Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters











Publication year range
1.
Brain Commun ; 5(1): fcad017, 2023.
Article in English | MEDLINE | ID: mdl-36793789

ABSTRACT

Superoxide dismutase-1 is a ubiquitously expressed antioxidant enzyme. Mutations in SOD1 can cause amyotrophic lateral sclerosis, probably via a toxic gain-of-function involving protein aggregation and prion-like mechanisms. Recently, homozygosity for loss-of-function mutations in SOD1 has been reported in patients presenting with infantile-onset motor neuron disease. We explored the bodily effects of superoxide dismutase-1 enzymatic deficiency in eight children homozygous for the p.C112Wfs*11 truncating mutation. In addition to physical and imaging examinations, we collected blood, urine and skin fibroblast samples. We used a comprehensive panel of clinically established analyses to assess organ function and analysed oxidative stress markers, antioxidant compounds, and the characteristics of the mutant Superoxide dismutase-1. From around 8 months of age, all patients exhibited progressive signs of both upper and lower motor neuron dysfunction, cerebellar, brain stem, and frontal lobe atrophy and elevated plasma neurofilament concentration indicating ongoing axonal damage. The disease progression seemed to slow down over the following years. The p.C112Wfs*11 gene product is unstable, rapidly degraded and no aggregates were found in fibroblast. Most laboratory tests indicated normal organ integrity and only a few modest deviations were found. The patients displayed anaemia with shortened survival of erythrocytes containing decreased levels of reduced glutathione. A variety of other antioxidants and oxidant damage markers were within normal range. In conclusion, non-neuronal organs in humans show a remarkable tolerance to absence of Superoxide dismutase-1 enzymatic activity. The study highlights the enigmatic specific vulnerability of the motor system to both gain-of-function mutations in SOD1 and loss of the enzyme as in the here depicted infantile superoxide dismutase-1 deficiency syndrome.

2.
J Neurochem ; 164(1): 77-93, 2023 01.
Article in English | MEDLINE | ID: mdl-36326589

ABSTRACT

Mutations in the human Superoxide dismutase 1 (hSOD1) gene are well-established cause of the motor neuron disease ALS. Patients and transgenic (Tg) ALS model mice carrying mutant variants develop hSOD1 aggregates in the CNS. We have identified two hSOD1 aggregate strains, which both transmit spreading template-directed aggregation and premature fatal paralysis when inoculated into adult transgenic mice. This prion-like spread of aggregation could be a primary disease mechanism in SOD1-induced ALS. Human SOD1 aggregation has been studied extensively both in cultured cells and under various conditions in vitro. To determine how the structure of aggregates formed in these model systems related to disease-associated aggregates in the CNS, we used a binary epitope-mapping assay to examine aggregates of hSOD1 variants G93A, G85R, A4V, D90A, and G127X formed in vitro, in four different cell lines and in the CNS of Tg mice. We found considerable variability between replicate sets of in vitro-generated aggregates. In contrast, there was a high similarity between replicates of a given hSOD1 mutant in a given cell line, but pronounced variations between different hSOD1 mutants and different cell lines in both structures and amounts of aggregates formed. The aggregates formed in vitro or in cultured cells did not replicate the aggregate strains that arise in the CNS. Our findings suggest that the distinct aggregate morphologies in the CNS could result from a micro-environment with stringent quality control combined with second-order selection by spreading ability. Explorations of pathogenesis and development of therapeutics should be conducted in models that replicate aggregate structures forming in the CNS.


