Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 318
Filter
Add more filters

Publication year range
1.
PLoS Biol ; 22(1): e3002462, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38289969

ABSTRACT

Mutations in the gene encoding Cu-Zn superoxide dismutase 1 (SOD1) cause a subset of familial amyotrophic lateral sclerosis (fALS) cases. A shared effect of these mutations is that SOD1, which is normally a stable dimer, dissociates into toxic monomers that seed toxic aggregates. Considerable research effort has been devoted to developing compounds that stabilize the dimer of fALS SOD1 variants, but unfortunately, this has not yet resulted in a treatment. We hypothesized that cyclic thiosulfinate cross-linkers, which selectively target a rare, 2 cysteine-containing motif, can stabilize fALS-causing SOD1 variants in vivo. We created a library of chemically diverse cyclic thiosulfinates and determined structure-cross-linking-activity relationships. A pre-lead compound, "S-XL6," was selected based upon its cross-linking rate and drug-like properties. Co-crystallographic structure clearly establishes the binding of S-XL6 at Cys 111 bridging the monomers and stabilizing the SOD1 dimer. Biophysical studies reveal that the degree of stabilization afforded by S-XL6 (up to 24°C) is unprecedented for fALS, and to our knowledge, for any protein target of any kinetic stabilizer. Gene silencing and protein degrading therapeutic approaches require careful dose titration to balance the benefit of diminished fALS SOD1 expression with the toxic loss-of-enzymatic function. We show that S-XL6 does not share this liability because it rescues the activity of fALS SOD1 variants. No pharmacological agent has been proven to bind to SOD1 in vivo. Here, using a fALS mouse model, we demonstrate oral bioavailability; rapid engagement of SOD1G93A by S-XL6 that increases SOD1G93A's in vivo half-life; and that S-XL6 crosses the blood-brain barrier. S-XL6 demonstrated a degree of selectivity by avoiding off-target binding to plasma proteins. Taken together, our results indicate that cyclic thiosulfinate-mediated SOD1 stabilization should receive further attention as a potential therapeutic approach for fALS.


Subject(s)
Amyotrophic Lateral Sclerosis , Animals , Mice , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Cysteine/genetics , Mutation , Superoxide Dismutase/genetics , Superoxide Dismutase/chemistry , Superoxide Dismutase/metabolism , Superoxide Dismutase-1/genetics
3.
EMBO J ; 41(2): e105531, 2022 12 17.
Article in English | MEDLINE | ID: mdl-34904718

ABSTRACT

Recessive gene mutations underlie many developmental disorders and often lead to disabling neurological problems. Here, we report identification of a homozygous c.170G>A (p.Cys57Tyr or C57Y) mutation in the gene coding for protein disulfide isomerase A3 (PDIA3, also known as ERp57), an enzyme that catalyzes formation of disulfide bonds in the endoplasmic reticulum, to be associated with syndromic intellectual disability. Experiments in zebrafish embryos show that PDIA3C57Y expression is pathogenic and causes developmental defects such as axonal disorganization as well as skeletal abnormalities. Expression of PDIA3C57Y in the mouse hippocampus results in impaired synaptic plasticity and memory consolidation. Proteomic and functional analyses reveal that PDIA3C57Y expression leads to dysregulation of cell adhesion and actin cytoskeleton dynamics, associated with altered integrin biogenesis and reduced neuritogenesis. Biochemical studies show that PDIA3C57Y has decreased catalytic activity and forms disulfide-crosslinked aggregates that abnormally interact with chaperones in the endoplasmic reticulum. Thus, rare disease gene variant can provide insight into how perturbations of neuronal proteostasis can affect the function of the nervous system.


Subject(s)
Developmental Disabilities/genetics , Endoplasmic Reticulum/metabolism , Protein Disulfide-Isomerases/genetics , Proteostasis , Adolescent , Adult , Animals , Axons/metabolism , Axons/pathology , Cell Adhesion , Cells, Cultured , Child , Cytoskeleton/metabolism , Developmental Disabilities/metabolism , Developmental Disabilities/pathology , Female , Hippocampus/metabolism , Hippocampus/pathology , Humans , Integrins/metabolism , Male , Mice , Mice, Inbred C57BL , Mutation, Missense , Neuronal Outgrowth , Neuronal Plasticity , Pedigree , Protein Disulfide-Isomerases/metabolism , Zebrafish
4.
Cell ; 140(1): 88-98, 2010 Jan 08.
Article in English | MEDLINE | ID: mdl-20074522

