Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Int J Mol Sci ; 24(7)2023 Mar 28.
Article in English | MEDLINE | ID: mdl-37047286

ABSTRACT

The self-association of amylogenic proteins to the fibril form is considered a pivotal factor in the pathogenesis of neurodegenerative diseases, including Parkinson's disease (PD). PD causes unintended or uncontrollable movements in its common symptoms. α-synuclein is the major cause of PD development and thus has been the main target of numerous studies to suppress and sequester its expression or effectively degrade it. Nonetheless, to date, there are no efficient and proven ways to prevent pathological protein aggregation. Recent investigations proposed applying an external electric field to interrupt the fibrils. This method is a non-invasive approach that has a certain benefit over others. We performed molecular dynamics (MD) simulations by applying an electric field on highly toxic fibrils of α-synuclein to gain a molecular-level insight into fibril disruption mechanisms. The results revealed that the applied external electric field induces substantial changes in the conformation of the α-synuclein fibrils. Furthermore, we show the threshold value for electric field strength required to completely disrupt the α-synuclein fibrils by opening the hydrophobic core of the fibril. Thus, our findings might serve as a valuable foundation to better understand molecular-level mechanisms of the α-synuclein fibrils disaggregation process under an applied external electric field.


Subject(s)
Parkinson Disease , alpha-Synuclein , Humans , alpha-Synuclein/metabolism , Molecular Dynamics Simulation , Parkinson Disease/metabolism , Protein Aggregation, Pathological/complications , Amyloid/metabolism
2.
Neurobiol Aging ; 109: 52-63, 2022 01.
Article in English | MEDLINE | ID: mdl-34655981

ABSTRACT

Pathological aggregation of tau and neuroinflammatory changes mark the clinical course of Alzheimer's disease and related tauopathies. To understand the correlation between these pathological hallmarks and functional deficits, we assessed behavioral and physiological deficits in the PS19 mouse model, a broadly utilized model of tauopathy. At 9 months, PS19 mice have characteristic hyperactive behavior, a decline in motor strength, and deterioration in physiological conditions marked by lower body temperature, reduced body weight, and an increase in measures of frailty. Correlation of these deficits with different pathological hallmarks revealed that pathological tau species, characterized by soluble p-tau species, and tau seeding bioactivity correlated with impairment in grip strength and thermal regulation. On the other hand, astrocyte reactivity showed a positive correlation with the hyperactive behavior of the PS19 mice. These results suggest that a diverse spectrum of soluble pathological tau species could be responsible for different symptoms and that neuroinflammation could contribute to functional deficits independently from tau pathology. These observations enhance the necessity of a multi-targeted approach for the treatment of neurodegenerative tauopathies.


Subject(s)
Gliosis/etiology , Neuroinflammatory Diseases/complications , Protein Aggregation, Pathological/complications , Tauopathies/etiology , tau Proteins/metabolism , Animals , Behavior, Animal , Body Temperature Regulation , Disease Models, Animal , Female , Frailty/etiology , Hand Strength , Humans , Male , Mice, Transgenic , Motor Activity , Tauopathies/pathology , Tauopathies/physiopathology , Tauopathies/psychology
3.
Int J Mol Sci ; 22(12)2021 Jun 21.
Article in English | MEDLINE | ID: mdl-34205606

ABSTRACT

Neurodegenerative disorders (NDDs), including Alzheimer's, Parkinson's, and Huntington's diseases, are a highly prevalent class of disorders that share the presence of aberrant aggregates called amyloids in the nervous system [...].


Subject(s)
Amyloid , Amyloidosis , Neurodegenerative Diseases/etiology , Protein Aggregates , Protein Aggregation, Pathological/complications , Animals , Humans
4.
Cell Death Dis ; 12(6): 592, 2021 06 08.
Article in English | MEDLINE | ID: mdl-34103467