Subject(s)
Amyotrophic Lateral Sclerosis , Mice , Humans , Animals , Superoxide Dismutase-1/genetics , Amyotrophic Lateral Sclerosis/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Mice, Transgenic , Cells, Cultured , Mutation/genetics , Disease Models, Animal
3.
Acta Neuropathol Commun ; 8(1): 161, 2020 09 14.
Article in English | MEDLINE | ID: mdl-32928301

ABSTRACT

Increasing evidence suggests that propagation of the motor neuron disease amyotrophic lateral sclerosis (ALS) involves the pathogenic aggregation of disease-associated proteins that spread in a prion-like manner. We have identified two aggregate strains of human superoxide dismutase 1 (hSOD1) that arise in the CNS of transgenic mouse models of SOD1-mediated ALS. Both strains transmit template-directed aggregation and premature fatal paralysis when inoculated into the spinal cord of adult hSOD1 transgenic mice. This spread of pathogenic aggregation could be a potential target for immunotherapeutic intervention. Here we generated mouse monoclonal antibodies (mAbs) directed to exposed epitopes in hSOD1 aggregate strains and identified an aggregate selective mAb that targets the aa 143-153 C-terminal extremity of hSOD1 (αSOD1143-153). Both pre-incubation of seeds with αSOD1143-153 prior to inoculation, and weekly intraperitoneal (i.p.) administration attenuated transmission of pathogenic aggregation and prolonged the survival of seed-inoculated hSOD1G85R Tg mice. In contrast, administration of a mAb targeting aa 65-72 (αSOD165-72), which exhibits high affinity towards monomeric disordered hSOD1, had an adverse effect and aggravated seed induced premature ALS-like disease. Although the mAbs reached similar concentrations in CSF, only αSOD1143-153 was found in association with aggregated hSOD1 in spinal cord homogenates. Our results suggest that an aggregate-selective immunotherapeutic approach may suppress seeded transmission of pathogenic aggregation in ALS. However, long-term administration of αSOD1143-153 was unable to prolong the lifespan of non-inoculated hSOD1G85R Tg mice. Thus, spontaneously initiated hSOD1 aggregation in spinal motor neurons may be poorly accessible to therapeutic antibodies.


Subject(s)
Amyotrophic Lateral Sclerosis , Antibodies, Monoclonal/pharmacology , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology , Superoxide Dismutase-1/metabolism , Animals , Disease Models, Animal , Disease Progression , Humans , Mice , Mice, Transgenic
6.
Acta Neuropathol ; 138(1): 85-101, 2019 07.
Article in English | MEDLINE | ID: mdl-30863976

ABSTRACT

Mutations in superoxide dismutase 1 (SOD1) cause amyotrophic lateral sclerosis (ALS). Disease pathogenesis is linked to destabilization, disorder and aggregation of the SOD1 protein. However, the non-genetic factors that promote disorder and the subsequent aggregation of SOD1 have not been studied. Mainly located to the reducing cytosol, mature SOD1 contains an oxidized disulfide bond that is important for its stability. Since O2 is required for formation of the bond, we reasoned that low O2 tension might be a risk factor for the pathological changes associated with ALS development. By combining biochemical approaches in an extensive range of genetically distinct patient-derived cell lines, we show that the disulfide bond is an Achilles heel of the SOD1 protein. Culture of patient-derived fibroblasts, astrocytes, and induced pluripotent stem cell-derived mixed motor neuron and astrocyte cultures (MNACs) under low O2 tensions caused reductive bond cleavage and increases in disordered SOD1. The effects were greatest in cells derived from patients carrying ALS-linked mutations in SOD1. However, significant increases also occurred in wild-type SOD1 in cultures derived from non-disease controls, and patients carrying mutations in other common ALS-linked genes. Compared to fibroblasts, MNACs showed far greater increases in SOD1 disorder and even aggregation of mutant SOD1s, in line with the vulnerability of the motor system to SOD1-mediated neurotoxicity. Our results show for the first time that O2 tension is a principal determinant of SOD1 stability in human patient-derived cells. Furthermore, we provide a mechanism by which non-genetic risk factors for ALS, such as aging and other conditions causing reduced vascular perfusion, could promote disease initiation and progression.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Fibroblasts/pathology , Motor Neurons/pathology , Oxygen/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Fibroblasts/metabolism , Humans , Mutation/genetics , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism
7.
Acta Neuropathol ; 136(6): 939-953, 2018 12.
Article in English | MEDLINE | ID: mdl-30284034