ABSTRACT

Thyrotoxic hypokalemic periodic paralysis (TPP) is characterized by acute attacks of weakness, hypokalemia, and thyrotoxicosis of various etiologies. These transient attacks resemble those of patients with familial hypokalemic periodic paralysis (hypoKPP) and resolve with treatment of the underlying hyperthyroidism. Because of the phenotypic similarity of these conditions, we hypothesized that TPP might also be a channelopathy. While sequencing candidate genes, we identified a previously unreported gene (not present in human sequence databases) that encodes an inwardly rectifying potassium (Kir) channel, Kir2.6. This channel, nearly identical to Kir2.2, is expressed in skeletal muscle and is transcriptionally regulated by thyroid hormone. Expression of Kir2.6 in mammalian cells revealed normal Kir currents in whole-cell and single-channel recordings. Kir2.6 mutations were present in up to 33% of the unrelated TPP patients in our collection. Some of these mutations clearly alter a variety of Kir2.6 properties, all altering muscle membrane excitability leading to paralysis.


Subject(s)
Genetic Predisposition to Disease , Hypokalemic Periodic Paralysis/genetics , Mutation , Potassium Channels, Inwardly Rectifying/genetics , Amino Acid Sequence , Base Sequence , DNA Mutational Analysis , Electrophysiological Phenomena , Humans , Hypokalemic Periodic Paralysis/metabolism , Molecular Sequence Data , Potassium Channels, Inwardly Rectifying/chemistry , Potassium Channels, Inwardly Rectifying/metabolism , Transcription, Genetic , Triiodothyronine/metabolism
5.
Gene Ther ; 31(3-4): 105-118, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37752346

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease that affects motor neurons, causing progressive muscle weakness and respiratory failure. The presence of an expanded hexanucleotide repeat in chromosome 9 open reading frame 72 (C9ORF72) is the most frequent mutation causing familial ALS and frontotemporal dementia (FTD). To determine if suppressing expression of C9ORF72 gene products can reduce toxicity, we designed a set of artificial microRNAs (amiRNA) targeting the human C9ORF72 gene. Here we report that an AAV9-mediated amiRNA significantly suppresses expression of the C9ORF72 mRNA, protein, and toxic dipeptide repeat proteins generated by the expanded repeat in the brain and spinal cord of C9ORF72 transgenic mice.


Subject(s)
Amyotrophic Lateral Sclerosis , MicroRNAs , Neurodegenerative Diseases , Animals , Humans , Mice , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/therapy , C9orf72 Protein/genetics , C9orf72 Protein/metabolism , Dipeptides/genetics , Dipeptides/metabolism , DNA Repeat Expansion/genetics , Mice, Transgenic , MicroRNAs/genetics , Proteins/genetics , Proteins/metabolism
6.
Neurobiol Dis ; 192: 106414, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38253209

ABSTRACT

Alteration in protein citrullination (PC), a common posttranslational modification (PTM), contributes to pathogenesis in various inflammatory disorders. We previously reported that PC and protein arginine deiminase 2 (PAD2), the predominant enzyme isoform that catalyzes this PTM in the central nervous system (CNS), are altered in mouse models of amyotrophic lateral sclerosis (ALS). We now demonstrate that PAD2 expression and PC are altered in human postmortem ALS spinal cord and motor cortex compared to controls, increasing in astrocytes while trending lower in neurons. Furthermore, PC is enriched in protein aggregates that contain the myelin proteins PLP and MBP in ALS. These results confirm our findings in ALS mouse models and suggest that altered PAD2 and PC contribute to neurodegeneration in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis , Citrullination , Animals , Humans , Mice , Amyotrophic Lateral Sclerosis/metabolism , Gliosis/metabolism , Hydrolases/genetics , Hydrolases/metabolism , Myelin Proteins/metabolism , Myelin Sheath/pathology , Protein Aggregates , Protein-Arginine Deiminase Type 2/metabolism , Protein-Arginine Deiminases/metabolism , Proteins/metabolism , Spinal Cord/pathology
7.
Muscle Nerve ; 69(6): 719-729, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38593477

