Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 103
Filter
1.
ACS Chem Neurosci ; 15(7): 1533-1547, 2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38507813

ABSTRACT

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.


Subject(s)
Alzheimer Disease , Neurodegenerative Diseases , Parkinson Disease , Prion Diseases , Prions , Proteostasis Deficiencies , Humans , Mice , Animals , Neurodegenerative Diseases/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NF-kappa B/metabolism , Alzheimer Disease/metabolism , Neuroinflammatory Diseases , Down-Regulation , Parkinson Disease/metabolism , Neurons/metabolism , Prion Diseases/drug therapy , Prion Diseases/metabolism , Prions/metabolism , Inflammation/metabolism , Proteostasis Deficiencies/drug therapy , Proteostasis Deficiencies/metabolism
2.
Rev Neurol ; 77(11): 277-281, 2023 Dec 01.
Article in Spanish | MEDLINE | ID: mdl-38010785

ABSTRACT

The detection by biomarkers of the pathophysiological and molecular processes involved in misfolding protein diseases making it possible to delineate the natural history of these processes. The great majority of protein misfolding diseases have a prolonged preclinical phase, in which the biological changes are patent. The clinical manifestations (i.e., phenotypes) do not have a univocal correspondence with the underlying pathology, despite the fact that pathological eponyms have been used for the description of the clinical syndromes, which has favored diagnostic inaccuracy. In order to perform an adequate clinical management, we must know the 3 planes that currently define the most common neurodegenerative processes. Diagnostic accuracy will be a prerequisite for new therapies aimed at modifying the course of brain protein misfolding diseases.


TITLE: La nueva era de las enfermedades neurodegenerativas. La base de los nuevos abordajes.La detección por biomarcadores de los procesos fisiopatológicos y moleculares implicados en las enfermedades cerebrales por plegamiento anormal de proteínas está permitiendo delinear la historia natural de estos procesos. La gran mayoría de ellos tiene una fase preclínica prolongada, en la que los cambios biológicos son patentes. Las manifestaciones clínicas (fenotipos) no tienen una correspondencia unívoca con la patología subyacente, a pesar de que se han utilizado los epónimos anatomopatológicos para la descripción de los síndromes clínicos, lo que ha favorecido la imprecisión diagnóstica. Para realizar un adecuado manejo clínico debemos conocer los tres planos que definen actualmente los procesos neurodegenerativos más frecuentes. La precisión diagnóstica será un prerrequisito para las nuevas terapias dirigidas a modificar el curso de las enfermedades por plegamiento proteico cerebrales.


Subject(s)
Neurodegenerative Diseases , Proteostasis Deficiencies , Humans , Neurodegenerative Diseases/diagnosis , Proteins , Proteostasis Deficiencies/drug therapy , Proteostasis Deficiencies/pathology , Biomarkers
3.
Expert Opin Drug Discov ; 18(8): 881-891, 2023.
Article in English | MEDLINE | ID: mdl-37276120

ABSTRACT

INTRODUCTION: Protein misfolding diseases, including Alzheimer's and Parkinson's diseases, are characterized by the aberrant aggregation of proteins. These conditions are still largely untreatable, despite having a major impact on our healthcare systems and societies. AREAS COVERED: We describe drug discovery strategies to target protein misfolding and aggregation. We compare thermodynamic approaches, which are based on the stabilization of the native states of proteins, with kinetic approaches, which are based on the slowing down of the aggregation process. This comparison is carried out in terms of the current knowledge of the process of protein misfolding and aggregation, the mechanisms of disease and the therapeutic targets. EXPERT OPINION: There is an unmet need for disease-modifying treatments that target protein misfolding and aggregation for the over 50 human disorders known to be associated with this phenomenon. With the approval of the first drugs that can prevent misfolding or inhibit aggregation, future efforts will be focused on the discovery of effective compounds with these mechanisms of action for a wide range of conditions.


Subject(s)
Parkinson Disease , Proteostasis Deficiencies , Humans , Protein Folding , Proteins , Proteostasis Deficiencies/drug therapy , Proteostasis Deficiencies/prevention & control , Thermodynamics , Protein Aggregates
4.
Int J Biol Macromol ; 223(Pt A): 143-160, 2022 Dec 31.
Article in English | MEDLINE | ID: mdl-36356861

ABSTRACT

Most of the cell's chemical reactions and structural components are facilitated by proteins. But proteins are highly dynamic molecules, where numerous modifications or changes in the cellular environment can affect their native conformational fold leading to protein aggregation. Various stress conditions, such as oxidative stress, mutations and metal toxicity may cause protein misfolding and aggregation by shifting the conformational equilibrium towards more aggregation-prone states. Most of the protein misfolding diseases (PMDs) involve aggregation of protein. We have discussed such proteins like Aß peptide, α-synuclein, amylin and lysozyme involved in Alzheimer's, Parkinson's, type II diabetes and non-neuropathic systemic amyloidosis respectively. Till date, all advances in PMDs therapeutics help symptomatically but do not prevent the root cause of the disease, i.e., the aggregation of protein involved in the diseases. Current efforts focused on developing therapies for PMDs have employed diverse strategies; repositioning pre-existing drugs as it saves time and money; natural compounds that are touted as potential drug candidates have an advantage of being taken in diet normally and will induce lesser side effects. This review also covers recently developed therapeutic strategies like antisense drugs and disaggregases which has yielded therapeutic agents that have transitioned from preclinical studies into human clinical trials.


