target antibody were coupled with active SERS substrates. This SERS substrates had been employed for quantitative detection of misfolded SOD1 levels in WMLs and exhibited excellent stability, reliability, and accuracy. Moreover, experimental results indicated that the level of misfolded SOD1 increased with the increase in age and the degree of WMLs. Hence, misfolded SOD1 may be a potential blood marker for WMLs and aging. Meanwhile, SERS-based gold nanostars have great clinical application potential in the screening, diagnosis and treatment of WMLs.


Assuntos
Doenças Neurodegenerativas , Deficiências na Proteostase , Superóxido Dismutase-1 , Substância Branca , Humanos , Anticorpos , Ouro , Doenças Neurodegenerativas/diagnóstico , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/metabolismo , Reprodutibilidade dos Testes , Superóxido Dismutase , Superóxido Dismutase-1/análise , Superóxido Dismutase-1/genética , Superóxido Dismutase-1/metabolismo , Substância Branca/metabolismo , Substância Branca/fisiopatologia , Deficiências na Proteostase/diagnóstico , Deficiências na Proteostase/genética , Deficiências na Proteostase/metabolismo
11.
Cells ; 11(21)2022 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-36359796

RESUMO

Copper is a transition metal essential for human life. Its homeostasis is regulated in the liver, which delivers copper to the whole body and excretes its excess outside the organism in the feces through the bile. These functions are regulated within hepatocytes, and the ATP7B copper transporter is central to making the switch between copper use and excretion. In Wilson disease, the gene coding for ATP7B is mutated, leading to copper overload, firstly, in the liver and the brain. To better understand the role of ATP7B in hepatocytes and to provide a smart tool for the development of novel therapies against Wilson disease, we used the CrispR/Cas9 tool to generate hepatocyte cell lines with the abolished expression of ATP7B. These cell lines revealed that ATP7B plays a major role at low copper concentrations starting in the micromolar range. Moreover, metal stress markers are induced at lower copper concentrations compared to parental cells, while redox stress remains not activated. As shown recently, the main drawback induced by copper exposure is protein unfolding that is drastically exacerbated in ATP7B-deficient cells. Our data enabled us to propose that the zinc finger domain of DNAJ-A1 would serve as a sensor of Cu stress. Therefore, these Wilson-like hepatocytes are of high interest to explore in more detail the role of ATP7B.


Assuntos
ATPases Transportadoras de Cobre , Cobre , Degeneração Hepatolenticular , Humanos , Linhagem Celular , Cobre/farmacologia , Cobre/metabolismo , ATPases Transportadoras de Cobre/genética , ATPases Transportadoras de Cobre/metabolismo , Hepatócitos/metabolismo , Degeneração Hepatolenticular/genética , Deficiências na Proteostase/genética , Deficiências na Proteostase/metabolismo
12.
Int J Mol Sci ; 23(19)2022 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-36233058

RESUMO

Motoneuron diseases (MNDs) are neurodegenerative conditions associated with death of upper and/or lower motoneurons (MNs). Proteostasis alteration is a pathogenic mechanism involved in many MNDs and is due to the excessive presence of misfolded and aggregated proteins. Protein misfolding may be the product of gene mutations, or due to defects in the translation process, or to stress agents; all these conditions may alter the native conformation of proteins making them prone to aggregate. Alternatively, mutations in members of the protein quality control (PQC) system may determine a loss of function of the proteostasis network. This causes an impairment in the capability to handle and remove aberrant or damaged proteins. The PQC system consists of the degradative pathways, which are the autophagy and the proteasome, and a network of chaperones and co-chaperones. Among these components, Heat Shock Protein 70 represents the main factor in substrate triage to folding, refolding, or degradation, and it is assisted in this task by a subclass of the chaperone network, the small heat shock protein (sHSPs/HSPBs) family. HSPBs take part in proteostasis by bridging misfolded and aggregated proteins to the HSP70 machinery and to the degradative pathways, facilitating refolding or clearance of the potentially toxic proteins. Because of its activity against proteostasis alteration, the chaperone system plays a relevant role in the protection against proteotoxicity in MNDs. Here, we discuss the role of HSPBs in MNDs and which HSPBs may represent a valid target for therapeutic purposes.


