Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Prog Mol Biol Transl Sci ; 206: 341-388, 2024.
Article in English | MEDLINE | ID: mdl-38811085

ABSTRACT

A family of maladies known as amyloid disorders, proteinopathy, or amyloidosis, are characterized by the accumulation of abnormal protein aggregates containing cross-ß-sheet amyloid fibrils in many organs and tissues. Often, proteins that have been improperly formed or folded make up these fibrils. Nowadays, most treatments for amyloid illness focus on managing symptoms rather than curing or preventing the underlying disease process. However, recent advances in our understanding of the biology of amyloid diseases have led to the development of innovative therapies that target the emergence and accumulation of amyloid fibrils. Examples of these treatments include the use of small compounds, monoclonal antibodies, gene therapy, and others. In the end, even if the majority of therapies for amyloid diseases are symptomatic, greater research into the biology behind these disorders is identifying new targets for potential therapy and paving the way for the development of more effective treatments in the future.


Subject(s)
Amyloidosis , Humans , Animals , Amyloidosis/therapy , Amyloidosis/pathology , Amyloid/metabolism , Proteostasis Deficiencies/therapy , Genetic Therapy
2.
Brain Dev ; 45(5): 251-259, 2023 May.
Article in English | MEDLINE | ID: mdl-36870919

ABSTRACT

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.


Subject(s)
Molecular Chaperones , Proteostasis Deficiencies , Humans , Molecular Chaperones/metabolism , Proteostasis Deficiencies/genetics , Proteostasis Deficiencies/therapy , Proteostasis Deficiencies/metabolism , Mutation , Lysosomes/metabolism
3.
J Biol Chem ; 299(2): 102806, 2023 02.
Article in English | MEDLINE | ID: mdl-36529289

ABSTRACT

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.


Subject(s)
Molecular Chaperones , Peptide Fragments , Prions , RNA-Binding Protein FUS , beta Karyopherins , Humans , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/therapy , beta Karyopherins/chemistry , beta Karyopherins/genetics , beta Karyopherins/metabolism , Cell Line , Dependovirus/metabolism , Frontotemporal Dementia/metabolism , Frontotemporal Dementia/therapy , In Vitro Techniques , Molecular Chaperones/chemistry , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Prions/chemistry , Prions/metabolism , Proteostasis Deficiencies/metabolism , Proteostasis Deficiencies/therapy , RNA-Binding Protein FUS/chemistry , RNA-Binding Protein FUS/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Protein Binding
4.
Methods Mol Biol ; 2282: 377-394, 2021.
Article in English | MEDLINE | ID: mdl-33928585

ABSTRACT

Nanoparticles have been used to deliver siRNA to tissues and cells to silence specific genes in diverse organisms. Research and clinical application of nanoparticles like liposomes for drug delivery requires targeting them to specific anatomic regions or cell types, while avoiding off-target effects or clearance by the liver, kidney, or the immune system. Delivery to the central nervous system (CNS) presents additional challenges to cross the blood-brain barrier (BBB) to specific cell types like neurons, astrocytes, or glia. Here, we describe the generation of three different liposomal siRNA delivery vehicles to the CNS using the thin film hydration method. Utilizing cationic or anionic liposomes protects the siRNA from serum nucleases and proteases en route. To deliver the siRNA specifically to the CNS, the liposomes are complexed to a peptide that acts as a neuronal address by binding to nicotinic acetylcholine receptors (nAchRs). When injected intravenously or instilled intranasally, these liposome-siRNA-peptide complexes (LSPCs) or peptide addressed liposome-encapsulated therapeutic siRNA (PALETS) resist serum degradation, effectively cross the BBB, and deliver siRNA to AchR-expressing cells to suppress protein expression in the CNS.


