Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 873
Filter
1.
Int J Mol Sci ; 25(8)2024 Apr 20.
Article in English | MEDLINE | ID: mdl-38674104

ABSTRACT

ABCA4-related retinopathy is the most common inherited Mendelian eye disorder worldwide, caused by biallelic variants in the ATP-binding cassette transporter ABCA4. To date, over 2200 ABCA4 variants have been identified, including missense, nonsense, indels, splice site and deep intronic defects. Notably, more than 60% are missense variants that can lead to protein misfolding, mistrafficking and degradation. Currently no approved therapies target ABCA4. In this study, we demonstrate that ABCA4 misfolding variants are temperature-sensitive and reduced temperature growth (30 °C) improves their traffic to the plasma membrane, suggesting the folding of these variants could be rescuable. Consequently, an in vitro platform was developed for the rapid and robust detection of ABCA4 traffic to the plasma membrane in transiently transfected cells. The system was used to assess selected candidate small molecules that were reported to improve the folding or traffic of other ABC transporters. Two candidates, 4-PBA and AICAR, were identified and validated for their ability to enhance both wild-type ABCA4 and variant trafficking to the cell surface in cell culture. We envision that this platform could serve as a primary screen for more sophisticated in vitro testing, enabling the discovery of breakthrough agents to rescue ABCA4 protein defects and mitigate ABCA4-related retinopathy.


Subject(s)
ATP-Binding Cassette Transporters , Protein Folding , Protein Transport , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Humans , Protein Folding/drug effects , HEK293 Cells , Cell Membrane/metabolism , Small Molecule Libraries/pharmacology
2.
Part Fibre Toxicol ; 19(1): 33, 2022 05 10.
Article in English | MEDLINE | ID: mdl-35538581

ABSTRACT

BACKGROUND: Copper oxide (CuO) nanoparticles (NPs) are known to trigger cytotoxicity in a variety of cell models, but the mechanism of cell death remains unknown. Here we addressed the mechanism of cytotoxicity in macrophages exposed to CuO NPs versus copper chloride (CuCl2). METHODS: The mouse macrophage cell line RAW264.7 was used as an in vitro model. Particle uptake and the cellular dose of Cu were investigated by transmission electron microscopy (TEM) and inductively coupled plasma mass spectrometry (ICP-MS), respectively. The deposition of Cu in lysosomes isolated from macrophages was also determined by ICP-MS. Cell viability (metabolic activity) was assessed using the Alamar Blue assay, and oxidative stress was monitored by a variety of methods including a luminescence-based assay for cellular glutathione (GSH), and flow cytometry-based detection of mitochondrial superoxide and mitochondrial membrane potential. Protein aggregation was determined by confocal microscopy using an aggresome-specific dye and protein misfolding was determined by circular dichroism (CD) spectroscopy. Lastly, proteasome activity was investigated using a fluorometric assay. RESULTS: We observed rapid cellular uptake of CuO NPs in macrophages with deposition in lysosomes. CuO NP-elicited cell death was characterized by mitochondrial swelling with signs of oxidative stress including the production of mitochondrial superoxide and cellular depletion of GSH. We also observed a dose-dependent accumulation of polyubiquitinated proteins and loss of proteasomal function in CuO NP-exposed cells, and we could demonstrate misfolding and mitochondrial translocation of superoxide dismutase 1 (SOD1), a Cu/Zn-dependent enzyme that plays a pivotal role in the defense against oxidative stress. The chelation of copper ions using tetrathiomolybdate (TTM) prevented cell death whereas inhibition of the cellular SOD1 chaperone aggravated toxicity. Moreover, CuO NP-triggered cell death was insensitive to the pan-caspase inhibitor, zVAD-fmk, and to wortmannin, an inhibitor of autophagy, implying that this was a non-apoptotic cell death. ZnO NPs, on the other hand, triggered autophagic cell death. CONCLUSIONS: CuO NPs undergo dissolution in lysosomes leading to copper-dependent macrophage cell death characterized by protein misfolding and proteasomal insufficiency. Specifically, we present novel evidence for Cu-induced SOD1 misfolding which accords with the pronounced oxidative stress observed in CuO NP-exposed macrophages. These results are relevant for our understanding of the consequences of inadvertent human exposure to CuO NPs.