Assuntos
Proteínas de Choque Térmico Pequenas , Doença dos Neurônios Motores , Deficiências na Proteostase , Proteínas de Choque Térmico HSP70/genética , Proteínas de Choque Térmico HSP70/metabolismo , Proteínas de Choque Térmico Pequenas/genética , Proteínas de Choque Térmico Pequenas/metabolismo , Humanos , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Doença dos Neurônios Motores/metabolismo , Neurônios Motores/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Dobramento de Proteína , Deficiências na Proteostase/metabolismo
13.
J Biol Chem ; 298(9): 102285, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35870554

RESUMO

Secretagogin (SCGN) is a three-domain hexa-EF-hand Ca2+-binding protein that plays a regulatory role in the release of several hormones. SCGN is expressed largely in pancreatic ß-cells, certain parts of the brain, and also in neuroendocrine tissues. The expression of SCGN is altered in several diseases, such as diabetes, cancers, and neurodegenerative disorders; however, the precise associations that closely link SCGN expression to such pathophysiologies are not known. In this work, we report that SCGN is an early responder to cellular stress, and SCGN expression is temporally upregulated by oxidative stress and heat shock. We show the overexpression of SCGN efficiently prevents cells from heat shock and oxidative damage. We further demonstrate that in the presence of Ca2+, SCGN efficiently prevents the aggregation of a broad range of model proteins in vitro. Small-angle X-ray scattering (BioSAXS) studies further reveal that Ca2+ induces the conversion of a closed compact apo-SCGN conformation into an open extended holo-SCGN conformation via multistate intermediates, consistent with the augmentation of chaperone activity of SCGN. Furthermore, isothermal titration calorimetry establishes that Ca2+ enables SCGN to bind α-synuclein and insulin, two target proteins of SCGN. Altogether, our data not only demonstrate that SCGN is a Ca2+-dependent generic molecular chaperone involved in protein homeostasis with broad substrate specificity but also elucidate the origin of its altered expression in several cancers. We describe a plausible mechanism of how perturbations in Ca2+ homeostasis and/or deregulated SCGN expression would hasten the process of protein misfolding, which is a feature of many aggregation-based proteinopathies.


Assuntos
Cálcio , Motivos EF Hand , Resposta ao Choque Térmico , Células Secretoras de Insulina , Chaperonas Moleculares , Estresse Oxidativo , Agregação Patológica de Proteínas , Deficiências na Proteostase , Secretagoginas , Animais , Cálcio/metabolismo , Células HEK293 , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Chaperonas Moleculares/química , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Agregação Patológica de Proteínas/metabolismo , Dobramento de Proteína , Deficiências na Proteostase/genética , Deficiências na Proteostase/metabolismo , Ratos , Secretagoginas/química , Secretagoginas/genética , Secretagoginas/metabolismo , alfa-Sinucleína/metabolismo
14.
Int J Mol Sci ; 23(9)2022 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-35563197

RESUMO

Neurodegenerative protein conformational diseases are characterized by the misfolding and aggregation of metastable proteins encoded within the host genome. The host is also home to thousands of proteins encoded within exogenous genomes harbored by bacteria, fungi, and viruses. Yet, their contributions to host protein-folding homeostasis, or proteostasis, remain elusive. Recent studies, including our previous work, suggest that bacterial products contribute to the toxic aggregation of endogenous host proteins. We refer to these products as bacteria-derived protein aggregates (BDPAs). Furthermore, antibiotics were recently associated with an increased risk for neurodegenerative diseases, including Parkinson's disease and amyotrophic lateral sclerosis-possibly by virtue of altering the composition of the human gut microbiota. Other studies have shown a negative correlation between disease progression and antibiotic administration, supporting their protective effect against neurodegenerative diseases. These contradicting studies emphasize the complexity of the human gut microbiota, the gut-brain axis, and the effect of antibiotics. Here, we further our understanding of bacteria's effect on host protein folding using the model Caenorhabditis elegans. We employed genetic and chemical methods to demonstrate that the proteotoxic effect of bacteria on host protein folding correlates with the presence of BDPAs. Furthermore, the abundance and proteotoxicity of BDPAs are influenced by gentamicin, an aminoglycoside antibiotic that induces protein misfolding, and by butyrate, a short-chain fatty acid that we previously found to affect host protein aggregation and the associated toxicity. Collectively, these results increase our understanding of host-bacteria interactions in the context of protein conformational diseases.