ABSTRACT

Motor neurons containing aggregates of superoxide dismutase 1 (SOD1) are hallmarks of amyotrophic lateral sclerosis (ALS) caused by mutations in the gene encoding SOD1. We have previously reported that two strains of mutant human (h) SOD1 aggregates (denoted A and B) can arise in hSOD1-transgenic models for ALS and that inoculation of such aggregates into the lumbar spinal cord of mice results in rostrally spreading, templated hSOD1 aggregation and premature fatal ALS-like disease. Here, we explored whether mutant hSOD1 aggregates with prion-like properties also exist in human ALS. Aggregate seeds were prepared from spinal cords from an ALS patient carrying the hSOD1G127Gfs*7 truncation mutation and from mice transgenic for the same mutation. To separate from mono-, di- or any oligomeric hSOD1 species, the seed preparation protocol included ultracentrifugation through a density cushion. The core structure of hSOD1G127Gfs*7 aggregates present in mice was strain A-like. Inoculation of the patient- or mouse-derived seeds into lumbar spinal cord of adult hSOD1-expressing mice induced strain A aggregation propagating along the neuraxis and premature fatal ALS-like disease (p < 0.0001). Inoculation of human or murine control seeds had no effect. The potencies of the ALS patient-derived seed preparations were high and disease was initiated in the transgenic mice by levels of hSOD1G127Gfs*7 aggregates much lower than those found in the motor system of patients carrying the mutation. The results suggest that prion-like growth and spread of hSOD1 aggregation could be the primary pathogenic mechanism, not only in hSOD1 transgenic rodent models, but also in human ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Mutation/genetics , Protein Aggregates/physiology , Spinal Cord/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Aged , Animals , Epitope Mapping , Female , Glial Fibrillary Acidic Protein/metabolism , Humans , Male , Mice , Mice, Transgenic , Spinal Cord/pathology , Superoxide Dismutase/chemistry
8.
J Clin Invest ; 126(6): 2249-53, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27140399

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is an adult-onset degeneration of motor neurons that is commonly caused by mutations in the gene encoding superoxide dismutase 1 (SOD1). Both patients and Tg mice expressing mutant human SOD1 (hSOD1) develop aggregates of unknown importance. In Tg mice, 2 different strains of hSOD1 aggregates (denoted A and B) can arise; however, the role of these aggregates in disease pathogenesis has not been fully characterized. Here, minute amounts of strain A and B hSOD1 aggregate seeds that were prepared by centrifugation through a density cushion were inoculated into lumbar spinal cords of 100-day-old mice carrying a human SOD1 Tg. Mice seeded with A or B aggregates developed premature signs of ALS and became terminally ill after approximately 100 days, which is 200 days earlier than for mice that had not been inoculated or were given a control preparation. Concomitantly, exponentially growing strain A and B hSOD1 aggregations propagated rostrally throughout the spinal cord and brainstem. The phenotypes provoked by the A and B strains differed regarding progression rates, distribution, end-stage aggregate levels, and histopathology. Together, our data indicate that the aggregate strains are prions that transmit a templated, spreading aggregation of hSOD1, resulting in a fatal ALS-like disease.


Subject(s)
Amyotrophic Lateral Sclerosis/etiology , Mutant Proteins/genetics , Prions/genetics , Superoxide Dismutase-1/genetics , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Animals , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Neurons/metabolism , Motor Neurons/pathology , Mutant Proteins/chemistry , Prions/chemistry , Protein Aggregates/genetics , Protein Aggregation, Pathological/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Spinal Cord/metabolism , Spinal Cord/pathology , Superoxide Dismutase-1/chemistry
9.
Proc Natl Acad Sci U S A ; 112(32): 9878-83, 2015 Aug 11.
Article in English | MEDLINE | ID: mdl-26221023

