Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 116
Filter
1.
J Biol Chem ; 299(8): 105040, 2023 08.
Article in English | MEDLINE | ID: mdl-37442237

ABSTRACT

Cu/Zn-superoxide dismutase (CuZnSOD) is an enzyme that binds a copper and zinc ion and also forms an intramolecular disulfide bond. Together with the copper ion as the active site, the disulfide bond is completely conserved among these proteins; indeed, the disulfide bond plays critical roles in maintaining the catalytically competent conformation of CuZnSOD. Here, we found that a CuZnSOD protein in Paenibacillus lautus (PaSOD) has no Cys residue but exhibits a significant level of enzyme activity. The crystal structure of PaSOD revealed hydrophobic and hydrogen-bonding interactions in substitution for the disulfide bond of the other CuZnSOD proteins. Also notably, we determined that PaSOD forms a homodimer through an additional domain with a novel fold at the N terminus. While the advantages of lacking Cys residues and adopting a novel dimer configuration remain obscure, PaSOD does not require a disulfide-introducing/correcting system for maturation and could also avoid misfolding caused by aberrant thiol oxidations under an oxidative environment.


Subject(s)
Bacterial Proteins , Disulfides , Superoxide Dismutase-1 , Copper , Cysteine , Disulfides/chemistry , Superoxide Dismutase-1/chemistry , Zinc , Bacterial Proteins/chemistry , Paenibacillus , Protein Folding
2.
J Biol Chem ; 299(6): 104798, 2023 06.
Article in English | MEDLINE | ID: mdl-37156398

ABSTRACT

Canine degenerative myelopathy (DM), a fatal neurodegenerative disease in dogs, shares clinical and genetic features with amyotrophic lateral sclerosis, a human motor neuron disease. Mutations in the SOD1 gene encoding Cu/Zn superoxide dismutase (SOD1) cause canine DM and a subset of inherited human amyotrophic lateral sclerosis. The most frequent DM causative mutation is homozygous E40K mutation, which induces the aggregation of canine SOD1 but not of human SOD1. However, the mechanism through which canine E40K mutation induces species-specific aggregation of SOD1 remains unknown. By screening human/canine chimeric SOD1s, we identified that the humanized mutation of the 117th residue (M117L), encoded by exon 4, significantly reduced aggregation propensity of canine SOD1E40K. Conversely, introducing a mutation of leucine 117 to methionine, a residue homologous to canine, promoted E40K-dependent aggregation in human SOD1. M117L mutation improved protein stability and reduced cytotoxicity of canine SOD1E40K. Furthermore, crystal structural analysis of canine SOD1 proteins revealed that M117L increased the packing within the hydrophobic core of the ß-barrel structure, contributing to the increased protein stability. Our findings indicate that the structural vulnerability derived intrinsically from Met 117 in the hydrophobic core of the ß-barrel structure induces E40K-dependent species-specific aggregation in canine SOD1.


Subject(s)
Dog Diseases , Mutation , Neurodegenerative Diseases , Superoxide Dismutase-1 , Animals , Dogs , Humans , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/veterinary , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism , Dog Diseases/genetics , Dog Diseases/metabolism , Species Specificity
3.
Analyst ; 147(20): 4442-4449, 2022 Oct 10.
Article in English | MEDLINE | ID: mdl-36129310

ABSTRACT

Monitoring drug concentration in blood and reflecting this in the dosage are crucial for safe and effective drug treatment. Most drug assays are based on total concentrations of bound and unbound proteins in the serum, although only the unbound concentration causes beneficial and adverse events. Monitoring the unbound concentration alone is expected to provide a means for further optimisation of drug treatment. However, unbound concentration monitoring has not been routinely used for drug treatment due to the long analysis time and the high cost of conventional methods. Here, we have developed a rapid electrochemical method to determine the unbound concentration in ultrafiltered human serum using boron-doped diamond (BDD) electrodes. When the anticancer drug doxorubicin was used as the test drug, the catalytic doxorubicin-mediated reduction of dissolved oxygen provided a sensitive electrochemical signal, with a detection limit of 0.14 nM. In contrast, the sensitivity of glassy carbon (GC) was inferior under the same conditions due to interference from the dissolved oxygen reduction current. The signal background ratio (S/B) of BDD and GC was 11.5 (10 nM doxorubicin) and 1.1 (50 nM), respectively. The results show that a fast measurement time within ten seconds is possible in the clinical concentration range. Additionally, in the ultrafiltered human serum, the obtained values of unbound doxorubicin concentration showed good agreement with those quantified by conventional liquid chromatography-mass spectrometry. This approach has the potential for application in clinical settings where rapid and simple analysis methods would be beneficial.