ABSTRACT

Stress granules (SGs) are membraneless cell compartments formed in response to different stress stimuli, wherein translation factors, mRNAs, RNA-binding proteins (RBPs) and other proteins coalesce together. SGs assembly is crucial for cell survival, since SGs are implicated in the regulation of translation, mRNA storage and stabilization and cell signalling, during stress. One defining feature of SGs is their dynamism, as they are quickly assembled upon stress and then rapidly dispersed after the stress source is no longer present. Recently, SGs dynamics, their components and their functions have begun to be studied in the context of human diseases. Interestingly, the regulated protein self-assembly that mediates SG formation contrasts with the pathological protein aggregation that is a feature of several neurodegenerative diseases. In particular, aberrant protein coalescence is a key feature of polyglutamine (PolyQ) diseases, a group of nine disorders that are caused by an abnormal expansion of PolyQ tract-bearing proteins, which increases the propensity of those proteins to aggregate. Available data concerning the abnormal properties of the mutant PolyQ disease-causing proteins and their involvement in stress response dysregulation strongly suggests an important role for SGs in the pathogenesis of PolyQ disorders. This review aims at discussing the evidence supporting the existence of a link between SGs functionality and PolyQ disorders, by focusing on the biology of SGs and on the way it can be altered in a PolyQ disease context.


Subject(s)
Cytoplasmic Granules/physiology , Neurodegenerative Diseases/etiology , Peptides/metabolism , RNA-Binding Proteins/physiology , Stress, Physiological/physiology , Animals , Cytoplasmic Granules/metabolism , Humans , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Protein Aggregation, Pathological/complications , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology , RNA-Binding Proteins/metabolism , Signal Transduction/physiology
5.
J Alzheimers Dis ; 79(3): 979-1002, 2021.
Article in English | MEDLINE | ID: mdl-33386802

ABSTRACT

Amyloid-ß (Aß) and tau oligomers have been identified as neurotoxic agents responsible for causing Alzheimer's disease (AD). Clinical trials using Aß and tau as targets have failed, giving rise to calls for new research approaches to combat AD. This paper provides such an approach. Most basic AD research has involved quiescent Aß and tau solutions. However, studies involving laminar and extensional flow of proteins have demonstrated that mechanical agitation of proteins induces or accelerates protein aggregation. Recent MRI brain studies have revealed high energy, chaotic motion of cerebrospinal fluid (CSF) in lower brain and brainstem regions. These and studies showing CSF flow within the brain have shown that there are two energetic hot spots. These are within the third and fourth brain ventricles and in the neighborhood of the circle of Willis blood vessel region. These two regions are also the same locations as those of the earliest Aß and tau AD pathology. In this paper, it is proposed that cardiac systolic pulse waves that emanate from the major brain arteries in the lower brain and brainstem regions and whose pulse waves drive CSF flows within the brain are responsible for initiating AD and possibly other amyloid diseases. It is further proposed that the triggering of these diseases comes about because of the strengthening of systolic pulses due to major artery hardening that generates intense CSF extensional flow stress. Such stress provides the activation energy needed to induce conformational changes of both Aß and tau within the lower brain and brainstem region, producing unique neurotoxic oligomer molecule conformations that induce AD.


Subject(s)
Alzheimer Disease/etiology , Amyloid beta-Peptides/metabolism , Brain Stem/pathology , Cerebrospinal Fluid/physiology , Plaque, Amyloid/etiology , tau Proteins/metabolism , Alzheimer Disease/pathology , Brain/metabolism , Brain/pathology , Disease Progression , Humans , Models, Biological , Plaque, Amyloid/pathology , Protein Aggregation, Pathological/complications , Protein Aggregation, Pathological/pathology , Stress, Physiological
6.
Int J Mol Sci ; 21(17)2020 Sep 03.
Article in English | MEDLINE | ID: mdl-32899160

ABSTRACT

Neurodegenerative proteinopathies are complex diseases that share some pathogenetic processes. One of these is the failure of the proteostasis network (PN), which includes all components involved in the synthesis, folding, and degradation of proteins, thus leading to the aberrant accumulation of toxic protein aggregates in neurons. The single components that belong to the three main modules of the PN are highly interconnected and can be considered as part of a single giant network. Several pharmacological strategies have been proposed to ameliorate neurodegeneration by targeting PN components. Nevertheless, effective disease-modifying therapies are still lacking. In this review article, after a general description of the PN and its failure in proteinopathies, we will focus on the available pharmacological tools to target proteostasis. In this context, we will discuss the main advantages of systems-based pharmacology in contrast to the classical targeted approach, by focusing on network pharmacology as a strategy to innovate rational drug design.