Subject(s)
Macrophages , Metal Nanoparticles , Nanoparticles , Superoxide Dismutase-1 , Animals , Cell Death/drug effects , Copper , Glutathione/metabolism , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Metal Nanoparticles/toxicity , Mice , Nanoparticles/chemistry , Nanoparticles/metabolism , Nanoparticles/toxicity , Oxidative Stress , Protein Folding/drug effects , RAW 264.7 Cells , Superoxide Dismutase-1/metabolism , Superoxides
3.
J Med Chem ; 65(7): 5212-5243, 2022 04 14.
Article in English | MEDLINE | ID: mdl-35377645

ABSTRACT

In cystic fibrosis (CF), the deletion of phenylalanine 508 (F508del) in the CF transmembrane conductance regulator (CFTR) leads to misfolding and premature degradation of the mutant protein. These defects can be targeted with pharmacological agents named potentiators and correctors. During the past years, several efforts have been devoted to develop and approve new effective molecules. However, their clinical use remains limited, as they fail to fully restore F508del-CFTR biological function. Indeed, the search for CFTR correctors with different and additive mechanisms has recently increased. Among them, drugs that modulate the CFTR proteostasis environment are particularly attractive to enhance therapy effectiveness further. This Perspective focuses on reviewing the recent progress in discovering CFTR proteostasis regulators, mainly describing the design, chemical structure, and structure-activity relationships. The opportunities, challenges, and future directions in this emerging and promising field of research are discussed, as well.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator , Cystic Fibrosis , Proteostasis , Cystic Fibrosis/drug therapy , Cystic Fibrosis/genetics , Cystic Fibrosis/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/drug effects , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Humans , Mutant Proteins/drug effects , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation , Protein Folding/drug effects , Proteostasis/drug effects , Proteostasis/physiology
4.
Cell Mol Life Sci ; 79(4): 192, 2022 Mar 16.
Article in English | MEDLINE | ID: mdl-35292885

ABSTRACT

The advent of Trikafta (Kaftrio in Europe) (a triple-combination therapy based on two correctors-elexacaftor/tezacaftor-and the potentiator ivacaftor) has represented a revolution for the treatment of patients with cystic fibrosis (CF) carrying the most common misfolding mutation, F508del-CFTR. This therapy has proved to be of great efficacy in people homozygous for F508del-CFTR and is also useful in individuals with a single F508del allele. Nevertheless, the efficacy of this therapy needs to be improved, especially in light of the extent of its use in patients with rare class II CFTR mutations. Using CFBE41o- cells expressing F508del-CFTR, we provide mechanistic evidence that targeting the E1 ubiquitin-activating enzyme (UBA1) by TAK-243, a small molecule in clinical trials for other diseases, boosts the rescue of F508del-CFTR induced by CFTR correctors. Moreover, TAK-243 significantly increases the F508del-CFTR short-circuit current induced by elexacaftor/tezacaftor/ivacaftor in differentiated human primary airway epithelial cells, a gold standard for the pre-clinical evaluation of patients' responsiveness to pharmacological treatments. This new combinatory approach also leads to an improvement in CFTR conductance on cells expressing other rare CF-causing mutations, including N1303K, for which Trikafta is not approved. These findings show that Trikafta therapy can be improved by the addition of a drug targeting the misfolding detection machinery at the beginning of the ubiquitination cascade and may pave the way for an extension of Trikafta to low/non-responding rare misfolded CFTR mutants.


Subject(s)
Aminophenols/administration & dosage , Benzodioxoles/administration & dosage , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/drug therapy , Cystic Fibrosis/genetics , Indoles/administration & dosage , Pyrazoles/administration & dosage , Pyridines/administration & dosage , Pyrimidines/administration & dosage , Pyrrolidines/administration & dosage , Quinolones/administration & dosage , Sulfides/administration & dosage , Sulfonamides/administration & dosage , Ubiquitin-Activating Enzymes/antagonists & inhibitors , Cells, Cultured , Cystic Fibrosis/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Drug Synergism , Drug Therapy, Combination , Enzyme Inhibitors/administration & dosage , Humans , Mutation , Protein Folding/drug effects , Sequence Deletion
5.
J Biol Chem ; 298(3): 101612, 2022 03.
Article in English | MEDLINE | ID: mdl-35065969

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease in which motor neurons progressively and rapidly degenerate, eventually leading to death. The first protein found to contain ALS-associated mutations was copper/zinc superoxide dismutase 1 (SOD1), which is conformationally stable when it contains its metal ligands and has formed its native intramolecular disulfide. Mutations in SOD1 reduce protein folding stability via disruption of metal binding and/or disulfide formation, resulting in misfolding, aggregation, and ultimately cellular toxicity. A great deal of effort has focused on preventing the misfolding and aggregation of SOD1 as a potential therapy for ALS; however, the results have been mixed. Here, we utilize a small-molecule polytherapy of diacetylbis(N(4)-methylthiosemicarbazonato)copper(II) (CuATSM) and ebselen to mimic the metal delivery and disulfide bond promoting activity of the cellular chaperone of SOD1, the "copper chaperone for SOD1." Using microscopy with automated image analysis, we find that polytherapy using CuATSM and ebselen is highly effective and acts in synergy to reduce inclusion formation in a cell model of SOD1 aggregation for multiple ALS-associated mutants. Polytherapy reduces mutant SOD1-associated cell death, as measured by live-cell microscopy. Measuring dismutase activity via zymography and immunoblotting for disulfide formation showed that polytherapy promoted more effective maturation of transfected SOD1 variants beyond either compound alone. Our data suggest that a polytherapy of CuATSM and ebselen may merit more study as an effective method of treating SOD1-associated ALS.