Subject(s)
Diabetes Mellitus, Type 2 , Proteostasis Deficiencies , Humans , Proteostasis Deficiencies/drug therapy , Proteostasis Deficiencies/prevention & control , Protein Aggregates , Protein Folding
5.
Drug Des Devel Ther ; 15: 3593-3604, 2021.
Article in English | MEDLINE | ID: mdl-34429588

ABSTRACT

INTRODUCTION: Endoplasmic reticulum (ER) stress condition is characterized as the accumulation of misfolded or unfolded proteins in lumen of ER. This condition has been implicated in various diseases and pathologies including ß-cell apoptosis, Alzheimer's disease and atherosclerosis. We have reported that hydroxynaphthoic acids (HNA), naphthalene analogues of salicylic acid (SA), reduced ER stress. In this study, we explored structural modification to bi-aryl analogues of SA. METHODS: Palladium-catalyzed cross-coupling was applied to synthesize bi-aryl analogues of SA. Anti-ER stress activity was monitored by using our cell-based assay system where ER stress is induced by tunicamycin. To monitor ER stress markers, ER stress was induced physiologically relevant palmitate system. RESULTS: Many analogues decreased ER stress signal induced by tunicamycin. Compounds creating dihedral angle between Ar group and SA moiety generally increased the activity but gave some cytotoxicity to indicate the crucial role of flat conformation of aromatic region. The best compound (16e) showed up to almost 6-fold and 90-fold better activity than 3-HNA and tauro-ursodeoxycholic acid, positive controls, respectively. ER stress markers such as p-PERK and p-JNK were accordingly decreased in Western blotting upon treatment of 16e under palmitate-induced condition. CONCLUSION: Anti-ER stress activity and toxicity profile of bi-aryl analogues of SA could provide a novel platform for potential therapy for protein misfolding diseases.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Endoplasmic Reticulum Stress/drug effects , Salicylates/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/chemistry , HEK293 Cells , Hep G2 Cells , Humans , Proteostasis Deficiencies/drug therapy , Proteostasis Deficiencies/pathology , Salicylates/chemical synthesis , Salicylates/chemistry , Structure-Activity Relationship , Tunicamycin
6.
Toxins (Basel) ; 13(6)2021 05 26.
Article in English | MEDLINE | ID: mdl-34073185

ABSTRACT

The B subunit pentamer verotoxin (VT aka Shiga toxin-Stx) binding to its cellular glycosphingolipid (GSL) receptor, globotriaosyl ceramide (Gb3) mediates internalization and the subsequent receptor mediated retrograde intracellular traffic of the AB5 subunit holotoxin to the endoplasmic reticulum. Subunit separation and cytosolic A subunit transit via the ER retrotranslocon as a misfolded protein mimic, then inhibits protein synthesis to kill cells, which can cause hemolytic uremic syndrome clinically. This represents one of the most studied systems of prokaryotic hijacking of eukaryotic biology. Similarly, the interaction of cholera AB5 toxin with its GSL receptor, GM1 ganglioside, is the key component of the gastrointestinal pathogenesis of cholera and follows the same retrograde transport pathway for A subunit cytosol access. Although both VT and CT are the cause of major pathology worldwide, the toxin-receptor interaction is itself being manipulated to generate new approaches to control, rather than cause, disease. This arena comprises two areas: anti neoplasia, and protein misfolding diseases. CT/CTB subunit immunomodulatory function and anti-cancer toxin immunoconjugates will not be considered here. In the verotoxin case, it is clear that Gb3 (and VT targeting) is upregulated in many human cancers and that there is a relationship between GSL expression and cancer drug resistance. While both verotoxin and cholera toxin similarly hijack the intracellular ERAD quality control system of nascent protein folding, the more widespread cell expression of GM1 makes cholera the toxin of choice as the means to more widely utilise ERAD targeting to ameliorate genetic diseases of protein misfolding. Gb3 is primarily expressed in human renal tissue. Glomerular endothelial cells are the primary VT target but Gb3 is expressed in other endothelial beds, notably brain endothelial cells which can mediate the encephalopathy primarily associated with VT2-producing E. coli infection. The Gb3 levels can be regulated by cytokines released during EHEC infection, which complicate pathogenesis. Significantly Gb3 is upregulated in the neovasculature of many tumours, irrespective of tumour Gb3 status. Gb3 is markedly increased in pancreatic, ovarian, breast, testicular, renal, astrocytic, gastric, colorectal, cervical, sarcoma and meningeal cancer relative to the normal tissue. VT has been shown to be effective in mouse xenograft models of renal, astrocytoma, ovarian, colorectal, meningioma, and breast cancer. These studies are herein reviewed. Both CT and VT (and several other bacterial toxins) access the cell cytosol via cell surface ->ER transport. Once in the ER they interface with the protein folding homeostatic quality control pathway of the cell -ERAD, (ER associated degradation), which ensures that only correctly folded nascent proteins are allowed to progress to their cellular destinations. Misfolded proteins are translocated through the ER membrane and degraded by cytosolic proteosome. VT and CT A subunits have a C terminal misfolded protein mimic sequence to hijack this transporter to enter the cytosol. This interface between exogenous toxin and genetically encoded endogenous mutant misfolded proteins, provides a new therapeutic basis for the treatment of such genetic diseases, e.g., Cystic fibrosis, Gaucher disease, Krabbe disease, Fabry disease, Tay-Sachs disease and many more. Studies showing the efficacy of this approach in animal models of such diseases are presented.