ABSTRACT

INTRODUCTION/AIMS: Biomarkers have shown promise in amyotrophic lateral sclerosis (ALS) research, but the quest for reliable biomarkers remains active. This study evaluates the effect of debamestrocel on cerebrospinal fluid (CSF) biomarkers, an exploratory endpoint. METHODS: A total of 196 participants randomly received debamestrocel or placebo. Seven CSF samples were to be collected from all participants. Forty-five biomarkers were analyzed in the overall study and by two subgroups characterized by the ALS Functional Rating Scale-Revised (ALSFRS-R). A prespecified model was employed to predict clinical outcomes leveraging biomarkers and disease characteristics. Causal inference was used to analyze relationships between neurofilament light chain (NfL) and ALSFRS-R. RESULTS: We observed significant changes with debamestrocel in 64% of the biomarkers studied, spanning pathways implicated in ALS pathology (63% neuroinflammation, 50% neurodegeneration, and 89% neuroprotection). Biomarker changes with debamestrocel show biological activity in trial participants, including those with advanced ALS. CSF biomarkers were predictive of clinical outcomes in debamestrocel-treated participants (baseline NfL, baseline latency-associated peptide/transforming growth factor beta1 [LAP/TGFß1], change galectin-1, all p < .01), with baseline NfL and LAP/TGFß1 remaining (p < .05) when disease characteristics (p < .005) were incorporated. Change from baseline to the last measurement showed debamestrocel-driven reductions in NfL were associated with less decline in ALSFRS-R. Debamestrocel significantly reduced NfL from baseline compared with placebo (11% vs. 1.6%, p = .037). DISCUSSION: Following debamestrocel treatment, many biomarkers showed increases (anti-inflammatory/neuroprotective) or decreases (inflammatory/neurodegenerative) suggesting a possible treatment effect. Neuroinflammatory and neuroprotective biomarkers were predictive of clinical response, suggesting a potential multimodal mechanism of action. These results offer preliminary insights that need to be confirmed.


Subject(s)
Amyotrophic Lateral Sclerosis , Biomarkers , Neurofilament Proteins , Adult , Aged , Female , Humans , Male , Middle Aged , Amyotrophic Lateral Sclerosis/cerebrospinal fluid , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/diagnosis , Biomarkers/cerebrospinal fluid , Double-Blind Method , Neurofilament Proteins/cerebrospinal fluid , Treatment Outcome
8.
Brain ; 146(12): 5139-5152, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37527465

ABSTRACT

Frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) are fatal neurodegenerative diseases that represent ends of the spectrum of a single disease. The most common genetic cause of FTD and ALS is a hexanucleotide repeat expansion in the C9orf72 gene. Although epidemiological data suggest that traumatic brain injury (TBI) represents a risk factor for FTD and ALS, its role in exacerbating disease onset and course remains unclear. To explore the interplay between traumatic brain injury and genetic risk in the induction of FTD/ALS pathology we combined a mild repetitive traumatic brain injury paradigm with an established bacterial artificial chromosome transgenic C9orf72 (C9BAC) mouse model without an overt motor phenotype or neurodegeneration. We assessed 8-10 week-old littermate C9BACtg/tg (n = 21), C9BACtg/- (n = 20) and non-transgenic (n = 21) mice of both sexes for the presence of behavioural deficits and cerebral histopathology at 12 months after repetitive TBI. Repetitive TBI did not affect body weight gain, general neurological deficit severity, nor survival over the 12-month observation period and there was no difference in rotarod performance, object recognition, social interaction and acoustic characteristics of ultrasonic vocalizations of C9BAC mice subjected to repetitive TBI versus sham injury. However, we found that repetitive TBI increased the time to the return of the righting reflex, reduced grip force, altered sociability behaviours and attenuated ultrasonic call emissions during social interactions in C9BAC mice. Strikingly, we found that repetitive TBI caused widespread microglial activation and reduced neuronal density that was associated with loss of histological markers of axonal and synaptic integrity as well as profound neuronal transactive response DNA binding protein 43 kDa mislocalization in the cerebral cortex of C9BAC mice at 12 months; this was not observed in non-transgenic repetitive TBI and C9BAC sham mice. Our data indicate that repetitive TBI can be an environmental risk factor that is sufficient to trigger FTD/ALS-associated neuropathology and behavioural deficits, but not paralysis, in mice carrying a C9orf72 hexanucleotide repeat expansion.