ABSTRACT

A longstanding challenge in studies of neurodegenerative disease has been that the pathologic protein aggregates in live tissue are not amenable to structural and kinetic analysis by conventional methods. The situation is put in focus by the current progress in demarcating protein aggregation in vitro, exposing new mechanistic details that are now calling for quantitative in vivo comparison. In this study, we bridge this gap by presenting a direct comparison of the aggregation kinetics of the ALS-associated protein superoxide dismutase 1 (SOD1) in vitro and in transgenic mice. The results based on tissue sampling by quantitative antibody assays show that the SOD1 fibrillation kinetics in vitro mirror with remarkable accuracy the spinal cord aggregate buildup and disease progression in transgenic mice. This similarity between in vitro and in vivo data suggests that, despite the complexity of live tissue, SOD1 aggregation follows robust and simplistic rules, providing new mechanistic insights into the ALS pathology and organism-level manifestation of protein aggregation phenomena in general.


Subject(s)
Amyotrophic Lateral Sclerosis/enzymology , Amyotrophic Lateral Sclerosis/pathology , Protein Aggregates , Superoxide Dismutase/chemistry , Superoxide Dismutase/metabolism , Animals , Apoproteins/chemistry , Apoproteins/metabolism , Disease Models, Animal , Kinetics , Mice, Transgenic , Mutation/genetics , Protein Unfolding , Spinal Cord/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , Survival Analysis
10.
Proc Natl Acad Sci U S A ; 112(14): 4489-94, 2015 Apr 07.
Article in English | MEDLINE | ID: mdl-25802384

ABSTRACT

Despite considerable progress in uncovering the molecular details of protein aggregation in vitro, the cause and mechanism of protein-aggregation disease remain poorly understood. One reason is that the amount of pathological aggregates in neural tissue is exceedingly low, precluding examination by conventional approaches. We present here a method for determination of the structure and quantity of aggregates in small tissue samples, circumventing the above problem. The method is based on binary epitope mapping using anti-peptide antibodies. We assessed the usefulness and versatility of the method in mice modeling the neurodegenerative disease amyotrophic lateral sclerosis, which accumulate intracellular aggregates of superoxide dismutase-1. Two strains of aggregates were identified with different structural architectures, molecular properties, and growth kinetics. Both were different from superoxide dismutase-1 aggregates generated in vitro under a variety of conditions. The strains, which seem kinetically under fragmentation control, are associated with different disease progressions, complying with and adding detail to the growing evidence that seeding, infectivity, and strain dependence are unifying principles of neurodegenerative disease.


Subject(s)
Epitope Mapping/methods , Proteins/chemistry , Superoxide Dismutase/genetics , Amino Acid Sequence , Amyotrophic Lateral Sclerosis/genetics , Animals , Brain/metabolism , Epitopes/chemistry , Humans , Mice , Mice, Transgenic , Molecular Sequence Data , Neurodegenerative Diseases/metabolism , Protein Conformation , Protein Folding , Protein Multimerization , Spinal Cord/metabolism , Superoxide Dismutase/chemistry , Superoxide Dismutase-1
11.
Hum Mol Genet ; 22(1): 51-60, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23026746

ABSTRACT

A common cause of amyotrophic lateral sclerosis (ALS) is mutations in the gene encoding superoxide dismutase-1. There is evolving circumstantial evidence that the wild-type protein can also be neurotoxic and that it may more generally be involved in the pathogenesis of ALS. To test this proposition more directly, we generated mice that express wild-type human superoxide dismutase-1 at a rate close to that of mutant superoxide dismutase-1 in the commonly studied G93A transgenic model. These mice developed an ALS-like syndrome and became terminally ill after around 370 days. The loss of spinal ventral neurons was similar to that in the G93A and other mutant superoxide dismutase-1 models, and large amounts of aggregated superoxide dismutase-1 were found in spinal cords, but also in the brain. The findings show that wild-type human superoxide dismutase-1 has the ability to cause ALS in mice, and they support the hypothesis of a more general involvement of the protein in the disease in humans.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Superoxide Dismutase/genetics , Amyotrophic Lateral Sclerosis/enzymology , Animals , Base Sequence , Blotting, Northern , Blotting, Western , DNA Primers , Humans , Mice , Mice, Transgenic , Polymerase Chain Reaction , Spinal Cord/enzymology , Superoxide Dismutase-1
12.
Neuromolecular Med ; 15(1): 147-58, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23076707