Subject(s)
Neurodegenerative Diseases/pathology , Protein Aggregation, Pathological/complications , Proteins/metabolism , Proteostasis Deficiencies/pathology , Proteostasis , Animals , Homeostasis , Humans , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/metabolism , Proteostasis Deficiencies/etiology
7.
Rev Mal Respir ; 37(2): 180-186, 2020 Feb.
Article in French | MEDLINE | ID: mdl-32014310

ABSTRACT

INTRODUCTION: Light chain deposition disease is a rare anatomo-clinical disorder, which rarely leads to cystic lung destruction. CASE REPORT: We report the case of a 62years old female patient with a history of a monoclonal gammopathy of unknown significance who developed progressive dyspnea. Thoracic CT-scan demonstrated a diffuse pulmonary cystic disorder with predominance in the right lower lobe. Thoracoscopic surgical resection of that lobe led to a diagnosis of non-amyloid kappa light chain deposits. Surgery also resulted in a lung volume reduction effect with clinical and functional benefits related to improved ventilation of adjacent segments. CONCLUSION: This report of pulmonary cystic disorder related to a light chain deposition disease highlights the potential clinical and functional benefits observed after lung volume reduction surgery.


Subject(s)
Cysts/surgery , Immunoglobulin Light Chains/metabolism , Lung Diseases/surgery , Monoclonal Gammopathy of Undetermined Significance/surgery , Protein Aggregation, Pathological/pathology , Cysts/metabolism , Cysts/pathology , Diagnosis, Differential , Female , Humans , Immunoglobulin Light-chain Amyloidosis/diagnosis , Lung Diseases/metabolism , Lung Diseases/pathology , Middle Aged , Monoclonal Gammopathy of Undetermined Significance/complications , Monoclonal Gammopathy of Undetermined Significance/diagnosis , Monoclonal Gammopathy of Undetermined Significance/pathology , Pneumonectomy , Protein Aggregation, Pathological/complications , Protein Aggregation, Pathological/diagnosis , Protein Aggregation, Pathological/metabolism , Treatment Outcome
8.
Annu Rev Pathol ; 15: 261-285, 2020 01 24.
Article in English | MEDLINE | ID: mdl-31594457

ABSTRACT

Autophagy is an evolutionarily conserved catabolic process that targets different types of cytoplasmic cargo (such as bulk cytoplasm, damaged cellular organelles, and misfolded protein aggregates) for lysosomal degradation. Autophagy is activated in response to biological stress and also plays a critical role in the maintenance of normal cellular homeostasis; the latter function is particularly important for the integrity of postmitotic, metabolically active tissues, such as skeletal muscle. Through impairment of muscle homeostasis, autophagy dysfunction contributes to the pathogenesis of many different skeletal myopathies; the observed autophagy defects differ from disease to disease but have been shown to involve all steps of the autophagic cascade (from induction to lysosomal cargo degradation) and to impair both bulk and selective autophagy. To highlight the molecular and cellular mechanisms that are shared among different myopathies with deficient autophagy, these disorders are discussed based on the nature of the underlying autophagic defect rather than etiology or clinical presentation.


Subject(s)
Autophagy/physiology , Muscular Diseases/etiology , Animals , Autophagy/genetics , Humans , Lysosomes/physiology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Diseases/genetics , Protein Aggregation, Pathological/complications , Protein Aggregation, Pathological/genetics
9.
Cell Mol Life Sci ; 77(9): 1721-1744, 2020 May.
Article in English | MEDLINE | ID: mdl-31667556

ABSTRACT

Accumulation of misfolded and aggregated forms of tau protein in the brain is a neuropathological hallmark of tauopathies, such as Alzheimer's disease and frontotemporal lobar degeneration. Tau aggregates have the ability to transfer from one cell to another and to induce templated misfolding and aggregation of healthy tau molecules in previously healthy cells, thereby propagating tau pathology across different brain areas in a prion-like manner. The molecular mechanisms involved in cell-to-cell transfer of tau aggregates are diverse, not mutually exclusive and only partially understood. Intracellular accumulation of misfolded tau induces several mechanisms that aim to reduce the cellular burden of aggregated proteins and also promote secretion of tau aggregates. However, tau may also be released from cells physiologically unrelated to protein aggregation. Tau secretion involves multiple vesicular and non-vesicle-mediated pathways, including secretion directly through the plasma membrane. Consequently, extracellular tau can be found in various forms, both as a free protein and in vesicles, such as exosomes and ectosomes. Once in the extracellular space, tau aggregates can be internalized by neighboring cells, both neurons and glial cells, via endocytic, pinocytic and phagocytic mechanisms. Importantly, accumulating evidence suggests that prion-like propagation of misfolding protein pathology could provide a general mechanism for disease progression in tauopathies and other related neurodegenerative diseases. Here, we review the recent literature on cellular mechanisms involved in cell-to-cell transfer of tau, with a particular focus in tau secretion.