Subject(s)
Amyotrophic Lateral Sclerosis , Organocopper Compounds , Superoxide Dismutase-1 , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Biomimetic Materials/pharmacology , Copper/metabolism , Disulfides/chemistry , Humans , Isoindoles/pharmacology , Molecular Chaperones/metabolism , Mutation , Organocopper Compounds/pharmacology , Organoselenium Compounds/pharmacology , Protein Folding/drug effects , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism
6.
ChemMedChem ; 17(2): e202100611, 2022 01 19.
Article in English | MEDLINE | ID: mdl-34704363

ABSTRACT

The development of imaging agents for in vivo detection of alpha-synuclein (α-syn) pathologies faces several challenges. A major gap in the field is the lack of diverse molecular scaffolds with high affinity and selectivity to α-syn fibrils for in vitro screening assays. Better in vitro scaffolds can instruct the discovery of better in vivo agents. We report the rational design, synthesis, and in vitro evaluation of a series of novel 1-indanone and 1,3-indandione derivatives from a Structure-Activity Relationship (SAR) study centered on some existing α-syn fibril binding ligands. Our results from fibril saturation binding experiments show that two of the lead candidates compounds 8 and 32 bind α-syn fibrils with binding constants (Kd ) of 9.0 and 18.8 nM, respectively, and selectivity of greater than 10× for α-syn fibrils compared with amyloid-ß (Aß) and tau fibrils. Our results demonstrate that the lead ligands avidly label all forms of α-syn on PD brain tissue sections, but only the dense core of senile plaques in AD brain tissue, respectively. These results are corroborated by ligand-antibody colocalization data from Syn211, which shows immunoreactivity toward all forms of α-syn aggregates, and Syn303, which displays preferential reactivity toward mature Lewy pathology. Our results reveal that 1-indanone derivatives have desirable properties for the biological evaluation of α-synucleinopathies.


Subject(s)
Alzheimer Disease/drug therapy , Indans/pharmacology , Neuroprotective Agents/pharmacology , alpha-Synuclein/antagonists & inhibitors , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Dose-Response Relationship, Drug , Drug Design , Humans , Indans/chemical synthesis , Indans/chemistry , Ligands , Molecular Structure , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/chemistry , Protein Aggregates/drug effects , Protein Folding/drug effects , Structure-Activity Relationship , alpha-Synuclein/metabolism
7.
Plant Physiol ; 188(1): 241-254, 2022 01 20.
Article in English | MEDLINE | ID: mdl-34609517

ABSTRACT

Disulfide bonds play essential roles in the folding of secretory and plasma membrane proteins in the endoplasmic reticulum (ER). In eukaryotes, protein disulfide isomerase (PDI) is an enzyme catalyzing the disulfide bond formation and isomerization in substrates. The Arabidopsis (Arabidopsis thaliana) genome encodes diverse PDIs including structurally distinct subgroups PDI-L and PDI-M/S. It remains unclear how these AtPDIs function to catalyze the correct disulfide formation. We found that one Arabidopsis ER oxidoreductin-1 (Ero1), AtERO1, can interact with multiple PDIs. PDI-L members AtPDI2/5/6 mainly serve as an isomerase, while PDI-M/S members AtPDI9/10/11 are more efficient in accepting oxidizing equivalents from AtERO1 and catalyzing disulfide bond formation. Accordingly, the pdi9/10/11 triple mutant exhibited much stronger inhibition than pdi1/2/5/6 quadruple mutant under dithiothreitol treatment, which caused disruption of disulfide bonds in plant proteins. Furthermore, AtPDI2/5 work synergistically with PDI-M/S members in relaying disulfide bonds from AtERO1 to substrates. Our findings reveal the distinct but overlapping roles played by two structurally different AtPDI subgroups in oxidative protein folding in the ER.


Subject(s)
Arabidopsis/genetics , Arabidopsis/metabolism , Catalysis/drug effects , Disulfides/metabolism , Oxidation-Reduction/drug effects , Protein Disulfide-Isomerases/metabolism , Protein Folding/drug effects , Genetic Variation , Genotype , Mutation , Protein Disulfide-Isomerases/genetics
8.
Nutrients ; 13(12)2021 Dec 08.
Article in English | MEDLINE | ID: mdl-34959950