Subject(s)
Amyotrophic Lateral Sclerosis , Brain Concussion , C9orf72 Protein , Frontotemporal Dementia , Pick Disease of the Brain , Animals , Female , Male , Mice , Amyotrophic Lateral Sclerosis/genetics , Brain Concussion/pathology , C9orf72 Protein/genetics , C9orf72 Protein/metabolism , DNA Repeat Expansion , Frontotemporal Dementia/genetics , Frontotemporal Dementia/pathology , Mice, Transgenic
9.
N Engl J Med ; 383(2): 151-158, 2020 07 09.
Article in English | MEDLINE | ID: mdl-32640133

ABSTRACT

Two patients with familial amyotrophic lateral sclerosis (ALS) and mutations in the gene encoding superoxide dismutase 1 (SOD1) were treated with a single intrathecal infusion of adeno-associated virus encoding a microRNA targeting SOD1. In Patient 1, SOD1 levels in spinal cord tissue as analyzed on autopsy were lower than corresponding levels in untreated patients with SOD1-mediated ALS and in healthy controls. Levels of SOD1 in cerebrospinal fluid were transiently and only slightly lower in Patient 1 but were not affected in Patient 2. In Patient 1, meningoradiculitis developed after the infusion; Patient 2 was pretreated with immunosuppressive drugs and did not have this complication. Patient 1 had transient improvement in the strength of his right leg, a measure that had been relatively stable throughout his disease course, but there was no change in his vital capacity. Patient 2 had stable scores on a composite measure of ALS function and a stable vital capacity during a 12-month period. This study showed that intrathecal microRNA can be used as a potential treatment for SOD1-mediated ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/therapy , MicroRNAs/therapeutic use , Superoxide Dismutase-1/cerebrospinal fluid , Amyotrophic Lateral Sclerosis/cerebrospinal fluid , Amyotrophic Lateral Sclerosis/genetics , Dependovirus , Fatal Outcome , Gene Silencing , Genetic Therapy , Genetic Vectors , Humans , Injections, Spinal , Male , Meningoencephalitis , Middle Aged , Mutation , Proof of Concept Study , Spinal Cord/chemistry , Spinal Cord/pathology , Superoxide Dismutase-1/analysis , Superoxide Dismutase-1/genetics , Vital Capacity , Young Adult
10.
Ann Neurol ; 91(5): 716-729, 2022 05.
Article in English | MEDLINE | ID: mdl-35178738

ABSTRACT

OBJECTIVE: The objective of this study is to develop a novel method for monitoring the integrity of motor neurons in vivo by quantifying net retrograde axonal transport. METHODS: The method uses single photon emission computed tomography to quantify retrograde transport to spinal cord of tetanus toxin fragment C (125 I-TTC) following intramuscular injection. We characterized the transport profiles in 3 transgenic mouse models carrying amyotrophic lateral sclerosis (ALS)-associated genes, aging mice, and SOD1G93A transgenic mice following CRISPR/Cas9 gene editing. Lastly, we studied the effect of prior immunization of tetanus toxoid on the transport profile of TTC. RESULTS: This technique defines a quantitative profile of net retrograde axonal transport of TTC in living mice. The profile is distinctly abnormal in transgenic SOD1G93A mice as young as 65 days (presymptomatic) and worsens with disease progression. Moreover, this method detects a distinct therapeutic benefit of gene editing in transgenic SOD1G93A mice well before other clinical parameters (eg, grip strength) show improvement. Symptomatic transgenic PFN1C71G/C71G ALS mice display gross reductions in net retrograde axonal transport, which is also disturbed in asymptomatic mice harboring a human C9ORF72 transgene with an expanded GGGGCC repeat motif. In wild-type mice, net retrograde axonal transport declines with aging. Lastly, prior immunization with tetanus toxoid does not preclude use of this assay. INTERPRETATION: This assay of net retrograde axonal transport has broad potential clinical applications and should be particularly valuable as a physiological biomarker that permits early detection of benefit from potential therapies for motor neuron diseases. ANN NEUROL 2022;91:716-729.


Subject(s)
Amyotrophic Lateral Sclerosis , Axonal Transport , Amyotrophic Lateral Sclerosis/diagnostic imaging , Amyotrophic Lateral Sclerosis/genetics , Animals , Axonal Transport/genetics , Biomarkers , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Profilins , Spinal Cord/diagnostic imaging , Spinal Cord/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , Tetanus Toxoid
11.
Chem Res Toxicol ; 36(5): 734-746, 2023 05 15.
Article in English | MEDLINE | ID: mdl-37126467