Subject(s)
Protein Aggregation, Pathological/complications , Tauopathies/pathology , tau Proteins/metabolism , Animals , Disease Progression , Humans , Tauopathies/etiology , Tauopathies/metabolism
11.
Nat Neurosci ; 21(10): 1332-1340, 2018 10.
Article in English | MEDLINE | ID: mdl-30250260

ABSTRACT

A hallmark event in neurodegenerative diseases (NDs) is the misfolding, aggregation, and accumulation of proteins, leading to cellular dysfunction, loss of synaptic connections, and brain damage. Despite the involvement of distinct proteins in different NDs, the process of protein misfolding and aggregation is remarkably similar. A recent breakthrough in the field was the discovery that misfolded protein aggregates can self-propagate through seeding and spread the pathological abnormalities between cells and tissues in a manner akin to the behavior of infectious prions in prion diseases. This discovery has vast implications for understanding the mechanisms involved in the initiation and progression of NDs, as well as for the design of novel strategies for treatment and diagnosis. In this Review, we provide a critical discussion of the role of protein misfolding and aggregation in NDs. Commonalities and differences between distinct protein aggregates will be highlighted, in addition to evidence supporting the hypothesis that misfolded aggregates can be transmissible by the prion principle. We will also describe the molecular basis and implications for prion-like conformational strains, cross-interaction between different misfolded proteins in the brain, and how these concepts can be applied to the development of novel strategies for therapy and diagnosis.


Subject(s)
Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/metabolism , Protein Aggregation, Pathological/complications , Protein Folding , Animals , Humans , Protein Conformation
12.
Prog Neurobiol ; 169: 24-54, 2018 10.
Article in English | MEDLINE | ID: mdl-30077775

ABSTRACT

Oligodendrocytes are in contact with neurons, wrap axons with a myelin sheath that protects their structural integrity, and facilitate nerve conduction. Oligodendrocytes also form a syncytium with astrocytes which interacts with neurons, promoting reciprocal survival mediated by activity and by molecules involved in energy metabolism and trophism. Therefore, oligodendrocytes are key elements in the normal functioning of the central nervous system. Oligodendrocytes are affected following different insults to the central nervous system including ischemia, traumatism, and inflammation. The term oligodendrogliopathy highlights the prominent role of altered oligodendrocytes in the pathogenesis of certain neurological diseases, not only in demyelinating diseases and most leukodystrophies, but also in aging and age-related neurodegenerative diseases with abnormal protein aggregates. Most of these diseases are characterized by the presence of abnormal protein deposits, forming characteristic and specific inclusions in neurons and astrocytes but also in oligodendrocytes, thus signaling their involvement in the disease. Emerging evidence suggests that such deposits in oligodendrocytes are not mere bystanders but rather are associated with functional alterations. Moreover, operative modifications in oligodendrocytes are also detected in the absence of oligodendroglial inclusions in certain diseases. The present review focuses first on general aspects of oligodendrocytes and precursors, and their development and functions, and then introduces and updates alterations and dysfunction of oligodendrocytes in selected neurodegenerative diseases with abnormal protein aggregates such as multiple system atrophy, Lewy body diseases, tauopathies, Alzheimer's disease, amyotrophic lateral sclerosis, frontotemporal lobar degeneration with TDP-43 inclusions (TDP-43 proteinopathies), and Creutzfeldt-Jakob´s disease as a prototypical human prionopathy.