Subject(s)
Boron , Carbon , Boron/chemistry , Doxorubicin , Electrodes , Humans , Oxidation-Reduction , Oxygen
4.
J Clin Biochem Nutr ; 71(2): 73-77, 2022 Sep.
Article in English | MEDLINE | ID: mdl-36213785

ABSTRACT

Mutations in the gene coding Cu/Zn-superoxide dismutase (SOD1) are linked to a familial form of amyotrophic lateral sclerosis (ALS), and its pathological hallmark includes abnormal accumulation of mutant SOD1 proteins in spinal motorneurons. Mutant SOD1 proteins are considered to be susceptible to misfolding, resulting in the accumulation as oligomers/aggregates. While it remains obscure how and why SOD1 becomes misfolded under pathological conditions in vivo, the failure to bind a copper and zinc ion in SOD1 in vitro leads to the significant destabilization of its natively folded structure. Therefore, genetic and pharmacological attempts to promote the metal binding in mutant SOD1 could serve as an effective treatment of ALS. Here, I briefly review the copper and zinc binding process of SOD1 in vivo and discuss a copper chaperone for SOD1 as a potential target for developing ALS therapeutics.

5.
Biochem Biophys Res Commun ; 508(3): 729-734, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30528390

ABSTRACT

Alpha-synuclein (a-syn) aggregation in brain is implicated in several synucleinopathies, including Parkinson's disease (PD), dementia with Lewy bodies (DLB), and multiple system atrophy (MSA). Until date, at least six disease-associated mutations in a-syn (namely A30P, E46K, H50Q, G51D, A53T, and A53E) are known to cause dominantly inherited familial forms of synucleinopathies. Previous studies using recombinant proteins have reported that a subset of disease-associated mutants show higher aggregation propensities and form spectroscopically distinguishable aggregates compared to wild-type (WT). However, morphological and biochemical comparison of the aggregates for all disease-associated a-syn mutants have not yet been performed. In this study, we performed electron microscopic examination, guanidinium hydrochloride (GdnHCl) denaturation, and protease digestion to classify the aggregates from their respective point mutations. Using electron microscopy we observed variations of amyloid fibrillar morphologies among the aggregates of a-syn mutants, mainly categorized into two groups: twisted fibrils observed for both WT and E46K while straight fibrils for the other mutants. GdnHCl denaturation experiments revealed the a-syn mutants except for E46K were more resistant than WT against the denaturation. Mass spectrometry analysis of protease-treated aggregates showed a variety of protease-resistant cores, which may correspond to their morphological properties. The difference of their properties could be implicated in the clinicopathological difference of synucleinopathies with those mutations.


Subject(s)
Mutant Proteins/metabolism , Parkinson Disease/metabolism , Protein Aggregates , alpha-Synuclein/metabolism , Animals , Endopeptidase K/metabolism , Humans , Mice , Mutant Proteins/chemistry , Mutant Proteins/ultrastructure , Mutation/genetics , alpha-Synuclein/chemistry , alpha-Synuclein/ultrastructure
6.
Phys Rev Lett ; 122(15): 157401, 2019 Apr 19.
Article in English | MEDLINE | ID: mdl-31050541

ABSTRACT

This study aims to reveal the full potential of ZnO as an ultrafast photofunctional material. Based on nonlocal response theory to incorporate the spatially inhomogeneous quality of the samples coupled with experimental observations of linear and nonlinear optical responses, we establish the ultrafast radiative decay of excitons in ZnO thin films that reaches the speed of excitonic dephasing at room temperature in typical semiconductors at a couple tens of femtoseconds. The consistency between the observed delay-time dependence of the transient-grating signals and the theoretical prediction reveals that the ultrafast radiative decay is due to the synergetic effects of the giant light-exciton interaction volume and the radiative coupling between multicomponent excitons.