ABSTRACT

The impact of dietary advanced glycation end products (dAGEs) on human health has been discussed in many studies but, to date, no consensual pathophysiological process has been demonstrated. The intestinal absorption pathways which have so far been described for dAGEs, the passive diffusion of free AGE adducts and transport of glycated di-tripeptides by the peptide transporter 1 (PEPT-1), are not compatible with certain pathophysiological processes described. To get new insight into the intestinal absorption pathways and the pathophysiological mechanisms of dAGEs, we initiated an in vivo study with a so-called simple animal model with a complete digestive tract, Caenorhabditis elegans. Dietary bacteria were chemically modified with glyoxylic acid to mainly produce Nε-carboxymethyllysine (CML) and used to feed the worms. We performed different immunotechniques using an anti-CML antibody for the relative quantification of ingested CML and localization of this AGE in the worms' intestine. The relative expression of genes encoding different biological processes such as response to stresses and intestinal digestion were determined. The physiological development of the worms was verified. All the results were compared with those obtained with the control bacteria. The results revealed a new route for the intestinal absorption of dietary CML (dCML), endocytosis, which could be mediated by scavenger receptors. The exposure of worms to dCML induced a reproductive defect and a transcriptional response reflecting oxidative, carbonyl and protein folding stresses. These data, in particular the demonstration of endocytosis of dCML by enterocytes, open up new perspectives to better characterize the pathophysiological mechanisms of dAGEs.


Subject(s)
Caenorhabditis elegans/metabolism , Endocytosis/drug effects , Glycation End Products, Advanced/adverse effects , Glycation End Products, Advanced/metabolism , Intestinal Absorption/drug effects , Lysine/analogs & derivatives , Animals , Enterocytes/metabolism , Gastrointestinal Tract/metabolism , Lysine/administration & dosage , Lysine/adverse effects , Models, Animal , Oxidative Stress/drug effects , Protein Folding/drug effects , Reproduction/drug effects
9.
Int J Mol Sci ; 22(22)2021 Nov 18.
Article in English | MEDLINE | ID: mdl-34830348

ABSTRACT

Dysfunction of cellular homeostasis can lead to misfolding of proteins thus acquiring conformations prone to polymerization into pathological aggregates. This process is associated with several disorders, including neurodegenerative diseases, such as Parkinson's disease (PD), and endoplasmic reticulum storage disorders (ERSDs), like alpha-1-antitrypsin deficiency (AATD) and hereditary hypofibrinogenemia with hepatic storage (HHHS). Given the shared pathophysiological mechanisms involved in such conditions, it is necessary to deepen our understanding of the basic principles of misfolding and aggregation akin to these diseases which, although heterogeneous in symptomatology, present similarities that could lead to potential mutual treatments. Here, we review: (i) the pathological bases leading to misfolding and aggregation of proteins involved in PD, AATD, and HHHS: alpha-synuclein, alpha-1-antitrypsin, and fibrinogen, respectively, (ii) the evidence linking each protein aggregation to the stress mechanisms occurring in the endoplasmic reticulum (ER) of each pathology, (iii) a comparison of the mechanisms related to dysfunction of proteostasis and regulation of homeostasis between the diseases (such as the unfolded protein response and/or autophagy), (iv) and clinical perspectives regarding possible common treatments focused on improving the defensive responses to protein aggregation for diseases as different as PD, and ERSDs.


Subject(s)
Afibrinogenemia/genetics , Fibrinogen/chemistry , Parkinson Disease/genetics , alpha 1-Antitrypsin Deficiency/genetics , alpha 1-Antitrypsin/chemistry , alpha-Synuclein/chemistry , Afibrinogenemia/drug therapy , Afibrinogenemia/metabolism , Afibrinogenemia/pathology , Animals , Autophagy/drug effects , Autophagy/genetics , Coagulants/therapeutic use , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Fibrinogen/genetics , Fibrinogen/metabolism , Gene Expression Regulation , Humans , Liver/metabolism , Liver/pathology , Neuroprotective Agents/therapeutic use , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Parkinson Disease/pathology , Protease Inhibitors/therapeutic use , Protein Aggregates/drug effects , Protein Folding/drug effects , Unfolded Protein Response/drug effects , alpha 1-Antitrypsin/genetics , alpha 1-Antitrypsin/metabolism , alpha 1-Antitrypsin Deficiency/drug therapy , alpha 1-Antitrypsin Deficiency/metabolism , alpha 1-Antitrypsin Deficiency/pathology , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
10.
Int J Mol Sci ; 22(22)2021 Nov 19.
Article in English | MEDLINE | ID: mdl-34830391

ABSTRACT

Parkinson's disease (PD) is the second most common neurodegenerative disorder. An important hallmark of PD involves the pathological aggregation of proteins in structures known as Lewy bodies. The major component of these proteinaceous inclusions is alpha (α)-synuclein. In different conditions, α-synuclein can assume conformations rich in either α-helix or ß-sheets. The mechanisms of α-synuclein misfolding, aggregation, and fibrillation remain unknown, but it is thought that ß-sheet conformation of α-synuclein is responsible for its associated toxic mechanisms. To gain fundamental insights into the process of α-synuclein misfolding and aggregation, the secondary structure of this protein in the presence of charged and non-charged surfactant solutions was characterized. The selected surfactants were (anionic) sodium dodecyl sulphate (SDS), (cationic) cetyltrimethylammonium chloride (CTAC), and (uncharged) octyl ß-D-glucopyranoside (OG). The effect of surfactants in α-synuclein misfolding was assessed by ultra-structural analyses, in vitro aggregation assays, and secondary structure analyses. The α-synuclein aggregation in the presence of negatively charged SDS suggests that SDS-monomer complexes stimulate the aggregation process. A reduction in the electrostatic repulsion between N- and C-terminal and in the hydrophobic interactions between the NAC (non-amyloid beta component) region and the C-terminal seems to be important to undergo aggregation. Fourier transform infrared spectroscopy (FTIR) measurements show that ß-sheet structures comprise the assembly of the fibrils.