Subject(s)
Neurodegenerative Diseases/complications , Oligodendroglia/pathology , Protein Aggregation, Pathological/complications , Animals , Humans
13.
FEBS Lett ; 592(14): 2383-2391, 2018 07.
Article in English | MEDLINE | ID: mdl-29790176

ABSTRACT

The ordered assembly of Tau protein into abnormal filamentous inclusions is a defining characteristic of many human neurodegenerative diseases. Thirty years ago, we reported that Tau is an integral component of the intraneuronal filaments of Alzheimer's disease. All six brain Tau isoforms make up those filaments. Twenty years ago, we and others showed that mutations in MAPT, the Tau gene, cause familial forms of frontotemporal dementia, thus proving that dysfunction of Tau protein is sufficient to cause neurodegeneration and dementia. More recently, we showed that high-resolution structures of Tau filaments from human brain can be determined by electron cryo-microscopy. These filaments may form the seeds that underlie the prion-like properties of aggregated tau.


Subject(s)
Neurodegenerative Diseases/metabolism , Protein Aggregates/physiology , tau Proteins/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Brain/metabolism , Humans , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Protein Aggregation, Pathological/complications , Protein Aggregation, Pathological/pathology , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , tau Proteins/chemistry , tau Proteins/genetics
14.
Curr Comput Aided Drug Des ; 14(2): 125-130, 2018.
Article in English | MEDLINE | ID: mdl-29564986

ABSTRACT

BACKGROUND: Transthyretin (TTR) is the transporter protein (55 kDa) that carries retinolbinding protein and Thyroxin (T4) in its functional tetramer form. Presence of the mutation in this protein (TTR) may lead to the dissociation of tetramers to monomer which unfolds and self-associates to form amyloid aggregates. Aggregation of this protein has been found to be associated with various lifethreatening disorders such as Coronary Artery Disease (CAD) which is the major cause of mortality and morbidity worldwide. METHODS: In the present communication, we have predicted mutation prone residues of TTR with the help of suspect server. Substitution (T139R with 95 score) occurring at the thyroid hormone binding site was selected for studying the mutational consequences on TTR. The effect of mutation on stability, functionality, aggregation and folding rate was analyzed by MuPro, DUET, SDM, SNAP2, Polyphen2, PASTA2.0, Aggrescan and Folding RaCe servers. The presence of TTR monomer in CAD plasma has been observed through Western blot analysis. RESULTS: T139R mutation may expose the buried regions of TTR protein which help in the self association and the increase in the stability may help in the TTR deposition. Structural analysis indicated that F and H strands of TTR are more prone to aggregation. Thus, T139R mutation might cause these residues to be aggregation prone and change in folding rate and validated TTR monomer in diseased cases by Western blot analysis. CONCLUSION: The observed results clearly indicated that the occurrence of this mutation is causing the impact on the structural and functional significance of TTR by interfering in the formation of tetramer. Thus, hindrance created to thyroxin transportation resulted in higher lipid levels in the blood that ultimately might promote the progression of the CAD.


Subject(s)
Heart Diseases/genetics , Point Mutation , Prealbumin/genetics , Protein Aggregation, Pathological/genetics , Amino Acid Substitution , Coronary Artery Disease/blood , Coronary Artery Disease/complications , Coronary Artery Disease/genetics , Heart Diseases/blood , Heart Diseases/complications , Humans , Prealbumin/chemistry , Protein Aggregates , Protein Aggregation, Pathological/blood , Protein Aggregation, Pathological/complications , Protein Conformation , Protein Folding , Protein Stability
15.
Acta Neuropathol Commun ; 5(1): 99, 2017 Dec 19.
Article in English | MEDLINE | ID: mdl-29258615