7.
Biochim Biophys Acta Mol Basis Dis ; 1864(6 Pt A): 2119-2130, 2018 06.
Article in English | MEDLINE | ID: mdl-29551730

ABSTRACT

Dominant mutations in the gene encoding copper and zinc-binding superoxide dismutase (SOD1) cause amyotrophic lateral sclerosis (ALS). Abnormal accumulation of misfolded SOD1 proteins in spinal motoneurons is a major pathological hallmark in SOD1-related ALS. Dissociation of copper and/or zinc ions from SOD1 has been shown to trigger the protein aggregation/oligomerization in vitro, but the pathological contribution of such metal dissociation to the SOD1 misfolding still remains obscure. Here, we tested the relevance of the metal-deficient SOD1 in the misfolding in vivo by developing a novel antibody (anti-apoSOD), which exclusively recognized mutant SOD1 deficient in metal ions at its copper-binding site. Notably, anti-apoSOD-reactive species were detected specifically in the spinal cords of the ALS model mice only at their early pre-symptomatic stages but not at the end stage of the disease. The cerebrospinal fluid as well as the spinal cord homogenate of one SOD1-ALS patient also contained the anti-apoSOD-reactive species. Our results thus suggest that metal-deficiency in mutant SOD1 at its copper-binding site is one of the earliest pathological features in SOD1-ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/diagnosis , Copper/metabolism , Protein Aggregation, Pathological/diagnosis , Superoxide Dismutase-1/metabolism , Adult , Aged , Aged, 80 and over , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/immunology , Amyotrophic Lateral Sclerosis/pathology , Animals , Antibodies/immunology , Asymptomatic Diseases , Binding Sites/genetics , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Transgenic , Middle Aged , Motor Neurons/pathology , Mutation , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/immunology , Protein Aggregation, Pathological/pathology , Protein Binding/genetics , Protein Folding , Sensitivity and Specificity , Spinal Cord/cytology , Spinal Cord/pathology , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/immunology , Zinc/metabolism
8.
Brain ; 140(9): 2460-2474, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-29050386

ABSTRACT

See Jellinger (doi:10.1093/awx190) for a scientific commentary on this article. The enzyme monoamine oxidases (B and A subtypes, encoded by MAOB and MAOA, respectively) are drug targets in the treatment of Parkinson's disease. Inhibitors of MAOB are used clinically in Parkinson's disease for symptomatic purposes whereas the potential disease-modifying effect of monoamine oxidase inhibitors is debated. As astroglial cells express high levels of MAOB, the enzyme has been proposed as a brain imaging marker of astrogliosis, a cellular process possibly involved in Parkinson's disease pathogenesis as elevation of MAOB in astrocytes might be harmful. Since brain monoamine oxidase status in Parkinson's disease is uncertain, our objective was to measure, by quantitative immunoblotting in autopsied brain homogenates, protein levels of both monoamine oxidases in three different degenerative parkinsonian disorders: Parkinson's disease (n = 11), multiple system atrophy (n = 11), and progressive supranuclear palsy (n = 16) and in matched controls (n = 16). We hypothesized that if MAOB is 'substantially' localized to astroglial cells, MAOB levels should be generally associated with standard astroglial protein measures (e.g. glial fibrillary acidic protein). MAOB levels were increased in degenerating putamen (+83%) and substantia nigra (+10%, non-significant) in multiple system atrophy; in caudate (+26%), putamen (+27%), frontal cortex (+31%) and substantia nigra (+23%) of progressive supranuclear palsy; and in frontal cortex (+33%), but not in substantia nigra of Parkinson's disease, a region we previously reported no increase in astrocyte protein markers. Although the magnitude of MAOB increase was less than those of standard astrocytic markers, significant positive correlations were observed amongst the astrocyte proteins and MAOB. Despite suggestions that MAOA (versus MAOB) is primarily responsible for metabolism of dopamine in dopamine neurons, there was no loss of the enzyme in the parkinsonian substantia nigra; instead, increased nigral levels of a MAOA fragment and 'turnover' of the enzyme were observed in the conditions. Our findings provide support that MAOB might serve as a biochemical imaging marker, albeit not entirely specific, for astrocyte activation in human brain. The observation that MAOB protein concentration is generally increased in degenerating brain areas in multiple system atrophy (especially putamen) and in progressive supranuclear palsy, but not in the nigra in Parkinson's disease, also distinguishes astrocyte behaviour in Parkinson's disease from that in the two 'Parkinson-plus' conditions. The question remains whether suppression of either MAOB in astrocytes or MAOA in dopamine neurons might influence progression of the parkinsonian disorders.