Subject(s)
Central Nervous System Diseases/therapy , Gene Transfer Techniques , Lipids/chemistry , Proteins/chemistry , Proteostasis Deficiencies/therapy , RNA Interference , RNA, Small Interfering/genetics , RNAi Therapeutics , Animals , Blood-Brain Barrier/metabolism , Central Nervous System Diseases/genetics , Central Nervous System Diseases/metabolism , Liposomes , Mice, Inbred Strains , Protein Folding , Proteostasis Deficiencies/genetics , Proteostasis Deficiencies/metabolism , RNA, Small Interfering/chemistry , RNA, Small Interfering/metabolism , Receptors, Nicotinic/genetics , Receptors, Nicotinic/metabolism
6.
Brain Dev ; 43(1): 45-54, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32736903

ABSTRACT

In lysosomal diseases, enzyme deficiency is caused by misfolding of mutant enzyme protein with abnormal steric structure that is expressed by gene mutation. Chaperone therapy is a new molecular therapeutic approach primarily for lysosomal diseases. The misfolded mutant enzyme is digested rapidly or aggregated to induce endoplasmic reticulum stress. As a result, the catalytic activity is lost. The following sequence of events results in chaperone therapy to achieve correction of molecular pathology. An orally administered low molecular competitive inhibitor (chaperone) is absorbed into the bloodstream and reaches the target cells and tissues. The mutant enzyme is stabilized by the chaperone and subjected to normal enzyme proteinfolding (proteostasis). The first chaperone drug was developed for Fabry disease and is currently available in medical practice. At present three types of chaperones are available: competitive chaperone with enzyme inhibitory bioactivity (exogenous), non-competitive (or allosteric) chaperone without inhibitory bioactivity (exogenous), and molecular chaperone (heat shock protein; endogenous). The third endogenous chaperone would be directed to overexpression or activated by an exogenous low-molecular inducer. This new molecular therapeutic approach, utilizing the three types of chaperone, is expected to apply to a variety of diseases, genetic or non-genetic, and neurological or non-neurological, in addition to lysosomal diseases.


Subject(s)
Lysosomal Storage Diseases/therapy , Molecular Chaperones/therapeutic use , Proteostasis Deficiencies/therapy , Endoplasmic Reticulum Stress/physiology , Fabry Disease/drug therapy , Gangliosidosis, GM1/drug therapy , Humans , Lysosomal Storage Diseases/metabolism , Lysosomal Storage Diseases/physiopathology , Lysosomes/metabolism , Molecular Chaperones/metabolism , Proteostasis Deficiencies/metabolism , Proteostasis Deficiencies/physiopathology
7.
Int J Mol Sci ; 21(22)2020 Nov 17.
Article in English | MEDLINE | ID: mdl-33212787

ABSTRACT

The aberrant aggregation of proteins is implicated in the onset and pathogenesis of a wide range of neurodegenerative disorders, including Alzheimer's and Parkinson's diseases. Mounting evidence indicates that misfolded protein oligomers produced as intermediates in the aggregation process are potent neurotoxic agents in these diseases. Because of the transient and heterogeneous nature of these elusive aggregates, however, it has proven challenging to develop therapeutics that can effectively target them. Here, we review approaches aimed at reducing oligomer toxicity, including (1) modulating the oligomer populations (e.g., by altering the kinetics of aggregation by inhibiting, enhancing, or redirecting the process), (2) modulating the oligomer properties (e.g., through the size-hydrophobicity-toxicity relationship), (3) modulating the oligomer interactions (e.g., by protecting cell membranes by displacing oligomers), and (4) reducing oligomer toxicity by potentiating the protein homeostasis system. We analyze examples of these complementary approaches, which may lead to the development of compounds capable of preventing or treating neurodegenerative disorders associated with protein aggregation.


