Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 161
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 185(13): 2201-2203, 2022 06 23.
Article in English | MEDLINE | ID: mdl-35750028

ABSTRACT

The ε4 variant in the APOE gene is the strongest genetic risk factor for Alzheimer's disease. How does this gene impact different cell types in the brain to increase disease risk? In this issue of Cell, TCW and colleagues report APOE-driven cell-type-specific changes that may contribute to Alzheimer's disease risk.


Subject(s)
Alzheimer Disease , Apolipoprotein E4 , Apolipoproteins E/metabolism , Alzheimer Disease/genetics , Apolipoprotein E4/genetics , Brain , Humans , Risk Factors
2.
Cell ; 184(16): 4284-4298.e27, 2021 08 05.
Article in English | MEDLINE | ID: mdl-34233164

ABSTRACT

Many organisms evolved strategies to survive desiccation. Plant seeds protect dehydrated embryos from various stressors and can lay dormant for millennia. Hydration is the key trigger to initiate germination, but the mechanism by which seeds sense water remains unresolved. We identified an uncharacterized Arabidopsis thaliana prion-like protein we named FLOE1, which phase separates upon hydration and allows the embryo to sense water stress. We demonstrate that biophysical states of FLOE1 condensates modulate its biological function in vivo in suppressing seed germination under unfavorable environments. We find intragenic, intraspecific, and interspecific natural variation in FLOE1 expression and phase separation and show that intragenic variation is associated with adaptive germination strategies in natural populations. This combination of molecular, organismal, and ecological studies uncovers FLOE1 as a tunable environmental sensor with direct implications for the design of drought-resistant crops, in the face of climate change.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/growth & development , Germination , Intercellular Signaling Peptides and Proteins/metabolism , Prions/metabolism , Seeds/growth & development , Water/metabolism , Arabidopsis/genetics , Arabidopsis/ultrastructure , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/ultrastructure , Dehydration , Imaging, Three-Dimensional , Intercellular Signaling Peptides and Proteins/chemistry , Mutation/genetics , Plant Dormancy , Plants, Genetically Modified , Protein Domains , Protein Isoforms/metabolism , Seeds/ultrastructure
3.
Cell ; 184(3): 689-708.e20, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33482083

ABSTRACT

The most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) is a GGGGCC repeat expansion in the C9orf72 gene. We developed a platform to interrogate the chromatin accessibility landscape and transcriptional program within neurons during degeneration. We provide evidence that neurons expressing the dipeptide repeat protein poly(proline-arginine), translated from the C9orf72 repeat expansion, activate a highly specific transcriptional program, exemplified by a single transcription factor, p53. Ablating p53 in mice completely rescued neurons from degeneration and markedly increased survival in a C9orf72 mouse model. p53 reduction also rescued axonal degeneration caused by poly(glycine-arginine), increased survival of C9orf72 ALS/FTD-patient-induced pluripotent stem cell (iPSC)-derived motor neurons, and mitigated neurodegeneration in a C9orf72 fly model. We show that p53 activates a downstream transcriptional program, including Puma, which drives neurodegeneration. These data demonstrate a neurodegenerative mechanism dynamically regulated through transcription-factor-binding events and provide a framework to apply chromatin accessibility and transcription program profiles to neurodegeneration.


Subject(s)
C9orf72 Protein/metabolism , DNA Repeat Expansion/genetics , Nerve Degeneration/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Axons/metabolism , C9orf72 Protein/genetics , Cell Death , Cells, Cultured , Cerebral Cortex/pathology , Chromatin/metabolism , DNA Damage , Disease Models, Animal , Drosophila , Mice, Inbred C57BL , Nerve Degeneration/pathology , Protein Stability , Transcription, Genetic , Tumor Suppressor Proteins/metabolism
4.
Cell ; 173(4): 958-971.e17, 2018 05 03.
Article in English | MEDLINE | ID: mdl-29628143

ABSTRACT

Defects in nucleocytoplasmic transport have been identified as a key pathogenic event in amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) mediated by a GGGGCC hexanucleotide repeat expansion in C9ORF72, the most common genetic cause of ALS/FTD. Furthermore, nucleocytoplasmic transport disruption has also been implicated in other neurodegenerative diseases with protein aggregation, suggesting a shared mechanism by which protein stress disrupts nucleocytoplasmic transport. Here, we show that cellular stress disrupts nucleocytoplasmic transport by localizing critical nucleocytoplasmic transport factors into stress granules, RNA/protein complexes that play a crucial role in ALS pathogenesis. Importantly, inhibiting stress granule assembly, such as by knocking down Ataxin-2, suppresses nucleocytoplasmic transport defects as well as neurodegeneration in C9ORF72-mediated ALS/FTD. Our findings identify a link between stress granule assembly and nucleocytoplasmic transport, two fundamental cellular processes implicated in the pathogenesis of C9ORF72-mediated ALS/FTD and other neurodegenerative diseases.