ABSTRACT

A common cause of amyotrophic lateral sclerosis is mutations in superoxide dismutase-1, which provoke the disease by an unknown mechanism. We have previously found that soluble hydrophobic misfolded mutant human superoxide dismutase-1 species are enriched in the vulnerable spinal cords of transgenic model mice. The levels were broadly inversely correlated with life spans, suggesting involvement in the pathogenesis. Here, we used methods based on antihuman superoxide dismutase-1 peptide antibodies specific for misfolded species to explore the composition and amounts of soluble misfolded human superoxide dismutase-1 in tissue extracts. Mice expressing 5 different human superoxide dismutase-1 variants with widely variable structural characteristics were examined. The levels were generally higher in spinal cords than in other tissues. The major portion of misfolded superoxide dismutase-1 was shown to be monomers lacking the C57-C146 disulfide bond with large hydrodynamic volume, indicating a severely disordered structure. The remainder of the misfolded protein appeared to be non-covalently associated in 130- and 250-kDa complexes. The malleable monomers should be prone to aggregate and associate with other cellular components, and should be easily translocated between compartments. They may be the primary cause of toxicity in superoxide dismutase-1-induced amyotrophic lateral sclerosis.


Subject(s)
Amyotrophic Lateral Sclerosis/enzymology , Disease Models, Animal , Protein Folding , Spinal Cord/enzymology , Superoxide Dismutase/chemistry , Amyotrophic Lateral Sclerosis/genetics , Animals , Chelating Agents/pharmacology , Chromatography, Gel , Copper , Cysteine/analysis , Cystine/analysis , Enzyme-Linked Immunosorbent Assay , Humans , Hydrophobic and Hydrophilic Interactions , Longevity/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation, Missense , Oxidation-Reduction , Protein Conformation , Protein Folding/drug effects , Recombinant Proteins/chemistry , Solubility , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , Zinc
13.
PLoS One ; 7(4): e36104, 2012.
Article in English | MEDLINE | ID: mdl-22558346

ABSTRACT

Neurodegeneration in protein-misfolding disease is generally assigned to toxic function of small, soluble protein aggregates. Largely, these assignments are based on observations of cultured neural cells where the suspect protein material is titrated directly into the growth medium. In the present study, we use this approach to shed light on the cytotoxic action of the metalloenzyme Cu/Zn superoxide dismutase 1 (SOD1), associated with misfolding and aggregation in amyotrophic lateral sclerosis (ALS). The results show, somewhat unexpectedly, that the toxic species of SOD1 in this type of experimental setting is not an aggregate, as typically observed for proteins implicated in other neuro-degenerative diseases, but the folded and fully soluble apo protein. Moreover, we demonstrate that the toxic action of apoSOD1 relies on the protein's ability to chelate Zn(2+) ions from the growth medium. The decreased cell viability that accompanies this extraction is presumably based on disturbed Zn(2+) homeostasis. Consistently, mutations that cause global unfolding of the apoSOD1 molecule or otherwise reduce its Zn(2+) affinity abolish completely the cytotoxic response. So does the addition of surplus Zn(2+). Taken together, these observations point at a case where the toxic response of cultured cells might not be related to human pathology but stems from the intrinsic limitations of a simplified cell model. There are several ways proteins can kill cultured neural cells but all of these need not to be relevant for neurodegenerative disease.