Subject(s)
Neurodegenerative Diseases/drug therapy , Parkinson Disease/drug therapy , Protein Aggregation, Pathological/drug therapy , alpha-Synuclein/genetics , Amyloid/antagonists & inhibitors , Amyloid/genetics , Cetrimonium/pharmacology , Circular Dichroism , Galactosides/pharmacology , Humans , Lewy Bodies/drug effects , Lewy Bodies/ultrastructure , Neurodegenerative Diseases/pathology , Parkinson Disease/genetics , Parkinson Disease/pathology , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/pathology , Protein Conformation , Protein Conformation, beta-Strand/genetics , Protein Folding/drug effects , Protein Structure, Secondary/drug effects , Sodium Dodecyl Sulfate/pharmacology , Spectroscopy, Fourier Transform Infrared , alpha-Synuclein/antagonists & inhibitors
11.
J Mol Biol ; 433(24): 167323, 2021 12 03.
Article in English | MEDLINE | ID: mdl-34695381

ABSTRACT

Misfolding and aggregation of alpha-synuclein (αS) within dopaminergic neurons is a key factor in the development and progression of a group of age-related neurodegenerative diseases, termed synucleinopathies, that include Parkinson's disease (PD). We previously derived a peptide inhibitor from a 209,952-member intracellular library screen by employing the preNAC region (45-54) as a design template. At least six single-point mutations firmly linked to early-onset Parkinson's disease (E46K, H50Q, G51D, A53T/E/V) are located within this region, strongly implicating a pathogenic role within αS that leads to increased cytotoxicity. A library-derived ten residue peptide, 4554W, was consequently shown to block αS aggregation at the point of primary nucleation via lipid induction, inhibiting its conversion into downstream cytotoxic species. Here we couple truncation with a full alanine scan analysis, to establish the effect upon the αS aggregation pathway relative to 4554W. This revealed the precise residues responsible for eliciting inhibitory interaction and function, as well as those potentially amenable to modification or functionalisation. We find that modification N6A combined with N-terminal truncation results in a peptide of significantly increased efficacy. Importantly, our data demonstrate that the peptide does not directly disrupt αS lipid-binding, a desirable trait since antagonists of αS aggregation and toxicity should not impede association with small synaptic neurotransmitter vesicles, and thus not disrupt dopaminergic vesicle fusion and recycling. This work paves the way toward the major aim of deriving a highly potent peptide antagonist of αS pathogenicity without impacting on native αS function.


Subject(s)
Antiparkinson Agents/chemistry , Parkinson Disease/metabolism , Peptidomimetics/chemistry , Protein Aggregates/drug effects , Protein Folding/drug effects , alpha-Synuclein/antagonists & inhibitors , alpha-Synuclein/chemistry , Alanine/chemistry , Alanine/genetics , Antiparkinson Agents/pharmacology , Cryoelectron Microscopy , Cytoplasmic Vesicles/metabolism , Dopaminergic Neurons/metabolism , Humans , Lipids/chemistry , Parkinson Disease/genetics , Peptide Library , Peptidomimetics/pharmacology , Point Mutation , alpha-Synuclein/genetics
12.
Biomolecules ; 11(10)2021 10 16.
Article in English | MEDLINE | ID: mdl-34680161

ABSTRACT

Rhodopsin (RHO) misfolding mutations are a common cause of the blinding disease autosomal dominant retinitis pigmentosa (adRP). The most prevalent mutation, RHOP23H, results in its misfolding and retention in the endoplasmic reticulum (ER). Under homeostatic conditions, misfolded proteins are selectively identified, retained at the ER, and cleared via ER-associated degradation (ERAD). Overload of these degradation processes for a prolonged period leads to imbalanced proteostasis and may eventually result in cell death. ERAD of misfolded proteins, such as RHOP23H, includes the subsequent steps of protein recognition, targeting for ERAD, retrotranslocation, and proteasomal degradation. In the present study, we investigated and compared pharmacological modulation of ERAD at these four different major steps. We show that inhibition of the VCP/proteasome activity favors cell survival and suppresses P23H-mediated retinal degeneration in RHOP23H rat retinal explants. We suggest targeting this activity as a therapeutic approach for patients with currently untreatable adRP.