Subject(s)
Active Transport, Cell Nucleus/physiology , Amyotrophic Lateral Sclerosis/pathology , Ataxin-2/metabolism , C9orf72 Protein/genetics , Frontotemporal Dementia/pathology , Active Transport, Cell Nucleus/drug effects , Aged , Amyotrophic Lateral Sclerosis/metabolism , Arsenites/toxicity , Ataxin-2/antagonists & inhibitors , Ataxin-2/genetics , C9orf72 Protein/metabolism , DNA Repeat Expansion/genetics , Female , Frontotemporal Dementia/metabolism , HEK293 Cells , Humans , Male , Membrane Glycoproteins/metabolism , Middle Aged , Nuclear Pore Complex Proteins/metabolism , Oxidative Stress/drug effects , RNA Interference , RNA, Small Interfering/metabolism , Sodium Compounds/toxicity , alpha Karyopherins/antagonists & inhibitors , alpha Karyopherins/genetics , alpha Karyopherins/metabolism , beta Karyopherins/antagonists & inhibitors , beta Karyopherins/genetics , beta Karyopherins/metabolism , ran GTP-Binding Protein/antagonists & inhibitors , ran GTP-Binding Protein/genetics , ran GTP-Binding Protein/metabolism
5.
Cell ; 173(3): 677-692.e20, 2018 04 19.
Article in English | MEDLINE | ID: mdl-29677512

ABSTRACT

RNA-binding proteins (RBPs) with prion-like domains (PrLDs) phase transition to functional liquids, which can mature into aberrant hydrogels composed of pathological fibrils that underpin fatal neurodegenerative disorders. Several nuclear RBPs with PrLDs, including TDP-43, FUS, hnRNPA1, and hnRNPA2, mislocalize to cytoplasmic inclusions in neurodegenerative disorders, and mutations in their PrLDs can accelerate fibrillization and cause disease. Here, we establish that nuclear-import receptors (NIRs) specifically chaperone and potently disaggregate wild-type and disease-linked RBPs bearing a NLS. Karyopherin-ß2 (also called Transportin-1) engages PY-NLSs to inhibit and reverse FUS, TAF15, EWSR1, hnRNPA1, and hnRNPA2 fibrillization, whereas Importin-α plus Karyopherin-ß1 prevent and reverse TDP-43 fibrillization. Remarkably, Karyopherin-ß2 dissolves phase-separated liquids and aberrant fibrillar hydrogels formed by FUS and hnRNPA1. In vivo, Karyopherin-ß2 prevents RBPs with PY-NLSs accumulating in stress granules, restores nuclear RBP localization and function, and rescues degeneration caused by disease-linked FUS and hnRNPA2. Thus, NIRs therapeutically restore RBP homeostasis and mitigate neurodegeneration.


Subject(s)
Active Transport, Cell Nucleus , Prions/chemistry , RNA-Binding Proteins/chemistry , Receptors, Cytoplasmic and Nuclear/chemistry , Adult , Aged , Animals , Cytoplasm/chemistry , DNA-Binding Proteins/chemistry , Drosophila melanogaster , Female , Green Fluorescent Proteins/chemistry , HEK293 Cells , HeLa Cells , Homeostasis , Humans , Karyopherins/chemistry , Male , Middle Aged , Molecular Chaperones/chemistry , Mutation , Neurodegenerative Diseases/pathology , Protein Domains , RNA-Binding Protein EWS/chemistry , TATA-Binding Protein Associated Factors/chemistry , beta Karyopherins/chemistry
6.
Mol Cell ; 83(12): 2020-2034.e6, 2023 Jun 15.
Article in English | MEDLINE | ID: mdl-37295429

ABSTRACT

Biomolecular condensation underlies the biogenesis of an expanding array of membraneless assemblies, including stress granules (SGs), which form under a variety of cellular stresses. Advances have been made in understanding the molecular grammar of a few scaffold proteins that make up these phases, but how the partitioning of hundreds of SG proteins is regulated remains largely unresolved. While investigating the rules that govern the condensation of ataxin-2, an SG protein implicated in neurodegenerative disease, we unexpectedly identified a short 14 aa sequence that acts as a condensation switch and is conserved across the eukaryote lineage. We identify poly(A)-binding proteins as unconventional RNA-dependent chaperones that control this regulatory switch. Our results uncover a hierarchy of cis and trans interactions that fine-tune ataxin-2 condensation and reveal an unexpected molecular function for ancient poly(A)-binding proteins as regulators of biomolecular condensate proteins. These findings may inspire approaches to therapeutically target aberrant phases in disease.