Subject(s)
Protein Aggregation, Pathological/therapy , Protein Multimerization , Proteostasis Deficiencies/therapy , Animals , Humans , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology , Proteostasis Deficiencies/metabolism , Proteostasis Deficiencies/pathology
8.
Acta Neuropathol ; 139(1): 3-25, 2020 01.
Article in English | MEDLINE | ID: mdl-31686182

ABSTRACT

The term "propagon" is used to define proteins that may transmit misfolding in vitro, in tissues or in organisms. Among propagons, misfolded tau is thought to be involved in the pathogenic mechanisms of various "tauopathies" that include Alzheimer's disease, progressive supranuclear palsy, and argyrophilic grain disease. Here, we review the available data in the literature and point out how the prion-like tau propagation has been extended from Alzheimer's disease to tauopathies. First, in Alzheimer's disease, the progression of tau aggregation follows stereotypical anatomical stages which may be considered as spreading. The mechanisms of the propagation are now subject to intensive and controversial research. It has been shown that tau may be secreted in the interstitial fluid in an active manner as reflected by high and constant concentration of extracellular tau during Alzheimer's pathology. Animal and cell models have been devised to mimic tau seeding and propagation, and despite their limitations, they have further supported to the prion-like propagation hypothesis. Finally, such new ways of thinking have led to different therapeutic strategies in anti-tau immunotherapy among tauopathies and have stimulated new clinical trials. However, it appears that the prion-like propagation hypothesis mainly relies on data obtained in Alzheimer's disease. From this review, it appears that further studies are needed (1) to characterize extracellular tau species, (2) to find the right pathological tau species to target, (3) to follow in vivo tau pathology by brain imaging and biomarkers and (4) to interpret current clinical trial results aimed at reducing the progression of these pathologies. Such inputs will be essential to have a comprehensive view of these promising therapeutic strategies in tauopathies.


Subject(s)
Immunotherapy/methods , Proteostasis Deficiencies/pathology , Tauopathies/pathology , Animals , Humans , Proteostasis Deficiencies/therapy , Tauopathies/therapy
9.
Prog Mol Biol Transl Sci ; 168: 277-287, 2019.
Article in English | MEDLINE | ID: mdl-31699321

ABSTRACT

Disease-modifying therapies for proteinopathies are urgently needed yet clinical trials for the major neurodegenerative diseases, Alzheimer's and Parkinson's, have been failing at an alarming rate leaving patients and caregivers scrambling for any sign of hope. At the same time, for one family of proteinopathies, the rare TTR amyloidoses, disease-modifying therapy has existed for almost 3 decades and two new types of disease-modifying therapy have become available more recently. In this chapter, I discuss those therapies, examine to what extent they can be generalized for other diseases, and consider what we may learn from their relative success.


Subject(s)
Neurodegenerative Diseases/therapy , Proteostasis Deficiencies/therapy , Animals , Humans , Neurodegenerative Diseases/pathology , Proteostasis Deficiencies/pathology
10.
Lab Invest ; 99(7): 929-942, 2019 07.
Article in English | MEDLINE | ID: mdl-30918326

ABSTRACT

More than 40 different neurological diseases are caused by microsatellite repeat expansions. Since the discovery of repeat-associated non-AUG (RAN) translation by Zu et al. in 2011, nine expansion disorders have been identified as RAN-positive diseases. RAN proteins are translated from different types of nucleotide repeat expansions and can be produced from both sense and antisense transcripts. In some diseases, RAN proteins have been shown to accumulate in affected brain regions. Here we review the pathological and molecular aspects associated with RAN protein accumulation for each particular disorder, the correlation between disease pathology and the available in vivo models and the common aspects shared by some of the newly discovered RAN proteins.


Subject(s)
Microsatellite Repeats , Nervous System Diseases/genetics , Proteostasis Deficiencies/etiology , Animals , Gene Expression , Humans , Proteostasis Deficiencies/therapy
11.
J Neurosci ; 38(33): 7248-7254, 2018 08 15.
Article in English | MEDLINE | ID: mdl-30012684