Subject(s)
Endoplasmic Reticulum/drug effects , Retinal Degeneration/genetics , Retinitis Pigmentosa/genetics , Rhodopsin/genetics , Alkaloids/pharmacology , Animals , Animals, Genetically Modified , Benzoquinones/pharmacology , Disease Models, Animal , Endoplasmic Reticulum/genetics , Humans , Lactams, Macrocyclic/pharmacology , Mutation/genetics , Photoreceptor Cells, Vertebrate/drug effects , Photoreceptor Cells, Vertebrate/pathology , Proteasome Endopeptidase Complex/drug effects , Proteasome Endopeptidase Complex/genetics , Protein Folding/drug effects , Proteolysis/drug effects , Rats , Retina/drug effects , Retina/growth & development , Retina/pathology , Retinal Degeneration/pathology , Retinitis Pigmentosa/pathology , Rhodopsin/ultrastructure
13.
Molecules ; 26(19)2021 Oct 07.
Article in English | MEDLINE | ID: mdl-34641604

ABSTRACT

Heavy metals enter the human body through the gastrointestinal tract, skin, or via inhalation. Toxic metals have proven to be a major threat to human health, mostly because of their ability to cause membrane and DNA damage, and to perturb protein function and enzyme activity. These metals disturb native proteins' functions by binding to free thiols or other functional groups, catalyzing the oxidation of amino acid side chains, perturbing protein folding, and/or displacing essential metal ions in enzymes. The review shows the physiological and biochemical effects of selected toxic metals interactions with proteins and enzymes. As environmental contamination by heavy metals is one of the most significant global problems, some detoxification strategies are also mentioned.


Subject(s)
Biodegradation, Environmental , Environmental Exposure , Metals, Heavy/toxicity , Protein Binding/drug effects , Binding Sites , Cosmetics/toxicity , DNA Damage , Environmental Pollutants/toxicity , Enzymes/drug effects , Food/toxicity , Humans , Protein Folding/drug effects
14.
Cell Mol Life Sci ; 78(23): 7813-7829, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34714360

ABSTRACT

Protein misfolding is involved in a large number of diseases, among which cystic fibrosis. Complex intra- and inter-domain folding defects associated with mutations in the cystic fibrosis transmembrane regulator (CFTR) gene, among which p.Phe508del (F508del), have recently become a therapeutical target. Clinically approved correctors such as VX-809, VX-661, and VX-445, rescue mutant protein. However, their binding sites and mechanisms of action are still incompletely understood. Blind docking onto the 3D structures of both the first membrane-spanning domain (MSD1) and the first nucleotide-binding domain (NBD1), followed by molecular dynamics simulations, revealed the presence of two potential VX-809 corrector binding sites which, when mutated, abrogated rescue. Network of amino acids in the lasso helix 2 and the intracellular loops ICL1 and ICL4 allosterically coupled MSD1 and NBD1. Corrector VX-445 also occupied two potential binding sites on MSD1 and NBD1, the latter being shared with VX-809. Binding of both correctors on MSD1 enhanced the allostery between MSD1 and NBD1, hence the increased efficacy of the corrector combination. These correctors improve both intra-domain folding by stabilizing fragile protein-lipid interfaces and inter-domain assembly via distant allosteric couplings. These results provide novel mechanistic insights into the rescue of misfolded proteins by small molecules.


Subject(s)
Aminopyridines/pharmacology , Benzodioxoles/pharmacology , Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , Cystic Fibrosis/drug therapy , Mutation , Protein Folding/drug effects , Pyrazoles/pharmacology , Pyridines/pharmacology , Pyrrolidines/pharmacology , Binding Sites , Chloride Channel Agonists/pharmacology , Cystic Fibrosis/genetics , Cystic Fibrosis/metabolism , Cystic Fibrosis/pathology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Drug Therapy, Combination , HEK293 Cells , Humans , Protein Domains , Protein Structure, Tertiary
15.
Biochem J ; 478(15): 2953-2975, 2021 08 13.
Article in English | MEDLINE | ID: mdl-34375386

ABSTRACT

The Unfolded Protein response is an adaptive pathway triggered upon alteration of endoplasmic reticulum (ER) homeostasis. It is transduced by three major ER stress sensors, among which the Inositol Requiring Enzyme 1 (IRE1) is the most evolutionarily conserved. IRE1 is an ER-resident type I transmembrane protein exhibiting an ER luminal domain that senses the protein folding status and a catalytic kinase and RNase cytosolic domain. In recent years, IRE1 has emerged as a relevant therapeutic target in various diseases including degenerative, inflammatory and metabolic pathologies and cancer. As such several drugs altering IRE1 activity were developed that target either catalytic activity and showed some efficacy in preclinical pathological mouse models. In this review, we describe the different drugs identified to target IRE1 activity as well as their mode of action from a structural perspective, thereby identifying common and different modes of action. Based on this information we discuss on how new IRE1-targeting drugs could be developed that outperform the currently available molecules.