Subject(s)
Brain/enzymology , Dopamine/deficiency , Monoamine Oxidase/metabolism , Multiple System Atrophy/metabolism , Parkinson Disease/metabolism , Supranuclear Palsy, Progressive/metabolism , Adolescent , Adult , Case-Control Studies , Caudate Nucleus/metabolism , Female , Frontal Lobe/metabolism , Glial Fibrillary Acidic Protein/metabolism , Humans , Isoenzymes/metabolism , Male , Middle Aged , Multiple System Atrophy/pathology , Nerve Degeneration/pathology , Parkinson Disease/pathology , Peptide Fragments/metabolism , Phosphopyruvate Hydratase/metabolism , Putamen/metabolism , Substantia Nigra/metabolism , Supranuclear Palsy, Progressive/pathology , Tubulin/metabolism , Young Adult , alpha-Synuclein/metabolism
9.
J Biol Chem ; 291(8): 4144-55, 2016 Feb 19.
Article in English | MEDLINE | ID: mdl-26694608

ABSTRACT

Misfolding of Cu,Zn-superoxide dismutase (SOD1) is a pathological change in the familial form of amyotrophic lateral sclerosis caused by mutations in the SOD1 gene. SOD1 is an enzyme that matures through the binding of copper and zinc ions and the formation of an intramolecular disulfide bond. Pathogenic mutations are proposed to retard the post-translational maturation, decrease the structural stability, and hence trigger the misfolding of SOD1 proteins. Despite this, a misfolded and potentially pathogenic conformation of immature SOD1 remains obscure. Here, we show significant and distinct conformational changes of apoSOD1 that occur only upon reduction of the intramolecular disulfide bond in solution. In particular, loop regions in SOD1 lose their restraint and become significantly disordered upon dissociation of metal ions and reduction of the disulfide bond. Such drastic changes in the solution structure of SOD1 may trigger misfolding and fibrillar aggregation observed as pathological changes in the familial form of amyotrophic lateral sclerosis.


Subject(s)
Amyotrophic Lateral Sclerosis , Copper/chemistry , Protein Aggregation, Pathological , Superoxide Dismutase/chemistry , Zinc/chemistry , Copper/metabolism , Disulfides/chemistry , Disulfides/metabolism , Humans , Protein Structure, Secondary , Protein Structure, Tertiary , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , Zinc/metabolism
10.
Adv Exp Med Biol ; 925: 1-12, 2017.
Article in English | MEDLINE | ID: mdl-27311318

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal motor neuron disease that is characterized by the formation of abnormal inclusions in neurons. While the pathomechanism of ALS remains obscure, a number of proteins have been identified in the inclusion bodies, and the pathological roles of RNA-binding proteins have been increasingly emphasized. Among those, the FET proteins (FUS, EWSR1, TAF15) were recently identified as RNA-binding proteins in pathological inclusions of ALS and other neurodegenerative diseases; moreover, mutations in the genes encoding the FET proteins were found to be associated with familial forms of ALS. FET proteins are normally localized in the nucleus, but the introduction of pathogenic mutations in FET proteins leads to their abnormal redistribution to the cytoplasm, where they form aggregates. While further investigation will be required to understand the intracellular factors controlling the aggregation propensities of FET proteins, they are thought to lose their physiological functions and become toxic through their misfolding/aggregation. Here, we will briefly review recent advances of our understanding of the physiological functions and aggregation behavior of FET proteins in vivo as well as in vitro.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Calmodulin-Binding Proteins/genetics , Protein Aggregation, Pathological/genetics , RNA-Binding Protein FUS/genetics , RNA-Binding Proteins/genetics , TATA-Binding Protein Associated Factors/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Calmodulin-Binding Proteins/metabolism , Cell Nucleus/metabolism , Cytosol/metabolism , Gene Expression Regulation , Humans , Inclusion Bodies/chemistry , Inclusion Bodies/pathology , Mutation , Neurons/metabolism , Neurons/pathology , Protein Aggregates/genetics , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology , RNA-Binding Protein EWS , RNA-Binding Protein FUS/metabolism , RNA-Binding Proteins/metabolism , Signal Transduction , TATA-Binding Protein Associated Factors/metabolism
11.
Int J Mol Sci ; 17(5)2016 Apr 28.
Article in English | MEDLINE | ID: mdl-27136532