Subject(s)
Bacterial Toxins/therapeutic use , Animals , Cholera Toxin/therapeutic use , Endoplasmic Reticulum/metabolism , Gaucher Disease/drug therapy , Humans , Protein Subunits/therapeutic use , Proteostasis Deficiencies/drug therapy , Shiga Toxins/therapeutic use , Trihexosylceramides/metabolism
7.
Brain ; 144(10): 2915-2932, 2021 11 29.
Article in English | MEDLINE | ID: mdl-34019093

ABSTRACT

Neurodegenerative proteinopathies are characterized by progressive cell loss that is preceded by the mislocalization and aberrant accumulation of proteins prone to aggregation. Despite their different physiological functions, disease-related proteins like tau, α-synuclein, TAR DNA binding protein-43, fused in sarcoma and mutant huntingtin, all share low complexity regions that can mediate their liquid-liquid phase transitions. The proteins' phase transitions can range from native monomers to soluble oligomers, liquid droplets and further to irreversible, often-mislocalized aggregates that characterize the stages and severity of neurodegenerative diseases. Recent advances into the underlying pathogenic mechanisms have associated mislocalization and aberrant accumulation of disease-related proteins with defective nucleocytoplasmic transport and its mediators called karyopherins. These studies identify karyopherin abnormalities in amyotrophic lateral sclerosis, frontotemporal dementia, Alzheimer's disease, and synucleinopathies including Parkinson's disease and dementia with Lewy bodies, that range from altered expression levels to the subcellular mislocalization and aggregation of karyopherin α and ß proteins. The reported findings reveal that in addition to their classical function in nuclear import and export, karyopherins can also act as chaperones by shielding aggregation-prone proteins against misfolding, accumulation and irreversible phase-transition into insoluble aggregates. Karyopherin abnormalities can, therefore, be both the cause and consequence of protein mislocalization and aggregate formation in degenerative proteinopathies. The resulting vicious feedback cycle of karyopherin pathology and proteinopathy identifies karyopherin abnormalities as a common denominator of onset and progression of neurodegenerative disease. Pharmacological targeting of karyopherins, already in clinical trials as therapeutic intervention targeting cancers such as glioblastoma and viral infections like COVID-19, may therefore represent a promising new avenue for disease-modifying treatments in neurodegenerative proteinopathies.


Subject(s)
Karyopherins/metabolism , Neurodegenerative Diseases/metabolism , Proteostasis Deficiencies/metabolism , Animals , Brain/drug effects , Brain/metabolism , Humans , Karyopherins/genetics , Neurodegenerative Diseases/drug therapy , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Proteostasis Deficiencies/drug therapy
8.
Drug Discov Today ; 26(8): 1972-1979, 2021 08.
Article in English | MEDLINE | ID: mdl-33892144

ABSTRACT

Neurodegenerative disorders can arise as a result of amyloid-ß production and misfolding of its protein. The complex anatomy of the brain and the unresolved mechanics of the central nervous system hinder drug delivery; the brain is sheathed in a highly protective blood-brain barrier, a tightly packed layer of endothelial cells that restrict the entry of certain substances into the brain. Nanotechnology has achieved success in delivery to the brain, with preclinical assessments showing an acceptable concentration of active drugs in the therapeutic range, and nanoparticles can be fabricated to inhibit amyloid and enhance the delivery of the therapeutic molecule. This review focuses on the interactions of nanoparticles with amyloid-ß aggregates and provides an assessment of their theranostic potential.