Assuntos
Doenças Neurodegenerativas , Deficiências na Proteostase , Animais , Antibacterianos , Bactérias/metabolismo , Caenorhabditis elegans/metabolismo , Humanos , Doenças Neurodegenerativas/metabolismo , Agregados Proteicos , Dobramento de Proteína , Proteínas/metabolismo , Proteostase , Deficiências na Proteostase/metabolismo
15.
Nucl Med Biol ; 114-115: 108-120, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35487833

RESUMO

This review outlines small molecule radiotracers developed for positron emission tomography (PET) imaging of proteinopathies, neurodegenerative diseases characterised by accumulation of malformed proteins, over the last two decades with the focus on radioligands that have progressed to clinical studies. Introduction provides a short summary of proteinopathy targets used for PET imaging, including vastly studied proteins Aß and tau and emerging α-synuclein. In the main section, clinically relevant Aß and tau radioligand classes and their properties are discussed, including an overview of lead compounds and radioligand candidates studied as α-synuclein imaging agents in the early discovery and preclinical development phase. Lastly, the specific challenges and future directions in proteinopathy radioligand development are summarized.


Assuntos
Doença de Alzheimer , Deficiências na Proteostase , Humanos , alfa-Sinucleína/metabolismo , alfa-Sinucleína/farmacologia , Doença de Alzheimer/metabolismo , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Tomografia por Emissão de Pósitrons/métodos , Deficiências na Proteostase/diagnóstico por imagem , Deficiências na Proteostase/metabolismo , Proteínas tau/metabolismo
16.
J Biol Chem ; 298(5): 101930, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35421375

RESUMO

Immune checkpoint blockade therapy is perhaps the most important development in cancer treatment in recent memory. It is based on decades of investigation into the biology of immune cells and the role of the immune system in controlling cancer growth. While the molecular circuitry that governs the immune system in general-and antitumor immunity in particular-is intensely studied, far less attention has been paid to the role of cellular stress in this process. Proteostasis, intimately linked to cell stress responses, refers to the dynamic regulation of the cellular proteome and is maintained through a complex network of systems that govern the synthesis, folding, and degradation of proteins in the cell. Disruption of these systems can result in the loss of protein function, altered protein function, the formation of toxic aggregates, or pathologies associated with cell stress. However, the importance of proteostasis extends beyond its role in maintaining proper protein function; proteostasis governs how tolerant cells may be to mutations in protein-coding genes and the overall half-life of proteins. Such gene expression changes may be associated with human diseases including neurodegenerative diseases, metabolic disease, and cancer and manifest at the protein level against the backdrop of the proteostasis network in any given cellular environment. In this review, we focus on the role of proteostasis in regulating immune responses against cancer as well the role of proteostasis in determining immunogenicity of cancer cells.


Assuntos
Neoplasias , Proteostase , Humanos , Neoplasias/imunologia , Dobramento de Proteína , Proteoma/metabolismo , Proteostase/imunologia , Deficiências na Proteostase/metabolismo
17.
Molecules ; 27(3)2022 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-35164285

RESUMO

Amyloidosis is a common pathological event in which proteins self-assemble into misfolded soluble and insoluble molecular forms, oligomers and fibrils that are often toxic to cells. Notably, aggregation-prone human islet amyloid polypeptide (hIAPP), or amylin, is a pancreatic hormone linked to islet ß-cells demise in diabetics. The unifying mechanism by which amyloid proteins, including hIAPP, aggregate and kill cells is still matter of debate. The pathology of type-2 diabetes mellitus (T2DM) is characterized by extracellular and intracellular accumulation of toxic hIAPP species, soluble oligomers and insoluble fibrils in pancreatic human islets, eventually leading to loss of ß-cell mass. This review focuses on molecular, biochemical and cell-biology studies exploring molecular mechanisms of hIAPP synthesis, trafficking and degradation in the pancreas. In addition to hIAPP turnover, the dynamics and the mechanisms of IAPP-membrane interactions; hIAPP aggregation and toxicity in vitro and in situ; and the regulatory role of diabetic factors, such as lipids and cholesterol, in these processes are also discussed.