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a lethal neurodegenerative disease affecting both upper and lower motor neurons, and currently, there is no cure or effective treatment. Mutations in a gene encoding a ubiquitous antioxidant enzyme, Cu,Zn-superoxide dismutase (SOD1), have been first identified as a cause of familial forms of ALS. It is widely accepted that mutant SOD1 proteins cause the disease through a gain in toxicity but not through a loss of its physiological function. SOD1 is a major copper-binding protein and regulates copper homeostasis in the cell; therefore, a toxicity of mutant SOD1 could arise from the disruption of copper homeostasis. In this review, we will briefly review recent studies implying roles of copper homeostasis in the pathogenesis of SOD1-ALS and highlight the therapeutic interventions focusing on pharmacological as well as genetic regulations of copper homeostasis to modify the pathological process in SOD1-ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Copper/metabolism , Superoxide Dismutase-1/genetics , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/pathology , Animals , Chelating Agents/chemistry , Chelating Agents/metabolism , Chelating Agents/therapeutic use , Copper/chemistry , Disease Models, Animal , Humans , Polymorphism, Single Nucleotide
12.
J Biol Chem ; 289(29): 20139-49, 2014 Jul 18.
Article in English | MEDLINE | ID: mdl-24917671

ABSTRACT

Enzymatic activation of Cu,Zn-superoxide dismutase (SOD1) requires not only binding of a catalytic copper ion but also formation of an intramolecular disulfide bond. Indeed, the disulfide bond is completely conserved among all species possessing SOD1; however, it remains obscure how disulfide formation controls the enzymatic activity of SOD1. Here, we show that disulfide formation is a primary event in the folding process of prokaryotic SOD1 (SodC) localized to the periplasmic space. Escherichia coli SodC was found to attain ß-sheet structure upon formation of the disulfide bond, whereas disulfide-reduced SodC assumed little secondary structure even in the presence of copper and zinc ions. Moreover, reduction of the disulfide bond made SodC highly susceptible to proteolytic degradation. We thus propose that the thiol-disulfide status in SodC controls the intracellular stability of this antioxidant enzyme and that the oxidizing environment of the periplasm is required for the enzymatic activation of SodC.


Subject(s)
Escherichia coli Proteins/chemistry , Superoxide Dismutase/chemistry , Binding Sites , Copper/metabolism , Disulfides/chemistry , Enzyme Activation , Enzyme Stability , Escherichia coli/enzymology , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Models, Molecular , Protein Conformation , Protein Folding , Protein Structure, Secondary , Proteolysis , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Zinc/metabolism
13.
J Biol Chem ; 289(2): 1192-202, 2014 Jan 10.
Article in English | MEDLINE | ID: mdl-24280224

ABSTRACT

Dominant mutations in FUS/TLS cause a familial form of amyotrophic lateral sclerosis (fALS), where abnormal accumulation of mutant FUS proteins in cytoplasm has been observed as a major pathological change. Many of pathogenic mutations have been shown to deteriorate the nuclear localization signal in FUS and thereby facilitate cytoplasmic mislocalization of mutant proteins. Several other mutations, however, exhibit no effects on the nuclear localization of FUS in cultured cells, and their roles in the pathomechanism of fALS remain obscure. Here, we show that a pathogenic mutation, G156E, significantly increases the propensities for aggregation of FUS in vitro and in vivo. Spontaneous in vitro formation of amyloid-like fibrillar aggregates was observed in mutant but not wild-type FUS, and notably, those fibrils functioned as efficient seeds to trigger the aggregation of wild-type protein. In addition, the G156E mutation did not disturb the nuclear localization of FUS but facilitated the formation of intranuclear inclusions in rat hippocampal neurons with significant cytotoxicity. We thus propose that intranuclear aggregation of FUS triggered by a subset of pathogenic mutations is an alternative pathomechanism of FUS-related fALS diseases.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Cell Nucleus/metabolism , Mutation , RNA-Binding Protein FUS/metabolism , Amino Acid Sequence , Amino Acid Substitution , Amyloid/chemistry , Amyloid/metabolism , Amyloid/ultrastructure , Amyotrophic Lateral Sclerosis/genetics , Animals , Cell Line, Tumor , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Humans , Microscopy, Confocal , Microscopy, Electron , Neurons/cytology , Neurons/metabolism , Protein Denaturation , RNA-Binding Protein FUS/chemistry , RNA-Binding Protein FUS/genetics , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
14.
Neurobiol Dis ; 82: 243-253, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26102022