Subject(s)
Drug Delivery Systems , Nanoparticles , Neurodegenerative Diseases/drug therapy , Amyloid beta-Peptides/metabolism , Animals , Blood-Brain Barrier/metabolism , Humans , Nanotechnology , Neurodegenerative Diseases/physiopathology , Proteostasis Deficiencies/drug therapy , Proteostasis Deficiencies/physiopathology , Theranostic Nanomedicine
9.
ChemMedChem ; 16(13): 2146-2156, 2021 07 06.
Article in English | MEDLINE | ID: mdl-33760394

ABSTRACT

Defective protein folding and accumulation of misfolded proteins is associated with neurodegenerative, cardiovascular, secretory, and metabolic disorders. Efforts are being made to identify small-molecule modulators or structural-correctors for conformationally destabilized proteins implicated in various protein aggregation diseases. Using a metastable-reporter-based primary screen, we evaluated pharmacological chaperone activity of a diverse class of natural products. We found that a flavonoid glycoside (C-10, chrysoeriol-7-O-ß-D-glucopyranoside) stabilizes metastable proteins, prevents its aggregation, and remodels the oligomers into protease-sensitive species. Data was corroborated with additional secondary screen with disease-specific pathogenic protein. In vitro and cell-based experiments showed that C-10 inhibits α-synuclein aggregation which is implicated in synucleinopathies-related neurodegeneration. C-10 interferes in its structural transition into ß-sheeted fibrils and mitigates α-synuclein aggregation-associated cytotoxic effects. Computational modeling suggests that C-10 binds to unique sites in α-synuclein which may interfere in its aggregation amplification. These findings open an avenue for comprehensive SAR development for flavonoid glycosides as pharmacological chaperones for metastable and aggregation-prone proteins implicated in protein conformational diseases.


Subject(s)
Biological Products/pharmacology , Flavonoids/pharmacology , Glycosides/pharmacology , Proteostasis Deficiencies/drug therapy , Biological Products/chemistry , Biological Products/isolation & purification , Cells, Cultured , Dose-Response Relationship, Drug , Flavonoids/chemistry , Flavonoids/isolation & purification , Glycosides/chemistry , Glycosides/isolation & purification , HEK293 Cells , Humans , Molecular Structure , Protein Folding/drug effects , Proteostasis Deficiencies/metabolism , Seeds/chemistry , Structure-Activity Relationship , Trigonella/chemistry , alpha-Synuclein/antagonists & inhibitors , alpha-Synuclein/metabolism
10.
Neurochem Int ; 145: 105011, 2021 05.
Article in English | MEDLINE | ID: mdl-33711400

ABSTRACT

Defective proteostasis is associated with the gradual accumulations of misfolded proteins and is a hallmark of many age-associated neurodegenerative diseases. In the aged brain, maintenance of the proteostasis network presents a substantial challenge, and its loss contributes to the onset and progression of neurological diseases associated with cognitive decline due to the generation of toxic protein aggregates, a process termed 'proteinopathy'. Emerging evidence suggests that reversing proteinopathies by boosting proteostasis might provide an effective means of preventing neurodegeneration. From this perspective, phytochemicals may play significant roles as potent modulators of the proteostasis network, as previous reports have suggested they can interact with various network components to modify pathologies and confer neuroprotection. This review focuses on some potent phytochemicals that directly or indirectly modulate the proteostasis network and on their possible molecular targets. In addition, we propose strategies for the natural product-based modulation of proteostasis machinery that target proteinopathies.


Subject(s)
Biological Products/administration & dosage , Biological Products/metabolism , Drug Delivery Systems/methods , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Proteostasis/drug effects , Animals , Humans , Neurodegenerative Diseases/pathology , Protein Aggregation, Pathological/drug therapy , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology , Proteostasis/physiology , Proteostasis Deficiencies/drug therapy , Proteostasis Deficiencies/metabolism , Proteostasis Deficiencies/pathology
11.
Yakugaku Zasshi ; 141(1): 1-13, 2021.
Article in Japanese | MEDLINE | ID: mdl-33390437

ABSTRACT

Chemical biology and structural development studies performed at The University of Tokyo during 1977-2020 are outlined chronologically. The studies are divided into three parts, i.e., (i) chemical biology of chemical carcinogenesis and molecular design of anti-tumor agents, (ii) structural development studies on biological response modifiers, and (iii) studies on so-called dramatype drug discovery focusing on pharmacological chaperones and protein knockdown-inducers. The first part describes analysis of DNA modification by Glu-P-1, which is a typical carcinogenic heterocyclic amine found in cooked foods, as well as molecular design of DNA-cleaving agents with anti-tumor properties. The second part deals with structural development studies of nuclear receptor ligands and various biological response modifiers derived from thalidomide, including the ligand superfamily concept and the multi-template strategy. The third part describes pharmacological chaperones that should be useful for the treatment of protein misfolding diseases, including Niemann-Pick type C disease and retinitis pigmentosa, and a protein knockdown strategy aimed at degradation of neurodegenerative-disease-causing polyglutamic aggregative proteins.