ABSTRACT

In our earlier work (Golden et al., 2021), we showed 70-80% accuracies for several skin sensitization computational tools using human data. Here, we expanded the data set using the NICEATM human skin sensitization database to create a final data set of 1355 discrete chemicals (largely negative, ∼70%). Using this expanded data set, we analyzed model performance and evaluated mispredictions using Toxtree (v 3.1.0), OECD QSAR Toolbox (v 4.5), VEGA's (1.2.0 BETA) CAESAR (v 2.1.7), and a k-nearest-neighbor (kNN) classification approach. We show that the accuracy on this data set was lower than previous estimates, with balanced accuracies being 63% and 65% for Toxtree and OECD QSAR Toolbox, respectively, 46% for VEGA, and 59% for a kNN approach, with the lower accuracy likely due to the higher percentage of nonsensitizing chemicals. Two hundred eighty seven chemicals were mispredicted by both Toxtree and OECD QSAR Toolbox, which was approximately 20% of the entire data set, and 84% of these were false positives. The absence or presence of metabolic simulation in OECD QSAR Toolbox made no overall difference. While Toxtree is known for overpredicting, 60% of the chemicals in the data set had no alert for skin sensitization, and a substantial number of these chemicals were in fact sensitizers, pointing to sensitization mechanisms not recognized by Toxtree. Interestingly, we observed that chemicals with more than one Toxtree alert were more likely to be nonsensitizers. Finally, a kNN approach tended to mispredict different chemicals than either OECD QSAR Toolbox or Toxtree, suggesting that there was additional information to be garnered from a kNN approach. Overall, the results demonstrate that while there is merit in structural alerts as well as QSAR or read-across approaches (perhaps even more so in their combination), additional improvement will require a more nuanced understanding of mechanisms of skin sensitization.


Subject(s)
Quantitative Structure-Activity Relationship , Skin , Humans , Skin/metabolism , Computer Simulation
12.
Am J Respir Crit Care Med ; 206(7): 838-845, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35649189

ABSTRACT

Rationale: There are no pharmacologic agents that modify emphysema progression in patients with chronic obstructive pulmonary disease (COPD). Objectives: To evaluate the efficacy of losartan, an angiotensin receptor blocker, to reduce emphysema progression. Methods: The trial was a multicenter, randomized, placebo-controlled trial conducted between May 2017 and January 2021. Eligible participants were aged ⩾40 years, had moderate to severe airflow obstruction, ⩾10 pack-years of smoking, mild-moderate emphysema on high-resolution computed tomography, and no medical indication for or intolerance of angiotensin receptor blockers. Treatment with losartan 100 mg daily or matching placebo (1:1) was randomly assigned. The primary outcome was emphysema progression on high-resolution computed tomography over 48 weeks. Secondary outcomes included the St George's Respiratory Questionnaire, the modified Medical Research Council dyspnea scale, the COPD Assessment Test, and the Physical Function-Short Form 20a. Measurements and Main Results: A total of 220 participants were enrolled; 58% were men, 19% were African American, and 24% were current smokers. The medians (interquartile ranges) for age were 65 (61-73) years and 48 (36-59) for percent predicted FEV1 after bronchodilator use. The mean (95% confidence interval) percentage emphysema progression was 1.35% (0.67-2.03) in the losartan group versus 0.66% (0.09-1.23) in the placebo group (P = NS). Conclusions: Losartan did not prevent emphysema progression in people with COPD with mild-moderate emphysema. Clinical trial registered with www.clinicaltrials.gov (NCT02696564).


Subject(s)
Emphysema , Pulmonary Disease, Chronic Obstructive , Pulmonary Emphysema , Aged , Angiotensin Receptor Antagonists/therapeutic use , Bronchodilator Agents/therapeutic use , Disease Progression , Female , Forced Expiratory Volume , Humans , Losartan/therapeutic use , Male , Pulmonary Disease, Chronic Obstructive/complications , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Emphysema/complications , Pulmonary Emphysema/drug therapy
13.
Genome Res ; 29(5): 809-818, 2019 05.
Article in English | MEDLINE | ID: mdl-30940688

ABSTRACT

Large-scale sequencing efforts in amyotrophic lateral sclerosis (ALS) have implicated novel genes using gene-based collapsing methods. However, pathogenic mutations may be concentrated in specific genic regions. To address this, we developed two collapsing strategies: One focuses rare variation collapsing on homology-based protein domains as the unit for collapsing, and the other is a gene-level approach that, unlike standard methods, leverages existing evidence of purifying selection against missense variation on said domains. The application of these two collapsing methods to 3093 ALS cases and 8186 controls of European ancestry, and also 3239 cases and 11,808 controls of diversified populations, pinpoints risk regions of ALS genes, including SOD1, NEK1, TARDBP, and FUS While not clearly implicating novel ALS genes, the new analyses not only pinpoint risk regions in known genes but also highlight candidate genes as well.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , DNA Mutational Analysis/methods , Genetic Predisposition to Disease , Genome-Wide Association Study/methods , Female , Genetic Variation , Humans , Male , Mutation , NIMA-Related Kinase 1/genetics , Protein Domains/genetics , RNA-Binding Protein FUS/genetics , Risk Factors , Superoxide Dismutase-1/genetics , White People/genetics , Exome Sequencing/methods
14.
Muscle Nerve ; 65(3): 291-302, 2022 03.
Article in English | MEDLINE | ID: mdl-34890069