Subject(s)
Endoplasmic Reticulum Stress/physiology , Endoplasmic Reticulum/metabolism , Endoribonucleases/metabolism , Homeostasis/physiology , Protein Serine-Threonine Kinases/metabolism , Unfolded Protein Response/physiology , Animals , Endoplasmic Reticulum/drug effects , Endoribonucleases/antagonists & inhibitors , Endoribonucleases/chemistry , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Homeostasis/drug effects , Humans , Protein Folding/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/chemistry , Signal Transduction/drug effects , Signal Transduction/physiology , Unfolded Protein Response/drug effects
16.
J Phys Chem Lett ; 12(32): 7659-7664, 2021 Aug 19.
Article in English | MEDLINE | ID: mdl-34351767

ABSTRACT

From stem cell freeze-drying to organ storage, considerable recent efforts have been directed toward the development of new preservation technologies. A prominent protein stabilizing strategy involves vitrification in glassy matrices, most notably those formed of sugars such as the biologically relevant preservative trehalose. Here, we compare the folding thermodynamics of a model miniprotein in solution and in the glassy state of the sugars trehalose and glucose. Using synchrotron radiation circular dichroism (SRCD), we find that the same native structure persists in solution and glass. However, upon transition to the glass, a completely different, conformationally restricted unfolded state replaces the disordered denatured state found in solution, potentially inhibiting misfolding. Concomitantly, a large exothermic contribution is observed in glass, exposing the stabilizing effect of interactions with the sugar matrix on the native state. Our results shed light on the mechanism of protein stabilization in sugar glass and should aid in future preservation technologies.


Subject(s)
Protein Conformation/drug effects , Protein Stability/drug effects , Proteins/metabolism , Trehalose/chemistry , Amino Acid Sequence , Protein Folding/drug effects , Proteins/chemistry , Thermodynamics , Vitrification
17.
Int J Mol Sci ; 22(16)2021 Aug 10.
Article in English | MEDLINE | ID: mdl-34445303

ABSTRACT

Macromolecular associates, such as membraneless organelles or lipid-protein assemblies, provide a hydrophobic environment, i.e., a liquid protein phase (LP), where folding preferences can be drastically altered. LP as well as the associated phase change from water (W) is an intriguing phenomenon related to numerous biological processes and also possesses potential in nanotechnological applications. However, the energetic effects of a hydrophobic yet water-containing environment on protein folding are poorly understood. Here, we focus on small ß-sheets, the key motifs of proteins, undergoing structural changes in liquid-liquid phase separation (LLPS) and also model the mechanism of energy-coupled unfolding, e.g., in proteases, during W → LP transition. Due to the importance of the accurate description for hydrogen bonding patterns, the employed models were studied by using quantum mechanical calculations. The results demonstrate that unfolding is energetically less favored in LP by ~0.3-0.5 kcal·mol-1 per residue in which the difference further increased by the presence of explicit structural water molecules, where the folded state was preferred by ~1.2-2.3 kcal·mol-1 per residue relative to that in W. Energetics at the LP/W interfaces was also addressed by theoretical isodesmic reactions. While the models predict folded state preference in LP, the unfolding from LP to W renders the process highly favorable since the unfolded end state has >1 kcal·mol-1 per residue excess stabilization.


Subject(s)
Phase Transition/drug effects , Protein Conformation, beta-Strand/drug effects , Water/pharmacology , Amino Acid Motifs/drug effects , Chemical Fractionation/methods , Computer Simulation , Hydrophobic and Hydrophilic Interactions/drug effects , Kinetics , Macromolecular Substances/chemistry , Models, Molecular , Protein Conformation/drug effects , Protein Folding/drug effects , Protein Stability/drug effects , Quantum Theory , Viscosity , Water/chemistry
18.
Int J Mol Sci ; 22(12)2021 Jun 18.
Article in English | MEDLINE | ID: mdl-34207146

ABSTRACT

Human phenylalanine hydroxylase (PAH) is a metabolic enzyme involved in the catabolism of L-Phe in liver. Loss of conformational stability and decreased enzymatic activity in PAH variants result in the autosomal recessive disorder phenylketonuria (PKU), characterized by developmental and psychological problems if not treated early. One current therapeutic approach to treat PKU is based on pharmacological chaperones (PCs), small molecules that can displace the folding equilibrium of unstable PAH variants toward the native state, thereby rescuing the physiological function of the enzyme. Understanding the PAH folding equilibrium is essential to develop new PCs for different forms of the disease. We investigate here the urea and the thermal-induced denaturation of full-length PAH and of a truncated form lacking the regulatory and the tetramerization domains. For either protein construction, two distinct transitions are seen in chemical denaturation followed by fluorescence emission, indicating the accumulation of equilibrium unfolding intermediates where the catalytic domains are partly unfolded and dissociated from each other. According to analytical centrifugation, the chemical denaturation intermediates of either construction are not well-defined species but highly polydisperse ensembles of protein aggregates. On the other hand, each protein construction similarly shows two transitions in thermal denaturation measured by fluorescence or differential scanning calorimetry, also indicating the accumulation of equilibrium unfolding intermediates. The similar temperatures of mid denaturation of the two constructions, together with their apparent lack of response to protein concentration, indicate the catalytic domains are unfolded in the full-length PAH thermal intermediate, where they remain associated. That the catalytic domain unfolds in the first thermal transition is relevant for the choice of PCs identified in high throughput screening of chemical libraries using differential scanning fluorimetry.