ABSTRACT

Deregulation of cellular proteostasis due to the failure of the ubiquitin proteasome system to dispose of misfolded aggregation-prone proteins is a hallmark of various neurodegenerative diseases in humans. Microorganisms have evolved to survive massive protein misfolding and aggregation triggered by heat shock using their protein-unfolding ATPases (unfoldases) from the Hsp100 family. Because the Hsp100 chaperones are absent in homoeothermic mammals, we hypothesized that the vulnerability of mammalian neurons to misfolded proteins could be mitigated by expressing a xenogeneic unfoldase. To test this idea, we expressed proteasome-activating nucleotidase (PAN), a protein-unfolding ATPase from thermophilic Archaea, which is homologous to the 19S eukaryotic proteasome and similar to the Hsp100 family chaperones in rod photoreceptors of mice. We found that PAN had no obvious effect in healthy rods; however, it effectively counteracted protein-misfolding retinopathy in Gγ1 knock-out mice. We conclude that archaeal PAN can rescue a protein-misfolding neurodegenerative disease, likely by recognizing misfolded mammalian proteins.SIGNIFICANCE STATEMENT This study demonstrates successful therapeutic application of an archaeal molecular chaperone in an animal model of neurodegenerative disease. Introducing the archaeal protein-unfolding ATPase proteasome-activating nucleotidase (PAN) into the retinal photoreceptors of mice protected these neurons from the cytotoxic effect of misfolded proteins. We propose that xenogeneic protein-unfolding chaperones could be equally effective against other types of neurodegenerative diseases of protein-misfolding etiology.


Subject(s)
Adenosine Triphosphatases/physiology , Archaeal Proteins/physiology , Genetic Therapy , Methanocaldococcus/enzymology , Protein Folding , Proteostasis Deficiencies/therapy , Retinal Degeneration/therapy , Retinal Rod Photoreceptor Cells/metabolism , Adenosine Triphosphatases/genetics , Animals , Archaeal Proteins/genetics , Disease Models, Animal , Female , GTP-Binding Protein gamma Subunits/deficiency , GTP-Binding Protein gamma Subunits/genetics , Genes, Synthetic , HEK293 Cells , Humans , Methanocaldococcus/genetics , Mice , Mice, Knockout , Mice, Transgenic , Promoter Regions, Genetic , Proteasome Endopeptidase Complex/metabolism , Recombinant Fusion Proteins/metabolism , Retinal Degeneration/enzymology , Retinal Degeneration/genetics , Retinal Rod Photoreceptor Cells/pathology , Rhodopsin/genetics , Transfection , Transgenes
12.
Curr Protein Pept Sci ; 19(8): 805-812, 2018.
Article in English | MEDLINE | ID: mdl-29708066

ABSTRACT

Protein function is dependent on assumption of the correct three-dimensional structure, achieved through the folding process. As a central element in ensuring cellular homeostasis, proteostasis i.e. the control of correct protein folding, trafficking and degradation, is a highly regulated process ensured by three integrated molecular pathways: i) the unfolded protein response (UPR) which is activated by the engulfment of misfolded proteins and results in protein re-folding through the expression of chaperones; ii) the ubiquitin-proteasome system (UPS) which 'flags' misfolded proteins with ubiquitin, directing them to the 26S proteasome for proteolytic degradation; iii) autophagy that, through lysosomes, removes misfolded or aggregated proteins. All three of these proteostatic controls can be impaired by the aging process and by pathological mutations highlighting the potential role of proteostasis in conditions associated with aging such as neurodegeneration, type 2 diabetes and cancer. Indeed, neurodegenerative diseases are characterised by an interconnected triumvirate of deregulated proteostasis, neuroinflammation (i.e. the uncontrolled activation of microglial cells), and oxidative stress (i.e. the unbuffered increase in reactive oxygen species). The transcription factor Nrf2, classically associated with protection against oxidative stress, can also modulate the UPR, UPS and autophagy, while inhibiting the activation of NF-kB, the key transcription factor of the inflammatory response. In this review we focus on recent data from our laboratory and others demonstrating that the protective Nrf2 pathway can be activated by the endogenous cyclic dipeptide (His-Pro), thereby driving neuroprotective effects in different pathological settings. In this context we discuss the possible utility of clyclo (His-Pro) as a promising future therapeutic option for protein misfolding disorders.