Subject(s)
Antineoplastic Agents , Drug Development/methods , Drug Development/trends , Drug Discovery/methods , Drug Discovery/trends , Immunologic Factors , Chemistry, Organic , Drug Design , Gene Knockdown Techniques , Humans , Ligands , Molecular Chaperones/therapeutic use , Polyglutamic Acid , Protein Folding , Proteostasis Deficiencies/drug therapy , Thalidomide/chemistry , Time Factors , Tokyo , Universities
12.
Front Biosci (Landmark Ed) ; 26(12): 1627-1642, 2021 12 30.
Article in English | MEDLINE | ID: mdl-34994177

ABSTRACT

Cells have evolved sophisticated molecular control systems to maximize the efficiency of the folding process. However, any subtle alteration of the environment or the protein can lead to misfolding or affect the conformational plasticity of the native states. It has been widely demonstrated that misfolding and/or conformational instability are the underlying mechanisms of several rare disorders caused by enzymatic deficits. In fact, disease-causing mutations often lead to the substitution of amino acids that are crucial for the achievement of a folded conformation, or play a role on the equilibrium between native-state conformers. One of the promising approaches to treat conformational disorders is the use of pharmacological chaperones (PCs), small molecules that specifically bind a target protein and stabilize a functional fold, thus increasing the amount of functionally active enzyme. Molecules acting as PCs are usually coenzymes, substrate analogues behaving as competitive inhibitors, or allosteric modulators. In this review, the general features of PCs are described, along with three examples of diseases (Gaucher disease, Phenylketonuria, and Primary Hyperoxaluria) in which this approach is currently under study at preclinical and/or clinical level.


Subject(s)
Gaucher Disease , Proteostasis Deficiencies , Amino Acids , Humans , Molecular Chaperones/metabolism , Protein Folding , Proteostasis Deficiencies/drug therapy , Proteostasis Deficiencies/genetics
13.
Inorg Chem ; 59(23): 17519-17534, 2020 Dec 07.
Article in English | MEDLINE | ID: mdl-33226796

ABSTRACT

PBT2 (5,7-dichloro-2-[(dimethylamino)methyl]-8-hydroxyquinoline) is a small Cu(II)-binding drug that has been investigated in the treatment of neurodegenerative diseases, namely, Alzheimer's disease (AD). PBT2 is thought to be highly effective at crossing the blood-brain barrier and has been proposed to exert anti-Alzheimer's effects through the modulation of metal ion concentrations in the brain, specifically the sequestration of Cu(II) from amyloid plaques. However, despite promising initial results in animal models and in clinical trials where PBT2 was shown to improve cognitive function, larger-scale clinical trials did not find PBT2 to have a significant effect on the amyloid plaque burden compared with controls. We propose that the results of these clinical trials likely point to a more complex mechanism of action for PBT2 other than simple Cu(II) sequestration. To this end, herein we have investigated the solution chemistry of Cu(II) coordination by PBT2 primarily using X-ray absorption spectroscopy (XAS), high-energy-resolution fluorescence-detected XAS, and electron paramagnetic resonance. We propose that a novel bis-PBT2 Cu(II) complex with asymmetric coordination may coexist in solution with a symmetric four-coordinate Cu(II)-bis-PBT2 complex distorted from coplanarity. Additionally, PBT2 is a more flexible ligand than other 8HQs because it can act as both a bidentate and a tridentate ligand as well as coordinate Cu(II) in both 1:1 and 2:1 PBT2/Cu(II) complexes.


Subject(s)
Alzheimer Disease/drug therapy , Chelating Agents/therapeutic use , Clioquinol/analogs & derivatives , Coordination Complexes/therapeutic use , Copper/therapeutic use , Neuroprotective Agents/pharmacology , Proteostasis Deficiencies/drug therapy , Animals , Chelating Agents/chemical synthesis , Chelating Agents/chemistry , Clioquinol/chemistry , Clioquinol/therapeutic use , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , Copper/chemistry , Density Functional Theory , Humans , Ligands , Molecular Structure , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/chemistry , X-Ray Absorption Spectroscopy
14.
Int J Mol Sci ; 21(10)2020 May 13.
Article in English | MEDLINE | ID: mdl-32414108