Assuntos
Diabetes Mellitus Tipo 2/patologia , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Pâncreas/patologia , Agregação Patológica de Proteínas/patologia , Animais , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Polipeptídeo Amiloide das Ilhotas Pancreáticas/análise , Pâncreas/metabolismo , Agregados Proteicos , Agregação Patológica de Proteínas/metabolismo , Conformação Proteica , Dobramento de Proteína , Deficiências na Proteostase/metabolismo , Deficiências na Proteostase/patologia
18.
Mol Cell Neurosci ; 118: 103682, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34800621

RESUMO

Proteostasis dysfunction and activation of the unfolded protein response (UPR) are characteristic of all major neurodegenerative diseases. Nevertheless, although the UPR and proteostasis dysfunction has been studied in great detail in model organisms like yeast and mammalian cell lines, it has not yet been examined in neurons. In this study, we applied a viral vector-mediated expression of a reporter protein based on a UPR transcription factor, ATF4, and time-lapse fluorescent microscopy to elucidate how mouse primary neurons respond to pharmacological and genetic perturbations to neuronal proteostasis. In in vitro models of endoplasmic reticulum (ER) stress and proteasome inhibition, we used the ATF4 reporter to reveal the time course of the neuronal stress response relative to neurite degeneration and asynchronous cell death. We showed how potential neurodegenerative disease co-factors, ER stress and mutant α-synuclein overexpression, impacted neuronal stress response and overall cellular health. This work therefore introduces a viral vector-based reporter that yields a quantifiable readout suitable for non-cell destructive kinetic monitoring of proteostasis dysfunction in neurons by harnessing ATF4 signaling as part of the UPR activation.


Assuntos
Doenças Neurodegenerativas , Deficiências na Proteostase , Animais , Estresse do Retículo Endoplasmático/fisiologia , Mamíferos , Camundongos , Doenças Neurodegenerativas/metabolismo , Neurônios/metabolismo , Deficiências na Proteostase/metabolismo , Resposta a Proteínas não Dobradas
19.
Trends Cell Biol ; 32(6): 470-474, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-34863585

RESUMO

Chaperone expression is developmentally regulated, establishing tissue-specific networks. However, the molecular basis underlying this specificity is mainly unknown. Recent evidence suggests that chaperone network rewiring is mediated, in part, by differentiation transcription factors to fit the proteome folding demands, with implications for the tissue-specific manifestation of protein misfolding diseases.


Assuntos
Chaperonas Moleculares , Deficiências na Proteostase , Diferenciação Celular , Humanos , Chaperonas Moleculares/metabolismo , Dobramento de Proteína , Proteoma/metabolismo , Deficiências na Proteostase/metabolismo , Fatores de Transcrição
20.
Trends Cell Biol ; 32(3): 202-215, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34654604

RESUMO

The proteostasis network (PN) regulates protein synthesis, folding, and degradation and is critical for the health and function of all cells. The PN has been extensively studied in the context of aging and age-related diseases, and loss of proteostasis is regarded as a major contributor to many age-associated disorders. In contrast to somatic tissues, an important feature of germ cells is their ability to maintain a healthy proteome across generations. Accumulating evidence has now revealed multiple layers of PN regulation that support germ cell function, determine reproductive capacity during aging, and prioritize reproduction at the expense of somatic health. Here, we review recent insights into these different modes of regulation and their implications for reproductive and somatic aging.


Assuntos
Deficiências na Proteostase , Proteostase , Envelhecimento/fisiologia , Humanos , Dobramento de Proteína , Proteoma/metabolismo , Deficiências na Proteostase/metabolismo , Reprodução
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...