Subject(s)
Neurodegenerative Diseases/metabolism , Peptides, Cyclic/metabolism , Piperazines/metabolism , Proteostasis , Animals , Autophagy , Cell Death , Cell Survival , Humans , NF-kappa B/metabolism , Neurodegenerative Diseases/therapy , Oxidative Stress , Protein Conformation , Protein Folding , Proteolysis , Proteostasis Deficiencies/metabolism , Proteostasis Deficiencies/therapy , Signal Transduction
13.
Physiol Rev ; 98(2): 697-725, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29442594

ABSTRACT

After synthesis, proteins are folded into their native conformations aided by molecular chaperones. Dysfunction in folding caused by genetic mutations in numerous genes causes protein conformational diseases. Membrane proteins are more prone to misfolding due to their more intricate folding than soluble proteins. Misfolded proteins are detected by the cellular quality control systems, especially in the endoplasmic reticulum, and proteins may be retained there for eventual degradation by the ubiquitin-proteasome system or through autophagy. Some misfolded proteins aggregate, leading to pathologies in numerous neurological diseases. In vitro, modulating mutant protein folding by altering molecular chaperone expression can ameliorate some misfolding. Some small molecules known as chemical chaperones also correct mutant protein misfolding in vitro and in vivo. However, due to their lack of specificity, their potential as therapeutics is limited. Another class of compounds, known as pharmacological chaperones (pharmacoperones), binds with high specificity to misfolded proteins, either as enzyme substrates or receptor ligands, leading to decreased folding energy barriers and correction of the misfolding. Because many of the misfolded proteins are misrouted but do not have defects in function per se, pharmacoperones have promising potential in advancing to the clinic as therapeutics, since correcting routing may ameliorate the underlying mechanism of disease. This review will comprehensively summarize this exciting area of research, surveying the literature from in vitro studies in cell lines to transgenic animal models and clinical trials in several protein misfolding diseases.


Subject(s)
Endoplasmic Reticulum/metabolism , Molecular Chaperones/metabolism , Protein Transport/physiology , Proteostasis Deficiencies/metabolism , Animals , Humans , Molecular Chaperones/genetics , Protein Conformation , Protein Folding , Protein Transport/genetics , Proteostasis Deficiencies/therapy
14.
RNA ; 24(4): 486-498, 2018 04.
Article in English | MEDLINE | ID: mdl-29295891

ABSTRACT

Polyglutamine (polyQ) diseases are a class of progressive neurodegenerative disorders characterized by the expression of both expanded CAG RNA and misfolded polyQ protein. We previously reported that the direct interaction between expanded CAG RNA and nucleolar protein nucleolin (NCL) impedes preribosomal RNA (pre-rRNA) transcription, and eventually triggers nucleolar stress-induced apoptosis in polyQ diseases. Here, we report that a 21-amino acid peptide, named "beta-structured inhibitor for neurodegenerative diseases" (BIND), effectively suppresses toxicity induced by expanded CAG RNA. When administered to a cell model, BIND potently inhibited cell death induced by expanded CAG RNA with an IC50 value of ∼0.7 µM. We showed that the function of BIND is dependent on Glu2, Lys13, Gly14, Ile18, Glu19, and Phe20. BIND treatment restored the subcellular localization of nucleolar marker protein and the expression level of pre-45s rRNA Through isothermal titration calorimetry analysis, we demonstrated that BIND suppresses nucleolar stress via a direct interaction with CAG RNA in a length-dependent manner. The mean binding constants (KD) of BIND to SCA2CAG22 , SCA2CAG42 , SCA2CAG55 , and SCA2CAG72 RNA are 17.28, 5.60, 4.83, and 0.66 µM, respectively. In vivo, BIND ameliorates retinal degeneration and climbing defects, and extends the lifespan of Drosophila expressing expanded CAG RNA. These effects suggested that BIND can suppress neurodegeneration in diverse polyQ disease models in vivo and in vitro without exerting observable cytotoxic effect. Our results collectively demonstrated that BIND is an effective inhibitor of expanded CAG RNA-induced toxicity in polyQ diseases.