ABSTRACT

BACKGROUND: Accumulation of misfolded proteins is a common hallmark of several neurodegenerative disorders (NDs) which results from a failure or an impairment of the protein quality control (PQC) system. The PQC system is composed by chaperones and the degradative systems (proteasome and autophagy). Mutant proteins that misfold are potentially neurotoxic, thus strategies aimed at preventing their aggregation or at enhancing their clearance are emerging as interesting therapeutic targets for NDs. METHODS: We tested the natural alkaloid berberine (BBR) and some derivatives for their capability to enhance misfolded protein clearance in cell models of NDs, evaluating which degradative pathway mediates their action. RESULTS: We found that both BBR and its semisynthetic derivatives promote degradation of mutant androgen receptor (ARpolyQ) causative of spinal and bulbar muscular atrophy, acting mainly via proteasome and preventing ARpolyQ aggregation. Overlapping effects were observed on other misfolded proteins causative of amyotrophic lateral sclerosis, frontotemporal-lobar degeneration or Huntington disease, but with selective and specific action against each different mutant protein. CONCLUSIONS: BBR and its analogues induce the clearance of misfolded proteins responsible for NDs, representing potential therapeutic tools to counteract these fatal disorders.


Subject(s)
Berberine/pharmacology , Biological Products/pharmacology , Neurotoxicity Syndromes/drug therapy , Proteostasis Deficiencies/drug therapy , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Autophagy/drug effects , Autophagy/genetics , Cell Line , Frontotemporal Dementia/drug therapy , Frontotemporal Dementia/genetics , Frontotemporal Dementia/pathology , Humans , Molecular Chaperones/genetics , Mutant Proteins/genetics , Neurotoxicity Syndromes/genetics , Neurotoxicity Syndromes/pathology , Proteasome Endopeptidase Complex/drug effects , Proteasome Endopeptidase Complex/genetics , Protein Aggregation, Pathological/drug therapy , Protein Aggregation, Pathological/genetics , Proteostasis Deficiencies/genetics , Proteostasis Deficiencies/pathology
15.
Biomolecules ; 10(5)2020 05 07.
Article in English | MEDLINE | ID: mdl-32392767

ABSTRACT

An extensive catalog of plasma membrane (PM) protein mutations related to phenotypic diseases is associated with incorrect protein folding and/or localization. These impairments, in addition to dysfunction, frequently promote protein aggregation, which can be detrimental to cells. Here, we review PM protein processing, from protein synthesis in the endoplasmic reticulum to delivery to the PM, stressing the main repercussions of processing failures and their physiological consequences in pathologies, and we summarize the recent proposed therapeutic strategies to rescue misassembled proteins through different types of chaperones and/or small molecule drugs that safeguard protein quality control and regulate proteostasis.


Subject(s)
Channelopathies/metabolism , Membrane Proteins/metabolism , Molecular Chaperones/metabolism , Protein Folding , Proteostasis Deficiencies/metabolism , Animals , Channelopathies/drug therapy , Channelopathies/genetics , Humans , Membrane Proteins/chemistry , Protein Transport , Proteostasis Deficiencies/drug therapy , Proteostasis Deficiencies/genetics
16.
Eur J Med Chem ; 192: 112197, 2020 Apr 15.
Article in English | MEDLINE | ID: mdl-32172082

ABSTRACT

Protein misfolding diseases (PMDs) are chronic and progressive, with no effective therapy so far. Aggregation and misfolding of amyloidogenic proteins are closely associated with the onset and progression of PMDs, such as amyloid-ß (Aß) in Alzheimer's disease, α-Synuclein (α-Syn) in Parkinson's disease and human islet amyloid polypeptide (hIAPP) in type 2 diabetes. Inhibiting toxic aggregation of amyloidogenic proteins is regarded as a promising therapeutic approach in PMDs. The past decade has witnessed the rapid progresses of this field, dozens of inhibitors have been screened and verified in vitro and in vivo, demonstrating inhibitory effects against the aggregation and misfolding of amyloidogenic proteins, together with beneficial effects. Natural products are major sources of small molecule amyloid inhibitors, a number of natural derived compounds have been identified with great bioactivities and translational prospects. Here, we review the non-polyphenolic natural inhibitors that potentially applicable for PMDs treatment, along with their working mechanisms. Future directions are proposed for the development and clinical applications of these inhibitors.


Subject(s)
Amyloidogenic Proteins/antagonists & inhibitors , Biological Products/pharmacology , Proteostasis Deficiencies/drug therapy , Amyloidogenic Proteins/metabolism , Biological Products/chemistry , Humans , Molecular Structure , Protein Aggregates/drug effects , Proteostasis Deficiencies/metabolism
17.
Cell Mol Life Sci ; 77(6): 977-996, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31552448