ABSTRACT

Although gliosis is a normal response to brain injury, reports on the extent of astrogliosis in the degenerating substantia nigra in Parkinson's disease (PD) are conflicting. It has also been recently suggested that accumulation of nigral α-synuclein in this disorder might suppress astrocyte activation which in turn could exacerbate the degenerative process. This study examined brain protein levels (intact protein, fragments, and aggregates, if any) of astroglial markers and their relationship to α-synuclein in PD and in the positive control parkinson-plus conditions multiple system atrophy (MSA) and progressive supranuclear palsy (PSP). Autopsied brain homogenates of patients with PD (n=10), MSA (n=11), PSP (n=11) and matched controls (n=10) were examined for the astroglial markers glial fibrillary acidic protein (GFAP), vimentin, and heat shock protein-27 (Hsp27) by quantitative immunoblotting. As expected, both MSA (putamen>substantia nigra>caudate>frontal cortex) and PSP (substantia nigra>caudate>putamen, frontal cortex) showed widespread but regionally specific pattern of increased immunoreactivity of the markers, in particular for the partially proteolyzed fragments (all three) and aggregates (GFAP). In contrast, immunoreactivity of the three markers was largely normal in PD in brain regions examined with the exception of trends for variably increased levels of cleaved vimentin in substantia nigra and frontal cortex. In patients with PD, GFAP levels in the substantia nigra correlated inversely with α-synuclein accumulation whereas the opposite was true for MSA. Our biochemical findings of generally normal protein levels of astroglial markers in substantia nigra of PD, and negative correlation with α-synuclein concentration, are consistent with some recent neuropathology reports of mild astroglial response and with the speculation that astrogliosis might be suppressed in this disorder by excessive α-synuclein accumulation. Should astrogliosis protect, to some extent, the degenerating substantia nigra from damage, therapeutics aimed at normalization of astrocyte reaction in PD could be helpful.


Subject(s)
Astrocytes/metabolism , Caudate Nucleus/metabolism , Frontal Lobe/metabolism , Parkinson Disease/metabolism , Putamen/metabolism , Aged , Biomarkers/metabolism , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Glial Fibrillary Acidic Protein/metabolism , HSP27 Heat-Shock Proteins/metabolism , Heat-Shock Proteins , Humans , Molecular Chaperones , Multiple System Atrophy/metabolism , Supranuclear Palsy, Progressive/metabolism , Vimentin/metabolism
15.
Biochem Biophys Res Commun ; 463(4): 1196-202, 2015 Aug 07.
Article in English | MEDLINE | ID: mdl-26086102

ABSTRACT

Dominant mutations in Cu,Zn-superoxide dismutase (SOD1) cause a familial form of amyotrophic lateral sclerosis (ALS). A pathological hallmark of the familial ALS is the formation of mutant SOD1 aggregates, leading to the proposal that SOD1 gains toxicities through protein misfolding triggered by mutations. Nevertheless, molecular requirements for mutant SOD1 to acquire pathogenicity still remain obscure. Here, we show that Cys residues in SOD1 are essential to exerting toxicities of SOD1 in a Caenorhabditis elegans model. Exogenous expression of wild-type as well as pathogenic mutant SOD1 fused with a fluorescent protein in C. elegans resulted in the accumulation of disulfide-reduced SOD1 and retarded the worm's motility. In contrast, little effects of exogenously expressed SOD1 on the motility were observed when all four Cys residues in SOD1 were replaced with Ser. Taken together, we propose that deregulation of Cys chemistry in SOD1 proteins is involved in the pathogenesis of SOD1-related ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/enzymology , Caenorhabditis elegans/drug effects , Cysteine/metabolism , Superoxide Dismutase/metabolism , Animals , Disease Models, Animal , Superoxide Dismutase/chemistry
16.
Mov Disord ; 30(2): 160-6, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25641350