ABSTRACT

INTRODUCTION/AIMS: Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative illness with great unmet patient need. We aimed to evaluate whether mesenchymal stem cells induced to secrete high levels of neurotrophic factors (MSC-NTF), a novel autologous cell-therapy capable of targeting multiple pathways, could safely slow ALS disease progression. METHODS: This randomized, double-blind, placebo-controlled study enrolled ALS participants meeting revised El Escorial criteria, revised ALS Functional Rating Scale (ALSFRS-R) ≥25 (screening) and ≥3 ALSFRS-R points decline prior to randomization. Participants received three treatments of MSC-NTF or placebo intrathecally. The primary endpoint evaluated efficacy of MSC-NTF through a responder analysis and safety. A change in disease progression post-treatment of ≥1.25 points/mo defines a clinical response. A pre-specified analysis leveraged baseline ALSFRS-R of 35 as a subgroup threshold. RESULTS: Overall, MSC-NTF treatment was well tolerated; there were no safety concerns. Thirty-three percent of MSC-NTF and 28% of placebo participants met clinical response criteria at 28 wk (odds ratio [OR] = 1.33, P = .45); thus, the primary endpoint was not met. A pre-specified analysis of participants with baseline ALSFRS-R ≥ 35 (n = 58) showed a clinical response rate at 28 wk of 35% MSC-NTF and 16% placebo (OR = 2.6, P = .29). Significant improvements in cerebrospinal biomarkers of neuroinflammation, neurodegeneration, and neurotrophic factor support were observed with MSC-NTF, with placebo unchanged. DISCUSSION: The study did not reach statistical significance on the primary endpoint. However, a pre-specified subgroup suggests that MSC-NTF participants with less severe disease may have retained more function compared to placebo. Given the unmet patient need, the results of this trial warrant further investigation.


Subject(s)
Amyotrophic Lateral Sclerosis , Mesenchymal Stem Cells , Amyotrophic Lateral Sclerosis/diagnosis , Double-Blind Method , Humans , Nerve Growth Factors/metabolism , Transplantation, Autologous
15.
Nature ; 539(7628): 197-206, 2016 Nov 10.
Article in English | MEDLINE | ID: mdl-27830784

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a progressive and uniformly fatal neurodegenerative disease. A plethora of genetic factors have been identified that drive the degeneration of motor neurons in ALS, increase susceptibility to the disease or influence the rate of its progression. Emerging themes include dysfunction in RNA metabolism and protein homeostasis, with specific defects in nucleocytoplasmic trafficking, the induction of stress at the endoplasmic reticulum and impaired dynamics of ribonucleoprotein bodies such as RNA granules that assemble through liquid-liquid phase separation. Extraordinary progress in understanding the biology of ALS provides new reasons for optimism that meaningful therapies will be identified.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/physiopathology , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/therapy , Animals , Biological Transport , C9orf72 Protein , Endoplasmic Reticulum Stress/genetics , Frontotemporal Dementia/genetics , Humans , Nervous System/pathology , Nervous System/physiopathology , Organelles/genetics , Organelles/metabolism , Organelles/pathology , Prions/metabolism , Proteins/genetics , Proteins/metabolism , Proteolysis , RNA/biosynthesis , RNA/genetics , RNA/metabolism , RNA/toxicity
16.
Int J Mol Sci ; 23(24)2022 Dec 16.
Article in English | MEDLINE | ID: mdl-36555655