ABSTRACT

Emerging experimental evidence suggests that the spread of tau pathology in the brain in Tauopathies reflects the propagation of abnormal tau species along neuroanatomically connected brain areas. This propagation could occur through a "prion-like" mechanism involving transfer of abnormal tau seeds from a "donor cell" to a "recipient cell" and recruitment of normal tau in the latter to generate new tau seeds. This review critically appraises the evidence that the spread of tau pathology occurs via such a "prion-like" mechanism and proposes a number of recommendations for directing future research. Recommendations for definitions of frequently used terms in the tau field are presented in an attempt to clarify and standardize interpretation of research findings. Molecular and cellular factors affecting tau aggregation are briefly reviewed, as are potential contributions of physiological and pathological post-translational modifications of tau. Additionally, the experimental evidence for tau seeding and "prion-like" propagation of tau aggregation that has emerged from cellular assays and in vivo models is discussed. Propagation of tau pathology using "prion-like" mechanisms is expected to incorporate several steps including cellular uptake, templated seeding, secretion and intercellular transfer through synaptic and non-synaptic pathways. The experimental findings supporting each of these steps are reviewed. The clinical validity of these experimental findings is then debated by considering the supportive or contradictory findings from patient samples. Further, the role of physiological tau release in this scenario is examined because emerging data shows that tau is secreted but the physiological function (if any) of this secretion in the context of propagation of pathological tau seeds is unclear. Bona fide prions exhibit specific properties, including transmission from cell to cell, tissue to tissue and organism to organism. The propagation of tau pathology has so far not been shown to exhibit all of these steps and how this influences the debate of whether or not abnormal tau species can propagate in a "prion-like" manner is discussed. The exact nature of tau seeds responsible for propagation of tau pathology in human tauopathies remains controversial; it might be tightly linked to the existence of tau strains stably propagating peculiar patterns of neuropathological lesions, corresponding to the different patterns seen in human tauopathies. That this is a property shared by all seed-competent tau conformers is not yet firmly established. Further investigation is also required to clarify the relationship between propagation of tau aggregates and tau-induced toxicity. Genetic variants identified as risks factors for tauopathies might play a role in propagation of tau pathology, but many more studies are needed to document this. The contribution of selective vulnerability of neuronal populations, as an alternative to prion-like mechanisms to explain spreading of tau pathology needs to be clarified. Learning from the prion field will be helpful to enhance our understanding of propagation of tau pathology. Finally, development of better models is expected to answer some of these key questions and allow for the testing of propagation-centred therapies.


Subject(s)
Brain/pathology , Prions/pathogenicity , Protein Aggregation, Pathological/complications , Tauopathies/pathology , Humans , Models, Molecular
16.
Sci Rep ; 7(1): 9503, 2017 08 25.
Article in English | MEDLINE | ID: mdl-28842716

ABSTRACT

Renal ischemia/reperfusion is a major cause of acute kidney injury. However, the pathogenic mechanisms underlying renal ischemia/reperfusion injury (IRI) are not fully defined. Here, we investigated the role of PTEN, a dual protein/lipid phosphatase, in the development of ischemic AKI in mice. Pharmacological inhibition of PTEN with bpV(HOpic) exacerbated renal dysfunction and promoted tubular damage in mice with IRI compared with vehicle-treated mice with IRI. PTEN inhibition enhanced tubular cell apoptosis in kidneys with IRI, which was associated with excessive caspase-3 activation. Furthermore, PTEN inhibition expanded the infiltration of neutrophils and macrophages into kidneys with IRI, which was accompanied by increased expression of the proinflammatory molecules. These results have demonstrated that PTEN plays a crucial role in the pathogenesis of ischemic acute kidney injury through regulating tubular cell apoptosis and inflammation suggesting PTEN could be a potential therapeutic target for acute kidney injury.


Subject(s)
Acute Kidney Injury/etiology , Acute Kidney Injury/metabolism , Enzyme Inhibitors/pharmacology , PTEN Phosphohydrolase/antagonists & inhibitors , PTEN Phosphohydrolase/metabolism , Protein Aggregates/drug effects , Protein Aggregation, Pathological , Acute Kidney Injury/drug therapy , Acute Kidney Injury/physiopathology , Animals , Apoptosis/drug effects , Biomarkers , Cytokines/metabolism , Enzyme Activation , Immunohistochemistry , Inflammation Mediators/metabolism , Ischemia/complications , Ischemia/metabolism , Male , Mice , Protein Aggregation, Pathological/complications , Protein Aggregation, Pathological/metabolism , Protein Binding
17.
Neurobiol Dis ; 102: 105-112, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28286179

ABSTRACT

Huntington's disease (HD) is a genetic neurodegenerative disorder characterized by a triad of motor, psychiatric and cognitive deficits with the latter classically attributed to disruption of fronto-striatal circuits. However, emerging evidence suggests that some of the cognitive deficits in HD may have their origin in other structures including the hippocampus. Hippocampal abnormalities have been reported in HD mouse models particularly in terms of performance on the Morris Water Maze. However, in these animals, it is difficult to be certain whether the spatial memory deficits are due to local pathology within this structure or their poor mobility and motivation. Thus, a better model of hippocampal dysfunction in HD is needed especially given that we have previously shown that patients with HD have hippocampal-related problems from the very earliest stages of disease. In this study, our aim was therefore to understand the cellular and behavioural consequences of local overexpression of mutant huntingtin (mHTT) in the hippocampus of adult mice. We found that a targeted injection of a lentivirus, encoding an N-terminal of mHTT with 82 CAG repeats, into the murine hippocampus led to the focal formation of mHTT aggregates, long-term spatial memory impairments with decreased neurogenesis and expression of the immediate early gene c-fos. This study has therefore shown for the first time that local expression of mHTT in the dentate gyrus has deleterious effects, including its neurogenic capacity, with functional behavioural consequences, which fits well with recent data on hippocampal deficits seen in patients with HD.