Subject(s)
Huntington Disease/therapy , Peptides/pharmacology , Proteostasis Deficiencies/genetics , Spinocerebellar Ataxias/therapy , Trinucleotide Repeats/genetics , Animals , Cell Death/drug effects , Drosophila/genetics , HEK293 Cells , Humans , Huntington Disease/genetics , Huntington Disease/pathology , Peptides/metabolism , Phosphoproteins/genetics , Protein Folding , Proteostasis Deficiencies/pathology , Proteostasis Deficiencies/therapy , RNA, Ribosomal/genetics , RNA-Binding Proteins/genetics , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/pathology , Transcription, Genetic/genetics , Trinucleotide Repeats/drug effects , Nucleolin
15.
Prog Mol Biol Transl Sci ; 150: 123-145, 2017.
Article in English | MEDLINE | ID: mdl-28838658

ABSTRACT

Early genetic studies on scrapie, an infectious neurodegenerative disease of sheep that was adapted to mice, provided evidence in support of the hypothesis that the agent was a slow virus with a nucleic acid genome independent of the host. Particularly compelling support for an independent genome came from the existence of strains of scrapie agent, some of which were true breeding, while others appeared to mutate under selective pressure. Kuru, a neurodegenerative disease in the remote highlands of Papua New Guinea, had pathological changes similar to those in scrapie and also proved to be transmissible. Genetic studies with the tools of molecular biology and transgenic mice forced a reevaluation of earlier work and supported the prion hypothesis of a novel pathogen devoid of nucleic acid. In this chapter, I discuss the contributions of classical and molecular genetics to understanding PrP prion diseases and to determining that heritable information is enciphered in protein conformation.


Subject(s)
Genetic Predisposition to Disease , Prion Diseases/genetics , Prion Proteins/chemistry , Prion Proteins/metabolism , Proteostasis Deficiencies/genetics , Animals , Humans , Prion Diseases/therapy , Protein Conformation , Proteostasis Deficiencies/therapy
16.
Int J Biol Macromol ; 105(Pt 1): 993-1000, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28743576

ABSTRACT

This review aims to highlight the fundamental mechanism of protein misfolding leading to protein aggregation and associated diseases. It also aims to anticipate novel therapeutic strategies with which to prevent or treat these highly debilitating conditions linked to these pathologies. The failure of a protein to correctly fold de novo or to remain correctly folded can have profound consequences on a living system especially when the cellular quality control processes fail to eliminate the rogue proteins. The core cause of over 20 different human diseases which have now been designated as 'conformational diseases' including neurodegenerative diseases such as Alzheimer's disease (AD), Huntington's disease (HD) and Parkinson's disease (PD) etc. A comprehensive study on protein misfolding, aggregation, and the outcomes of the effects of cytotoxic aggregates will lead to understand the aggregation-mediated cell toxicity and serves as a foundation for future research in development of promising therapies and drugs. This review has also shed light on the mechanism of protein misfolding which leads to its aggregation and hence the neurodegeneration. From these considerations, one could also envisage the possibility that protein aggregation may be exploited by nature to perform specific physiological functions in differing biological contexts.


Subject(s)
Protein Aggregates , Proteostasis Deficiencies/prevention & control , Amyloid/chemistry , Animals , Humans , Neurodegenerative Diseases/prevention & control , Neurodegenerative Diseases/therapy , Protein Aggregates/drug effects , Protein Folding/drug effects , Proteostasis Deficiencies/therapy
17.
Biochem Biophys Res Commun ; 482(3): 450-453, 2017 01 15.
Article in English | MEDLINE | ID: mdl-28212730

ABSTRACT

A variety of different forms of cellular stress can cause protein misfolding and aggregation and proteotoxicity. The cytoprotective response to proteotoxicity is termed the integrated stress response and involves 4 distinct serine/threonine protein kinases that converge on the translation initiation factor eIF2α, resulting in phosphorylation at S51, cell cycle arrest, and a general inhibition of global protein synthesis. Phosphorylation of eIF2α also promotes translation of ATF4 and the expression of ATF4 target genes that ameliorate proteotoxic stress but can also promote apoptosis. This mini review provides a general overview of these mechanisms and discusses how the inter-tumor heterogeneity that involves them affects sensitivity and resistance to proteasome inhibitors, a new class of cancer therapeutics that promotes tumor cell killing via proteotoxic stress.