Subject(s)
Phenylalanine Hydroxylase/chemistry , Protein Denaturation , Protein Folding , Binding Sites , Calorimetry, Differential Scanning , Catalytic Domain , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Phenylalanine Hydroxylase/isolation & purification , Phenylketonurias , Protein Conformation , Protein Denaturation/drug effects , Protein Folding/drug effects , Protein Stability , Temperature , Thermodynamics , Urea/chemistry
19.
Bioorg Med Chem Lett ; 48: 128243, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34246753

ABSTRACT

A growing number of diseases are linked to the misfolding of integral membrane proteins, and many of these proteins are targeted for ubiquitin-proteasome-dependent degradation. One such substrate is a mutant form of the Cystic Fibrosis Transmembrane Conductance Regulator (F508del-CFTR). Protein folding "correctors" that repair the F508del-CFTR folding defect have entered the clinic, but they are unlikely to protect the entire protein from degradation. To increase the pool of F508del-CFTR protein that is available for correction by existing treatments, we determined a structure-activity relationship to improve the efficacy and reduce the toxicity of an inhibitor of the E1 ubiquitin activating enzyme that facilitates F508del-CFTR maturation. A resulting lead compound lacked measurable toxicity and improved the ability of an FDA-approved corrector to augment F508del-CFTR folding, transport the protein to the plasma membrane, and maintain its activity. These data support a proof-of-concept that modest inhibition of substrate ubiquitination improves the activity of small molecule correctors to treat CF and potentially other protein conformational disorders.


Subject(s)
Benzoates/pharmacology , Cystic Fibrosis Transmembrane Conductance Regulator/antagonists & inhibitors , Furans/pharmacology , Pyrazoles/pharmacology , Ubiquitin/antagonists & inhibitors , Benzoates/chemistry , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Dose-Response Relationship, Drug , Furans/chemistry , Humans , Molecular Structure , Protein Folding/drug effects , Pyrazoles/chemistry , Structure-Activity Relationship , Ubiquitin/metabolism , Ubiquitination/drug effects
20.
Int J Med Sci ; 18(13): 2890-2896, 2021.
Article in English | MEDLINE | ID: mdl-34220315

ABSTRACT

Background: When an imbalance occurs between the demand and capacity for protein folding, unfolded proteins accumulate in the endoplasmic reticulum (ER) lumen and activate the unfolded protein response (UPR). In addition, unfolded proteins are cleared from the ER lumen for ubiquitination and subsequent cytosolic proteasomal degradation, which is termed as the ER-associated degradation (ERAD) pathway. This study focused on changes in the UPR and ERAD pathways induced by the repeated inhalation anesthetic exposure in Caenorhabditis elegans. Methods: Depending on repeated isoflurane exposure, C. elegans was classified into the control or isoflurane group. To evaluate the expression of a specific gene, RNA was extracted from adult worms in each group and real-time polymerase chain reaction was performed. Ubiquitinated protein levels were measured using western blotting, and behavioral changes were evaluated by chemotaxis assay using various mutant strains. Results: Isoflurane upregulated the expression of ire-1 and pek-1 whereas the expression of atf-6 was unaffected. The expression of both sel-1 and sel-11 was decreased by isoflurane exposure, possibly indicating the inhibition of retro-translocation. The expression of cdc-48.1 and cdc-48.2 was decreased and higher ubiquitinated protein levels were observed in the isoflurane group than in the control, suggesting that deubiquitination and degradation of misfolded proteins were interrupted. The chemotaxis indices of ire-1, pek-1, sel-1, and sel-11 mutants decreased significantly compared to N2, and they were not suppressed further even after the repeated isoflurane exposure. Conclusion: Repeated isoflurane exposure caused significant ER stress in C. elegans. Following the increase in UPR, the ERAD pathway was disrupted by repeated isoflurane exposure and ubiquitinated proteins was accumulated subsequently. UPR and ERAD pathways are potential modifiable neuroprotection targets against anesthesia-induced neurotoxicity.


Subject(s)
Anesthesia, General/adverse effects , Anesthetics, Inhalation/adverse effects , Endoplasmic Reticulum-Associated Degradation/drug effects , Isoflurane/adverse effects , Postoperative Complications/chemically induced , Anesthesia, General/methods , Animals , Caenorhabditis elegans , Caenorhabditis elegans Proteins/metabolism , Chemotaxis/drug effects , Chemotaxis/genetics , Disease Models, Animal , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/pathology , Endoplasmic Reticulum Stress/drug effects , Gene Expression Regulation/drug effects , Humans , Nervous System/drug effects , Postoperative Complications/prevention & control , Protein Folding/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...