Subject(s)
Hippocampus/metabolism , Huntingtin Protein/metabolism , Memory Disorders/metabolism , Neurogenesis/physiology , Protein Aggregation, Pathological/metabolism , Spatial Memory/physiology , Animals , Disease Models, Animal , Female , Genetic Vectors , Hippocampus/pathology , Humans , Huntingtin Protein/administration & dosage , Huntingtin Protein/genetics , Huntington Disease , Lentivirus , Maze Learning/physiology , Memory Disorders/etiology , Memory Disorders/pathology , Mice, Inbred C57BL , Mutation , Neurons/metabolism , Neurons/pathology , Protein Aggregation, Pathological/complications , Protein Aggregation, Pathological/pathology , Protein Aggregation, Pathological/psychology , Proto-Oncogene Proteins c-fos/metabolism
18.
Int J Neurosci ; 127(11): 1047-1057, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28110595

ABSTRACT

Proteins are major components of the biological functions in a cell. Biology demands that a protein must fold into its stable three-dimensional structure to become functional. In an unfavorable cellular environment, protein may get misfolded resulting in its aggregation. These conformational disorders are directly related to the tissue damage resulting in cellular dysfunction giving rise to different diseases. This way, several neurodegenerative diseases such as Alzheimer, Parkinson Huntington diseases and amyotrophic lateral sclerosis are caused. Misfolding of the protein is prevented by innate molecular chaperones of different classes. It is envisaged that work on this line is likely to translate the knowledge into the development of possible strategies for early diagnosis and efficient management of such related human diseases. The present review deals with the human neurodegenerative diseases caused due to the protein misfolding highlighting pathomechanisms and therapeutic intervention.


Subject(s)
Genetic Therapy/methods , Immunotherapy/methods , Molecular Chaperones , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/therapy , Protein Aggregation, Pathological/complications , Proteostasis Deficiencies/complications , Stem Cell Transplantation/methods , Animals , Humans , Neurodegenerative Diseases/drug therapy
19.
JAMA Neurol ; 74(2): 163-172, 2017 Feb 01.
Article in English | MEDLINE | ID: mdl-27918765

ABSTRACT

IMPORTANCE: Parkinson disease (PD) is a highly prevalent and incurable neurodegenerative disease associated with the accumulation of misfolded α-synuclein (αSyn) aggregates. An important problem in this disease is the lack of a sensitive, specific, and noninvasive biochemical diagnosis to help in clinical evaluation, monitoring of disease progression, and early differential diagnosis from related neurodegenerative diseases. OBJECTIVE: To develop a novel assay with high sensitivity and specificity to detect small quantities of αSyn aggregates circulating in cerebrospinal fluid (CSF) of patients affected by PD and related synucleinopathies. DESIGN, SETTING, AND PARTICIPANTS: The strategy evaluated in this proof-of-concept study uses the protein misfolding cyclic amplification (PMCA) technology that detects minute amounts of misfolded oligomers by taking advantage of their ability to nucleate further aggregation, enabling a very high amplification of the signal. The technology was first adapted with synthetic αSyn oligomers prepared in vitro and used to screen in 2 blinded cohorts of CSF samples from German and Japanese patients with PD (n = 76) and individuals serving as controls affected by other neurologic disorders (n = 65), neurodegenerative diseases (n = 18), and Alzheimer disease (n = 14). The kinetics of αSyn aggregation were measured by αSyn-PMCA in the presence of CSF samples from the participants to detect αSyn oligomeric seeds present in this biological fluid. The assays were conducted from November 15, 2013, to August 28, 2015. MAIN OUTCOMES AND MEASURES: Kinetic parameters correlated with disease severity at the time of sample collection, measured by the Hoehn and Yahr scale, with the lowest grade indicating unilateral involvement with minimal or no functional impairment, and the highest grade defining patients with complete confinement to wheelchair or bed. RESULTS: Studies with synthetic αSyn aggregates showed that αSyn-PMCA enabled to detect as little as 0.1 pg/mL of αSyn oligomers. The αSyn-PMCA signal was directly proportional to the amount of αSyn oligomers added to the reaction. A blinded study of CSF samples correctly identified patients affected by PD with an overall sensitivity of 88.5% (95% CI, 79.2%-94.6%) and specificity of 96.9% (95% CI, 89.3%-99.6%). The αSyn-PMCA results for different patients correlated with the severity of the clinical symptoms of PD (Japanese cohort: rs = -0.54, P = .006; German cohort: rs = -0.36, P = .02). CONCLUSIONS AND RELEVANCE: The findings suggest that detection of αSyn oligomers by αSyn-PMCA in the CSF of patients affected by PD may offer a good opportunity for a sensitive and specific biochemical diagnosis of the disease. Further studies are needed to investigate the usefulness of αSyn-PMCA to monitor disease progression and for preclinical identification of patients who may develop PD.