Subject(s)
Chelating Agents/pharmacology , Superoxide Dismutase/toxicity , Zinc/metabolism , Amino Acid Sequence , Apoproteins/toxicity , Catalytic Domain , Cell Death/drug effects , Cell Survival/drug effects , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Ligands , Models, Biological , Molecular Sequence Data , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Mutation/genetics , Neuroblastoma/pathology , Pliability/drug effects , Protein Multimerization/drug effects , Protein Stability/drug effects , Protein Structure, Quaternary , Protein Structure, Secondary , Serum , Superoxide Dismutase/chemistry , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , Time Factors , Transition Temperature/drug effects , Tumor Cells, Cultured
14.
J Biol Chem ; 286(23): 20130-6, 2011 Jun 10.
Article in English | MEDLINE | ID: mdl-21493711

ABSTRACT

Mutant superoxide dismutase-1 (SOD1) has an unidentified toxic property that provokes ALS. Several ALS-linked SOD1 mutations cause long C-terminal truncations, which suggests that common cytotoxic SOD1 conformational species should be misfolded and that the C-terminal end cannot be involved. The cytotoxicity may arise from interaction of cellular proteins with misfolded SOD1 species. Here we specifically immunocaptured misfolded SOD1 by the C-terminal end, from extracts of spinal cords from transgenic ALS model mice. Associated proteins were identified with proteomic techniques. Two transgenic models expressing SOD1s with contrasting molecular properties were examined: the stable G93A mutant, which is abundant in the spinal cord with only a tiny subfraction misfolded, and the scarce disordered truncation mutant G127insTGGG. For comparison, proteins in spinal cord extracts with affinity for immobilized apo G93A mutant SOD1 were determined. Two-dimensional gel patterns with a limited number of bound proteins were found, which were similar for the two SOD1 mutants. Apart from neurofilament light, the proteins identified were all chaperones and by far most abundant was Hsc70. The immobilized apo G93A SOD1, which would populate a variety of conformations, was found to bind to a considerable number of additional proteins. A substantial proportion of the misfolded SOD1 in the spinal cord extracts appeared to be chaperone-associated. Still, only about 1% of the Hsc70 appeared to be associated with misfolded SOD1. The results argue against the notion that chaperone depletion is involved in ALS pathogenesis in the transgenic models and in humans carrying SOD1 mutations.


Subject(s)
Amyotrophic Lateral Sclerosis/enzymology , Mutation, Missense , Protein Folding , Spinal Cord/enzymology , Superoxide Dismutase/metabolism , Amino Acid Substitution , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Disease Models, Animal , Electrophoresis, Gel, Two-Dimensional/methods , HSC70 Heat-Shock Proteins/genetics , HSC70 Heat-Shock Proteins/metabolism , Humans , Mice , Mice, Transgenic , Proteomics/methods , Spinal Cord/pathology , Superoxide Dismutase/genetics , Superoxide Dismutase-1
15.
J Neurochem ; 117(1): 91-9, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21226712

ABSTRACT

Several of the superoxide dismutase-1 (SOD1) mutations linked to amyotrophic lateral sclerosis (ALS) lead to synthesis of structurally defective molecules, suggesting that any cytotoxic conformational species common for all mutations should be misfolded. SOD1 can be secreted and evidence from ALS model systems suggests that extracellular SOD1 may be involved in cytotoxicity. Three ELISAs specifically reacting with different sequence segments in misfolded SOD1 species were used for analysis of CSF from 38 neurological controls and from 96 ALS patients, 57 of whom were sporadic cases and 39 familial, including 22 patients carrying SOD1 mutations. Misfolded SOD1 was found in all samples. There were, however, no significant differences between patients with and without mutations, and between all the ALS patients and the controls. The estimated concentration of misfolded SOD1 in the interstitium of the CNS is a 1000 times lower than that required for appreciable cytotoxicity in model systems. The results argue against a direct cytotoxic role of extracellular misfolded SOD1 in ALS. Misfolded SOD1 in CSF cannot be used as a biomarker of ALS in patients with and without mutations in the enzyme.