Subject(s)
Neoplasms/metabolism , Neoplasms/therapy , Activating Transcription Factor 4/metabolism , Animals , Apoptosis , Eukaryotic Initiation Factor-2/metabolism , Humans , Phosphorylation , Proteasome Inhibitors/therapeutic use , Protein Serine-Threonine Kinases/metabolism , Proteostasis Deficiencies/metabolism , Proteostasis Deficiencies/therapy , Stress, Physiological , eIF-2 Kinase
18.
Expert Opin Ther Targets ; 21(1): 37-49, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27786579

ABSTRACT

INTRODUCTION: There is increasing evidence that endoplasmic reticulum (ER) chaperones Protein Disulphide Isomerase (PDI) and ERp57 (endoplasmic reticulum protein 57) are protective against neurodegenerative diseases related to protein misfolding, including Amyotrophic Lateral Sclerosis (ALS). PDI and ERp57 also possess disulphide interchange activity, in which protein disulphide bonds are oxidized, reduced and isomerized, to form their native conformation. Recently, missense and intronic variants of PDI and ERp57 were associated with ALS, implying that PDI proteins are relevant to ALS pathology. Areas covered: Here, we discuss possible implications of the PDI and ERp57 variants, as well as recent studies describing previously unrecognized roles for PDI and ERp57 in the nervous system. Therapeutics based on PDI may therefore be attractive candidates for ALS. However, in addition to its protective functions, aberrant, toxic roles for PDI have recently been described. These functions need to be fully characterized before effective therapeutic strategies can be designed. Expert opinion: These disease-associated variants of PDI and ERp57 provide additional evidence for an important role for PDI proteins in ALS. However, there are many questions remaining unanswered that need to be addressed before the potential of the PDI family in relation to ALS can be fully realized.


Subject(s)
Amyotrophic Lateral Sclerosis/physiopathology , Protein Disulfide-Isomerases/metabolism , Amyotrophic Lateral Sclerosis/therapy , Animals , Drug Design , Humans , Molecular Targeted Therapy , Proteostasis Deficiencies/physiopathology , Proteostasis Deficiencies/therapy
19.
Dis Model Mech ; 9(8): 823-38, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27491084

ABSTRACT

Chaperones and co-chaperones enable protein folding and degradation, safeguarding the proteome against proteotoxic stress. Chaperones display dynamic responses to exogenous and endogenous stressors and thus constitute a key component of the proteostasis network (PN), an intricately regulated network of quality control and repair pathways that cooperate to maintain cellular proteostasis. It has been hypothesized that aging leads to chronic stress on the proteome and that this could underlie many age-associated diseases such as neurodegeneration. Understanding the dynamics of chaperone function during aging and disease-related proteotoxic stress could reveal specific chaperone systems that fail to respond to protein misfolding. Through the use of suppressor and enhancer screens, key chaperones crucial for proteostasis maintenance have been identified in model organisms that express misfolded disease-related proteins. This review provides a literature-based analysis of these genetic studies and highlights prominent chaperone modifiers of proteotoxicity, which include the HSP70-HSP40 machine and small HSPs. Taken together, these studies in model systems can inform strategies for therapeutic regulation of chaperone functionality, to manage aging-related proteotoxic stress and to delay the onset of neurodegenerative diseases.


Subject(s)
Molecular Chaperones/metabolism , Proteostasis Deficiencies/pathology , Animals , Disease Models, Animal , HSP70 Heat-Shock Proteins/metabolism , Homeostasis , Humans , Molecular Targeted Therapy , Proteostasis Deficiencies/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...