ABSTRACT

The polyglutamine (polyQ) diseases are a group of nine fatal, adult-onset neurodegenerative disorders characterized by the misfolding and aggregation of mutant proteins containing toxic expansions of CAG/polyQ tracts. The heat shock protein 90 and 70 (Hsp90/Hsp70) chaperone machinery is a key component of cellular protein quality control, playing a role in the regulation of folding, aggregation, and degradation of polyQ proteins. The ability of Hsp70 to facilitate disaggregation and degradation of misfolded proteins makes it an attractive therapeutic target in polyQ diseases. Genetic studies have demonstrated that manipulation of Hsp70 and related co-chaperones can enhance the disaggregation and/or degradation of misfolded proteins in models of polyQ disease. Therefore, the development of small molecules that enhance Hsp70 activity is of great interest. However, it is still unclear if currently available Hsp70 modulators can selectively enhance disaggregation or degradation of misfolded proteins without perturbing other Hsp70 functions essential for cellular homeostasis. This review discusses the multifaceted role of Hsp70 in protein quality control and the opportunities and challenges Hsp70 poses as a potential therapeutic target in polyQ disease.


Subject(s)
HSP70 Heat-Shock Proteins/metabolism , Peptides/metabolism , Protein Aggregation, Pathological/metabolism , Animals , Humans , Huntington Disease/drug therapy , Huntington Disease/metabolism , Molecular Targeted Therapy , Muscular Atrophy, Spinal/drug therapy , Muscular Atrophy, Spinal/metabolism , Protein Aggregation, Pathological/drug therapy , Protein Folding/drug effects , Proteostasis Deficiencies/drug therapy , Proteostasis Deficiencies/metabolism , Spinocerebellar Ataxias/drug therapy , Spinocerebellar Ataxias/metabolism
18.
Curr Protein Pept Sci ; 20(12): 1226-1245, 2019.
Article in English | MEDLINE | ID: mdl-31187709

ABSTRACT

Protein folding is the process by which a polypeptide chain acquires its functional, native 3D structure. Protein misfolding, on the other hand, is a process in which protein fails to fold into its native functional conformation. This misfolding of proteins may lead to precipitation of a number of serious diseases such as Cystic Fibrosis (CF), Alzheimer's Disease (AD), Parkinson's Disease (PD), and Amyotrophic Lateral Sclerosis (ALS) etc. Protein Quality-control (PQC) systems, consisting of molecular chaperones, proteases and regulatory factors, help in protein folding and prevent its aggregation. At the same time, PQC systems also do sorting and removal of improperly folded polypeptides. Among the major types of PQC systems involved in protein homeostasis are cytosolic, Endoplasmic Reticulum (ER) and mitochondrial ones. The cytosol PQC system includes a large number of component chaperones, such as Nascent-polypeptide-associated Complex (NAC), Hsp40, Hsp70, prefoldin and T Complex Protein-1 (TCP-1) Ring Complex (TRiC). Protein misfolding diseases caused due to defective cytosolic PQC system include diseases involving keratin/collagen proteins, cardiomyopathies, phenylketonuria, PD and ALS. The components of PQC system of Endoplasmic Reticulum (ER) include Binding immunoglobulin Protein (BiP), Calnexin (CNX), Calreticulin (CRT), Glucose-regulated Protein GRP94, the thiol-disulphide oxidoreductases, Protein Disulphide Isomerase (PDI) and ERp57. ER-linked misfolding diseases include CF and Familial Neurohypophyseal Diabetes Insipidus (FNDI). The components of mitochondrial PQC system include mitochondrial chaperones such as the Hsp70, the Hsp60/Hsp10 and a set of proteases having AAA+ domains similar to the proteasome that are situated in the matrix or the inner membrane. Protein misfolding diseases caused due to defective mitochondrial PQC system include medium-chain acyl-CoA dehydrogenase (MCAD)/Short-chain Acyl-CoA Dehydrogenase (SCAD) deficiency diseases, hereditary spastic paraplegia. Among therapeutic approaches towards the treatment of various protein misfolding diseases, chaperones have been suggested as potential therapeutic molecules for target based treatment. Chaperones have been advantageous because of their efficient entry and distribution inside the cells, including specific cellular compartments, in therapeutic concentrations. Based on the chemical nature of the chaperones used for therapeutic purposes, molecular, chemical and pharmacological classes of chaperones have been discussed.


Subject(s)
Molecular Chaperones/chemistry , Proteostasis Deficiencies/drug therapy , Acyl-CoA Dehydrogenases/metabolism , Animals , Endoplasmic Reticulum/metabolism , High-Throughput Screening Assays/methods , Humans , Mitochondria/metabolism , Molecular Chaperones/pharmacology , Protein Conformation , Protein Folding/drug effects , Signal Transduction , Small Molecule Libraries/chemistry , Structure-Activity Relationship
19.
Neuropharmacology ; 161: 107572, 2019 12 15.
Article in English | MEDLINE | ID: mdl-30885608