Subject(s)
Amyotrophic Lateral Sclerosis/cerebrospinal fluid , Amyotrophic Lateral Sclerosis/enzymology , Protein Folding , Superoxide Dismutase/cerebrospinal fluid , Adult , Aged , Aged, 80 and over , Amino Acid Sequence , Amyotrophic Lateral Sclerosis/genetics , Animals , Biomarkers/cerebrospinal fluid , Contraindications , Humans , Male , Mice , Mice, Transgenic , Middle Aged , Molecular Sequence Data , Mutation/genetics , Superoxide Dismutase/genetics , Superoxide Dismutase-1
16.
Proc Natl Acad Sci U S A ; 106(24): 9667-72, 2009 Jun 16.
Article in English | MEDLINE | ID: mdl-19497878

ABSTRACT

The structural integrity of the ubiquitous enzyme superoxide dismutase (SOD1) relies critically on the correct coordination of Cu and Zn. Loss of these cofactors not only promotes SOD1 aggregation in vitro but also seems to be a key prerequisite for pathogenic misfolding in the neurodegenerative disease amyotrophic lateral sclerosis (ALS). We examine here the consequences of Zn(2+) loss by selectively removing the Zn site, which has been implicated as the main modulator of SOD1 stability and disease competence. After Zn-site removal, the remaining Cu ligands can coordinate a nonnative Zn(2+) ion with microM affinity in the denatured state, and then retain this ion throughout the folding reaction. Without the restriction of a metallated Zn site, however, the Cu ligands fail to correctly coordinate the nonnative Zn(2+) ion: Trapping of a water molecule causes H48 to change rotamer and swing outwards. The misligation is sterically incompatible with the native structure. As a consequence, SOD1 unfolds locally and interacts with neighboring molecules in the crystal lattice. The findings point to a critical role for the native Zn site in controlling SOD1 misfolding, and show that even subtle changes of the metal-loading sequence can render the wild-type protein the same structural properties as ALS-provoking mutations. This frustrated character of the SOD1 molecule seems to arise from a compromise between optimization of functional and structural features.


Subject(s)
Amyotrophic Lateral Sclerosis/enzymology , Protein Folding , Superoxide Dismutase/metabolism , Humans , Ligands , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Superoxide Dismutase/chemistry
17.
Proc Natl Acad Sci U S A ; 104(35): 14157-62, 2007 Aug 28.
Article in English | MEDLINE | ID: mdl-17715066

ABSTRACT

Mutants of superoxide dismutase-1 (SOD1) cause ALS by an unidentified cytotoxic mechanism. We have previously shown that the stable SOD1 mutants D90A and G93A are abundant and show the highest levels in liver and kidney in transgenic murine ALS models, whereas the unstable G85R and G127X mutants are scarce but enriched in the CNS. These data indicated that minute amounts of misfolded SOD1 enriched in the motor areas might exert the ALS-causing cytotoxicity. A hydrophobic interaction chromatography (HIC) protocol was developed with the aim to determine the abundance of soluble misfolded SOD1 in tissues in vivo. Most G85R and G127X mutant SOD1s bound in the assay, but only minute subfractions of the D90A and G93A mutants. The absolute levels of HIC-binding SOD1 were, however, similar and broadly inversely related to lifespans in the models. They were generally enriched in the susceptible spinal cord. The HIC-binding SOD1 was composed of disulfide-reduced subunits lacking metal ions and also subunits that apparently carried nonnative intrasubunit disulfide bonds. The levels were high from birth until death and were comparable to the amounts of SOD1 that become sequestered in aggregates in the terminal stage. The HIC-binding SOD1 species ranged from monomeric to trimeric in size. These species form a least common denominator amongst SOD1 mutants with widely different molecular characteristics and might be involved in the cytotoxicity that causes ALS.


Subject(s)
Motor Neuron Disease/genetics , Mutation , Spinal Cord/physiopathology , Superoxide Dismutase/genetics , Animals , Disease Models, Animal , Genetic Variation , Humans , Mice , Mice, Transgenic , Motor Neuron Disease/enzymology , Oxidation-Reduction , Protein Denaturation , Protein Folding , Protein Subunits , Sequence Deletion , Spinal Cord/growth & development , Superoxide Dismutase/metabolism , Superoxide Dismutase-1
SELECTION OF CITATIONS
SEARCH DETAIL