Subject(s)
Parkinson Disease , Protein Aggregation, Pathological/complications , Proteostasis Deficiencies/complications , alpha-Synuclein/cerebrospinal fluid , Amyloid beta-Peptides/cerebrospinal fluid , Amyloid beta-Peptides/chemistry , Biochemical Phenomena , Diagnostic Tests, Routine , Female , Humans , In Vitro Techniques , Lewy Body Disease/cerebrospinal fluid , Male , Multiple System Atrophy/cerebrospinal fluid , Outcome Assessment, Health Care , Parkinson Disease/cerebrospinal fluid , Parkinson Disease/diagnosis , Parkinson Disease/etiology , Peptide Fragments/cerebrospinal fluid , Peptide Fragments/chemistry , Predictive Value of Tests , Retrospective Studies , Sensitivity and Specificity , Severity of Illness Index , tau Proteins/cerebrospinal fluid , tau Proteins/chemistry
20.
Mol Genet Metab ; 116(4): 289-97, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26547561

ABSTRACT

Pantothenate kinase-associated neurodegeneration (PKAN) is a progressive movement disorder that is due to mutations in PANK2. Pathologically, it is a member of a class of diseases known as neurodegeneration with brain iron accumulation (NBIA) and features increased tissue iron and ubiquitinated proteinaceous aggregates in the globus pallidus. We have previously determined that these aggregates represent condensed residue derived from degenerated pallidal neurons. However, the protein content, other than ubiquitin, of these aggregates remains unknown. In the present study, we performed biochemical and immunohistochemical studies to characterize these aggregates and found them to be enriched in apolipoprotein E that is poorly soluble in detergent solutions. However, we did not determine a significant association between APOE genotype and the clinical phenotype of disease in our database of 81 cases. Rather, we frequently identified similar ubiquitin- and apolipoprotein E-enriched lesions in these neurons in non-PKAN patients in the penumbrae of remote infarcts that involve the globus pallidus, and occasionally in other brain sites that contain large γ-aminobutyric acid (GABA)ergic neurons. Our findings, taken together, suggest that tissue or cellular hypoxic/ischemic injury within the globus pallidus may underlie the pathogenesis of PKAN.


Subject(s)
Apolipoproteins E/chemistry , Brain Ischemia/genetics , GABAergic Neurons/chemistry , Pantothenate Kinase-Associated Neurodegeneration/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Protein Aggregation, Pathological/genetics , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Brain Ischemia/complications , Brain Ischemia/metabolism , Brain Ischemia/pathology , Case-Control Studies , Child , Female , GABAergic Neurons/metabolism , GABAergic Neurons/pathology , Gene Expression , Globus Pallidus/metabolism , Globus Pallidus/pathology , Humans , Male , Middle Aged , Mutation , Pantothenate Kinase-Associated Neurodegeneration/complications , Pantothenate Kinase-Associated Neurodegeneration/metabolism , Pantothenate Kinase-Associated Neurodegeneration/pathology , Phosphotransferases (Alcohol Group Acceptor)/deficiency , Protein Aggregation, Pathological/complications , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology , Ubiquitin/chemistry , Ubiquitin/genetics , Ubiquitin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...