ABSTRACT

Diseases arising from misfolding of SLC6 transporters have been reported over recent years, e.g. folding-deficient mutants of the dopamine transporter and of the glycine transporter-2 cause infantile/juvenile Parkinsonism dystonia and hyperekplexia, respectively. Mutations in the coding sequence of the human creatine transporter-1 (hCRT-1/SLC6A8) gene result in a creatine transporter deficiency syndrome, which varies in its clinical manifestation from epilepsy, mental retardation, autism, development delay and motor dysfunction to gastrointestinal symptoms. Some of the mutations in hCRT-1 occur at residues, which are highly conserved across the SLC6 family. Here, we examined 16 clinically relevant hCRT-1 variants to verify the conjecture that they were misfolded and that this folding defect was amenable to correction. Confocal microscopy imaging revealed that the heterologously expressed YFP-tagged mutant CRTs were trapped in the endoplasmic reticulum (ER), co-localised with the ER-resident chaperone calnexin. In contrast, the wild type hCRT-1 reached the plasma membrane. Preincubation of transiently transfected HEK293 cells with the chemical chaperone 4-phenylbutyrate (4-PBA) restored ER export and surface expression of as well as substrate uptake by several folding-deficient CRT-1 mutants. A representative mutant (hCRT-1-P544L) was expressed in rat primary hippocampal neurons to verify pharmacochaperoning in a target cell: 4-PBA promoted the delivery of hCRT-1-P544L to the neurite extensions. These observations show that several folding-deficient hCRT-1 mutants can be rescued. This proof-of-principle justifies the search for additional pharmacochaperones to restore folding of 4PBA-unresponsive hCRT-1 mutants. Finally, 4-PBA is an approved drug in paediatric use: this provides a rationale for translating the current insights into clinical trials. This article is part of the issue entitled 'Special Issue on Neurotransmitter Transporters'.


Subject(s)
Brain Diseases, Metabolic, Inborn/drug therapy , Creatine/deficiency , Mental Retardation, X-Linked/drug therapy , Nerve Tissue Proteins/drug effects , Phenylbutyrates/pharmacology , Plasma Membrane Neurotransmitter Transport Proteins/deficiency , Proteostasis Deficiencies/drug therapy , Animals , Brain Diseases, Metabolic, Inborn/genetics , Calnexin/metabolism , Cell Membrane/metabolism , Creatine/genetics , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , HEK293 Cells , Humans , Mental Retardation, X-Linked/genetics , Mutation/genetics , Nerve Tissue Proteins/genetics , Neurites/metabolism , Neurons/metabolism , Plasma Membrane Neurotransmitter Transport Proteins/drug effects , Plasma Membrane Neurotransmitter Transport Proteins/genetics , Primary Cell Culture , Proteostasis Deficiencies/genetics , Rats
20.
Int J Mol Sci ; 20(4)2019 Feb 19.
Article in English | MEDLINE | ID: mdl-30791416

ABSTRACT

The aim of this review is to critically analyze promises and limitations of pharmacological inducers of autophagy against protein misfolding-associated neurodegeneration. Effective therapies against neurodegenerative disorders can be developed by regulating the "self-defense" equipment of neurons, such as autophagy. Through the degradation and recycling of the intracellular content, autophagy promotes neuron survival in conditions of trophic factor deprivation, oxidative stress, mitochondrial and lysosomal damage, or accumulation of misfolded proteins. Autophagy involves the activation of self-digestive pathways, which is different for dynamics (macro, micro and chaperone-mediated autophagy), or degraded material (mitophagy, lysophagy, aggrephagy). All neurodegenerative disorders share common pathogenic mechanisms, including the impairment of autophagic flux, which causes the inability to remove the neurotoxic oligomers of misfolded proteins. Pharmacological activation of autophagy is typically achieved by blocking the kinase activity of mammalian target of rapamycin (mTOR) enzymatic complex 1 (mTORC1), removing its autophagy suppressor activity observed under physiological conditions; acting in this way, rapamycin provided the first proof of principle that pharmacological autophagy enhancement can induce neuroprotection through the facilitation of oligomers' clearance. The demand for effective disease-modifying strategies against neurodegenerative disorders is currently stimulating the development of a wide number of novel molecules, as well as the re-evaluation of old drugs for their pro-autophagic potential.


Subject(s)
Autophagy/drug effects , Drug Discovery , Neuroprotection/drug effects , Animals , Autophagy/genetics , Biomarkers , Drug Discovery/methods , Humans , Lysosomes/drug effects , Lysosomes/genetics , Lysosomes/metabolism , Mitochondria/drug effects , Mitochondria/genetics , Mitochondria/metabolism , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Protein Aggregates , Protein Aggregation, Pathological , Protein Binding , Protein Conformation , Protein Folding , Protein Multimerization , Proteostasis Deficiencies/drug therapy , Proteostasis Deficiencies/etiology , Proteostasis Deficiencies/metabolism , Proteostasis Deficiencies/pathology , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...