ABSTRACT

ALS-linked mutations induce aberrant conformations within the SOD1 protein that are thought to underlie the pathogenic mechanism of SOD1-mediated ALS. Although clinical trials are underway for gene silencing of SOD1, these approaches reduce both wild-type and mutated forms of SOD1. Here, we sought to develop anti-SOD1 nanobodies with selectivity for mutant and misfolded forms of human SOD1 over wild-type SOD1. Characterization of two anti-SOD1 nanobodies revealed that these biologics stabilize mutant SOD1 in vitro. Further, SOD1 expression levels were enhanced and the physiological subcellular localization of mutant SOD1 was restored upon co-expression of anti-SOD1 nanobodies in immortalized cells. In human motor neurons harboring the SOD1 A4V mutation, anti-SOD1 nanobody expression promoted neurite outgrowth, demonstrating a protective effect of anti-SOD1 nanobodies in otherwise unhealthy cells. In vitro assays revealed that an anti-SOD1 nanobody exhibited selectivity for human mutant SOD1 over endogenous murine SOD1, thus supporting the preclinical utility of anti-SOD1 nanobodies for testing in animal models of ALS. In sum, the anti-SOD1 nanobodies developed and presented herein represent viable biologics for further preclinical testing in human and mouse models of ALS.


Subject(s)
Amyotrophic Lateral Sclerosis , Single-Domain Antibodies , Humans , Mice , Animals , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Single-Domain Antibodies/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Protein Folding , Motor Neurons/metabolism , Neuronal Outgrowth , Mutation
17.
Neurobiol Dis ; 155: 105368, 2021 07.
Article in English | MEDLINE | ID: mdl-33892050

ABSTRACT

Parkinson's disease (PD) is the most common form of neurodegenerative movement disorder, associated with profound loss of dopaminergic neurons from the basal ganglia. Though loss of dopaminergic neuron cell bodies from the substantia nigra pars compacta is a well-studied feature, atrophy and loss of their axons within the nigrostriatal tract is also emerging as an early event in disease progression. Genes that drive the Wallerian degeneration, like Sterile alpha and toll/interleukin-1 receptor motif containing (Sarm1), are excellent candidates for driving this axon degeneration, given similarities in the morphology of axon degeneration after axotomy and in PD. In the present study we assessed whether Sarm1 contributes to loss of dopaminergic projections in mouse models of PD. In Sarm1 deficient mice, we observed a significant delay in the degeneration of severed dopaminergic axons distal to a 6-OHDA lesion of the medial forebrain bundle (MFB) in the nigrostriatal tract, and an accompanying rescue of morphological, biochemical and behavioural phenotypes. However, we observed no difference compared to controls when striatal terminals were lesioned with 6-OHDA to induce a dying back form of neurodegeneration. Likewise, when PD phenotypes were induced using AAV-induced alpha-synuclein overexpression, we observed similar modest loss of dopaminergic terminals in Sarm1 knockouts and controls. Our data argues that axon degeneration after MFB lesion is Sarm1-dependent, but that other models for PD do not require Sarm1, or that Sarm1 acts with other redundant genetic pathways. This work adds to a growing body of evidence indicating Sarm1 contributes to some, but not all types of neurodegeneration, and supports the notion that while axon degeneration in many context appears morphologically similar, a diversity of axon degeneration programs exist.


Subject(s)
Armadillo Domain Proteins/genetics , Axons/pathology , Cytoskeletal Proteins/genetics , Genetic Variation/physiology , Parkinsonian Disorders/genetics , Parkinsonian Disorders/pathology , Animals , Armadillo Domain Proteins/deficiency , Axons/metabolism , Cytoskeletal Proteins/deficiency , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nerve Degeneration/chemically induced , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Oxidopamine/toxicity , Parkinsonian Disorders/chemically induced
18.
Proc Natl Acad Sci U S A ; 115(32): 8209-8214, 2018 08 07.
Article in English | MEDLINE | ID: mdl-30038021