ABSTRACT

Dopamine agonist medications with high affinity for the D3 dopamine receptor are commonly used to treat Parkinson's disease, and have been associated with pathological behaviors categorized under the umbrella of impulse control disorders (ICD). The aim of this study was to investigate whether ICD in Parkinson's patients are associated with greater D3 dopamine receptor availability. We used positron emission tomography (PET) radioligand imaging with the D3 dopamine receptor preferring agonist [¹¹C]-(+)-propyl-hexahydro-naphtho-oxazin (PHNO) in Parkinson's patients with (n = 11) and without (n = 21) ICD, and age-, sex-, and education-matched healthy control subjects (n = 18). Contrary to hypotheses, [¹¹C]-(+)-PHNO binding in D3 -rich brain areas was not elevated in Parkinson's patients with ICD compared with those without; instead, [¹¹C]-(+)-PHNO binding in ventral striatum was 20% lower (P = 0.011), correlating with two measures of ICD severity (r = -0.8 and -0.9), which may reflect higher dopamine tone in ventral striatum. In dorsal striatum, where [¹¹C]-(+)-PHNO binding is associated with D2 receptor levels, [¹¹C]-(+)-PHNO binding was elevated across patients compared with controls. We conclude that although D3 dopamine receptors have been linked to the occurrence of ICD in Parkinson's patients. Our findings do not support the hypothesis that D3 receptor levels are elevated in Parkinson's patients with ICD. We also did not find ICD-related abnormalities in D2 receptor levels. Our findings argue against the possibility that differences in D2/3 receptor levels can account for the development of ICD in PD; however, we cannot rule out that differences in dopamine levels (particularly in ventral striatum) may be involved.


Subject(s)
Disruptive, Impulse Control, and Conduct Disorders/diagnostic imaging , Parkinson Disease/diagnostic imaging , Positron-Emission Tomography , Receptors, Dopamine D2/metabolism , Receptors, Dopamine D3/metabolism , Adult , Aged , Aged, 80 and over , Disruptive, Impulse Control, and Conduct Disorders/etiology , Disruptive, Impulse Control, and Conduct Disorders/metabolism , Dopamine/metabolism , Dopamine Agents , Female , Humans , Male , Middle Aged , Oxazines , Parkinson Disease/complications , Parkinson Disease/metabolism , Positron-Emission Tomography/methods
17.
J Biol Chem ; 288(7): 4970-80, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23264618

ABSTRACT

Dominant mutations in Cu,Zn-superoxide dismutase (SOD1) are a cause of a familial form of amyotrophic lateral sclerosis. Wild-type SOD1 forms a highly conserved intra-molecular disulfide bond, whereas pathological SOD1 proteins are cross-linked via intermolecular disulfide bonds and form insoluble oligomers. A thiol-disulfide status in SOD1 will thus play a regulatory role in determining its folding/misfolding pathways; however, it remains unknown how pathogenic mutations in SOD1 affect the thiol-disulfide status to facilitate the protein misfolding. Here, we show that the structural destabilization of SOD1 scrambles a disulfide bond among four Cys residues in an SOD1 molecule. The disulfide scrambling produces SOD1 monomers with distinct electrophoretic mobility and also reproduces the formation of disulfide-linked oligomers. We have also found that the familial form of amyotrophic lateral sclerosis-causing mutations facilitate the disulfide scrambling in SOD1. Based upon our results, therefore, scrambling of the conserved disulfide bond will be a key event to cause the pathological changes in disease-associated mutant SOD1 proteins.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Disulfides/chemistry , Gene Expression Regulation , Mutation , Superoxide Dismutase/metabolism , Cross-Linking Reagents/pharmacology , Cysteine/chemistry , Dose-Response Relationship, Drug , Humans , Kinetics , Mass Spectrometry/methods , Models, Biological , Protein Denaturation , Protein Folding , Sulfhydryl Compounds/chemistry , Temperature , Time Factors
18.
Biochim Biophys Acta ; 1832(8): 1271-8, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23597596

ABSTRACT

Many proteins exhibit propensities to form fibrillar aggregates called amyloids that are rich in ß-sheet structures. Abnormal accumulation of amyloids in the brain and spinal cords is well known as a major pathological change in neurodegenerative diseases; therefore, amyloids have long been considered as disease culprits formed via protein misfolding and should be avoided in healthy cells. Recently, however, increasing numbers of proteins have been identified that require formation of fibrillar states for exertion of their physiological functions, and the critical roles of such functional amyloids include a molecular switch for environmental adaptation, a structural template for catalysis, and a regulator of intracellular signaling. Protein amyloids will, therefore, be more prevailed in our physiologies than we have expected so far. Here, we have reviewed recent studies on such regulatory roles of protein fibrillar aggregates in various physiologies and further discussed possible relations of functional to pathological amyloids.