ABSTRACT

Abnormal modifications to mutant superoxide dismutase 1 (SOD1) are linked to familial amyotrophic lateral sclerosis (fALS). Misfolding of wild-type SOD1 (SOD1WT) is also observed in postmortem tissue of a subset of sporadic ALS (sALS) cases, but cellular and molecular mechanisms generating abnormal SOD1WT species are unknown. We analyzed aberrant human SOD1WT species over the lifetime of transgenic mice and found the accumulation of disulfide-cross-linked high-molecular-weight SOD1WT aggregates during aging. Subcellular fractionation of spinal cord tissue and protein overexpression in NSC-34 motoneuron-like cells revealed that endoplasmic reticulum (ER) localization favors oxidation and disulfide-dependent aggregation of SOD1WT We established a pharmacological paradigm of chronic ER stress in vivo, which recapitulated SOD1WTaggregation in young transgenic mice. These species were soluble in nondenaturing detergents and did not react with a SOD1 conformation-specific antibody. Interestingly, SOD1WT aggregation under ER stress correlated with astrocyte activation in the spinal cord of transgenic mice. Finally, the disulfide-cross-linked SOD1WT species were also found augmented in spinal cord tissue of sALS patients, correlating with the presence of ER stress markers. Overall, this study suggests that ER stress increases the susceptibility of SOD1WT to aggregate during aging, operating as a possible risk factor for developing ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Endoplasmic Reticulum Stress/physiology , Spinal Cord/pathology , Superoxide Dismutase-1/metabolism , Adult , Aged , Aged, 80 and over , Aging/pathology , Amyotrophic Lateral Sclerosis/genetics , Animals , Astrocytes/pathology , Brain/cytology , Brain/drug effects , Brain/pathology , Cell Line , Disease Models, Animal , Endoplasmic Reticulum Stress/drug effects , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Motor Neurons , Mutation , Oxidation-Reduction , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/pathology , Protein Folding , Proteostasis/physiology , Spinal Cord/cytology , Spinal Cord/drug effects , Superoxide Dismutase-1/genetics , Tryptophan/metabolism , Tunicamycin/pharmacology , Unfolded Protein Response/physiology
19.
Hum Mol Genet ; 27(21): 3761-3771, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30010873

ABSTRACT

Axon degeneration occurs in all neurodegenerative diseases, but the molecular pathways regulating axon destruction during neurodegeneration are poorly understood. Sterile Alpha and TIR Motif Containing 1 (Sarm1) is an essential component of the prodegenerative pathway driving axon degeneration after axotomy and represents an appealing target for therapeutic intervention in neurological conditions involving axon loss. Amyotrophic lateral sclerosis (ALS) is characterized by rapid, progressive motor neuron degeneration and muscle atrophy, causing paralysis and death. Patient tissue and animal models of ALS show destruction of upper and lower motor neuron cell bodies and loss of their associated axons. Here, we investigate whether loss of Sarm1 can mitigate motor neuron degeneration in the SOD1G93A mouse model of ALS. We found no change in survival, behavioral, electrophysiogical or histopathological outcomes in SOD1G93A mice null for Sarm1. Blocking Sarm1-mediated axon destruction alone is therefore not sufficient to suppress SOD1G93A-induced neurodegeneration. Our data suggest the molecular pathways driving axon loss in ALS may be Sarm1-independent or involve genetic pathways that act in a redundant fashion with Sarm1.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Armadillo Domain Proteins/metabolism , Cytoskeletal Proteins/metabolism , Motor Neurons/metabolism , Nerve Degeneration , Amyotrophic Lateral Sclerosis/pathology , Animals , Armadillo Domain Proteins/physiology , Axotomy , Cytoskeletal Proteins/physiology , Disease Models, Animal , Male , Mice , Mice, Transgenic , Superoxide Dismutase/genetics
20.
EMBO J ; 35(8): 845-65, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26869642

ABSTRACT

Disturbance of endoplasmic reticulum (ER) proteostasis is a common feature of amyotrophic lateral sclerosis (ALS). Protein disulfide isomerases (PDIs) areERfoldases identified as possibleALSbiomarkers, as well as neuroprotective factors. However, no functional studies have addressed their impact on the disease process. Here, we functionally characterized fourALS-linked mutations recently identified in two majorPDIgenes,PDIA1 andPDIA3/ERp57. Phenotypic screening in zebrafish revealed that the expression of thesePDIvariants induce motor defects associated with a disruption of motoneuron connectivity. Similarly, the expression of mutantPDIs impaired dendritic outgrowth in motoneuron cell culture models. Cellular and biochemical studies identified distinct molecular defects underlying the pathogenicity of thesePDImutants. Finally, targetingERp57 in the nervous system led to severe motor dysfunction in mice associated with a loss of neuromuscular synapses. This study identifiesERproteostasis imbalance as a risk factor forALS, driving initial stages of the disease.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Motor Neurons/pathology , Procollagen-Proline Dioxygenase/genetics , Protein Disulfide-Isomerases/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Animals, Genetically Modified , Electromyography , Embryo, Nonmammalian , Endoplasmic Reticulum Stress/genetics , Humans , Mice, Knockout , Mutation , Neurites/pathology , Procollagen-Proline Dioxygenase/metabolism , Protein Disulfide-Isomerases/metabolism , Zebrafish/embryology , Zebrafish/genetics
SELECTION OF CITATIONS
SEARCH DETAIL