Subject(s)
Amyloid/metabolism , Amyloidosis/genetics , Amyloidosis/metabolism , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , RNA/genetics , Amyloid/genetics , Animals , Humans , Protein Folding , RNA/metabolism
19.
Neurobiol Dis ; 67: 107-18, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24704312

ABSTRACT

Animal data show that high doses of the stimulant drug methamphetamine can damage brain dopamine neurones; however, it is still uncertain whether methamphetamine, at any dose, is neurotoxic to human brain. Since gliosis is typically associated with brain damage and is observed in animal models of methamphetamine exposure, we measured protein levels (intact protein and fragments, if any) of markers of microgliosis (glucose transporter-5, human leukocyte antigens HLA-DRα [TAL.1B5] and HLA-DR/DQ/DPß [CR3/43]) and astrogliosis (glial fibrillary acidic protein, vimentin, and heat shock protein-27) in homogenates of autopsied brain of chronic methamphetamine users (n=20) and matched controls (n=23). Intact protein levels of all markers were, as expected, elevated (+28%-1270%, P<0.05) in putamen of patients with the neurodegenerative disorder multiple system atrophy (as a positive control) as were concentrations of fragments of glial fibrillary acidic protein, vimentin and heat shock protein-27 (+170%-4700%, P<0.005). In contrast, intact protein concentrations of the markers were normal in dopamine-rich striatum (caudate, putamen) and in the frontal cortex of the drug users. However, striatal levels of cleaved vimentin and heat shock protein-27 were increased (by 98%-211%, P<0.05), with positive correlations (r=0.41-0.60) observed between concentrations of truncated heat shock protein-27 and extent of dopamine loss (P=0.006) and levels of lipid peroxidation products 4-hydroxynonenal (P=0.046) and malondialdehyde (P=0.11). Our failure to detect increased intact protein levels of commonly used markers of microgliosis and astrogliosis could be explained by exposure to methamphetamine insufficient to cause a toxic process associated with overt gliosis; however, about half of the subjects had died of drug intoxication suggesting that "high" drug doses might have been used. Alternatively, drug tolerance to toxic effects might have occurred in the subjects, who were all chronic methamphetamine users. Nevertheless, the finding of above-normal levels of striatal vimentin and heat shock protein-27 fragments (which constituted 10-28% of the intact protein), for which changes in the latter correlated with those of several markers possibly suggestive of damage, does suggest that some astrocytic "disturbance" had occurred, which might in principle be related to methamphetamine neurotoxicity or to a neuroplastic remodeling process. Taken together, our neurochemical findings do not provide strong evidence for either marked microgliosis or astrogliosis in at least a subgroup of human recreational methamphetamine users who used the drug chronically and shortly before death. However, a logistically more difficult quantitative histopathological study is needed to confirm whether glial changes occur or do not occur in brain of human methamphetamine (and amphetamine) users.


Subject(s)
Brain/drug effects , Central Nervous System Stimulants , Gliosis/chemically induced , Methamphetamine , Adolescent , Adult , Brain/metabolism , Female , Gliosis/metabolism , Humans , Male , Young Adult
20.
Protein Sci ; 33(4): e4961, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38511674

ABSTRACT

Misfolding of mutant Cu/Zn-superoxide dismutase (SOD1) has been implicated in familial form of amyotrophic lateral sclerosis (ALS). A natively folded SOD1 forms a tight homodimer, and the dimer dissociation has been proposed to trigger the oligomerization/aggregation of SOD1. Besides increasing demand for probes allowing the detection of monomerized forms of SOD1 in various applications, the development of probes has been limited to conventional antibodies. Here, we have developed Mb(S4) monobody, a small synthetic binding protein based on the fibronectin type III scaffold, that recognizes a monomeric but not dimeric form of SOD1 by performing combinatorial library selections using phage and yeast-surface display methods. Although Mb(S4) was characterized by its excellent selectivity to the monomeric conformation of SOD1, the monomeric SOD1/Mb(S4) complex was not so stable (apparent Kd ~ µM) as to be detected in conventional pull-down experiments. Instead, the complex of Mb(S4) with monomeric but not dimeric SOD1 was successfully trapped by proximity-enabled chemical crosslinking even when reacted in the cell lysates. We thus anticipate that Mb(S4) binding followed by chemical crosslinking would be a useful strategy for in vitro and also ex vivo detection of the monomeric SOD1 proteins.


Subject(s)
Amyotrophic Lateral Sclerosis , Humans , Superoxide Dismutase-1/chemistry , Amyotrophic Lateral Sclerosis/genetics , Protein Folding , Superoxide Dismutase/chemistry , Saccharomyces cerevisiae/metabolism , Zinc/metabolism , Mutation
SELECTION OF CITATIONS
SEARCH DETAIL