Subject(s)
Ataxin-2 , Neurodegenerative Diseases , Humans , Ataxin-2/genetics , Poly(A)-Binding Protein I , Neurodegenerative Diseases/metabolism , Biomolecular Condensates
7.
Mol Cell ; 82(22): 4194-4196, 2022 11 17.
Article in English | MEDLINE | ID: mdl-36400005

ABSTRACT

In this issue of Molecular Cell, Gropp et al. identify how the cellular background affects whether or not the Huntington's disease protein will form toxic pathological aggregates, providing insight into selective neuronal vulnerability in neurodegenerative disease.


Subject(s)
Huntington Disease , Neurodegenerative Diseases , Humans , Huntington Disease/pathology , Neurons/metabolism
8.
Cell ; 154(6): 1182-4, 2013 Sep 12.
Article in English | MEDLINE | ID: mdl-24034239

ABSTRACT

Alzheimer's disease (AD) is associated with the deposition of ß-amyloid (Aß) plaques in the brain. In this issue, by cleverly processing patient samples, Lu et al. define a novel structural model of Aß fibrils from AD brain, revealing surprising differences from in vitro fibrils. These findings may lead to structure-specific inhibitors and more selective amyloid-imaging methods.


Subject(s)
Alzheimer Disease/pathology , Amyloid/chemistry , Brain/pathology , Female , Humans
9.
Nature ; 603(7899): 124-130, 2022 03.
Article in English | MEDLINE | ID: mdl-35197626

ABSTRACT

A hallmark pathological feature of the neurodegenerative diseases amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) is the depletion of RNA-binding protein TDP-43 from the nucleus of neurons in the brain and spinal cord1. A major function of TDP-43 is as a repressor of cryptic exon inclusion during RNA splicing2-4. Single nucleotide polymorphisms in UNC13A are among the strongest hits associated with FTD and ALS in human genome-wide association studies5,6, but how those variants increase risk for disease is unknown. Here we show that TDP-43 represses a cryptic exon-splicing event in UNC13A. Loss of TDP-43 from the nucleus in human brain, neuronal cell lines and motor neurons derived from induced pluripotent stem cells resulted in the inclusion of a cryptic exon in UNC13A mRNA and reduced UNC13A protein expression. The top variants associated with FTD or ALS risk in humans are located in the intron harbouring the cryptic exon, and we show that they increase UNC13A cryptic exon splicing in the face of TDP-43 dysfunction. Together, our data provide a direct functional link between one of the strongest genetic risk factors for FTD and ALS (UNC13A genetic variants), and loss of TDP-43 function.


Subject(s)
Amyotrophic Lateral Sclerosis , Frontotemporal Dementia , Amyotrophic Lateral Sclerosis/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Exons/genetics , Frontotemporal Dementia/metabolism , Genome-Wide Association Study , Humans , Motor Neurons/pathology , Nerve Tissue Proteins
10.
Mol Cell ; 78(2): 193-194, 2020 04 16.
Article in English | MEDLINE | ID: mdl-32302539

ABSTRACT

Heterochromatin protein 1 (HP1) has been proposed to drive heterochromatin formation by liquid-liquid phase separation. In this issue of Molecular Cell, however, Erdel et al. establish that heterochromatin can adopt digital compaction states that are independent of HP1 phase separation.


Subject(s)
Chromosomal Proteins, Non-Histone , Heterochromatin , Animals , Chromatin Assembly and Disassembly , Chromobox Protein Homolog 5 , DNA , Mice
11.
Nature ; 599(7883): 102-107, 2021 11.
Article in English | MEDLINE | ID: mdl-34616039

ABSTRACT

Astrocytes regulate the response of the central nervous system to disease and injury and have been hypothesized to actively kill neurons in neurodegenerative disease1-6. Here we report an approach to isolate one component of the long-sought astrocyte-derived toxic factor5,6. Notably, instead of a protein, saturated lipids contained in APOE and APOJ lipoparticles mediate astrocyte-induced toxicity. Eliminating the formation of long-chain saturated lipids by astrocyte-specific knockout of the saturated lipid synthesis enzyme ELOVL1 mitigates astrocyte-mediated toxicity in vitro as well as in a model of acute axonal injury in vivo. These results suggest a mechanism by which astrocytes kill cells in the central nervous system.


Subject(s)
Astrocytes/chemistry , Astrocytes/metabolism , Cell Death/drug effects , Lipids/chemistry , Lipids/toxicity , Animals , Culture Media, Conditioned/chemistry , Culture Media, Conditioned/toxicity , Fatty Acid Elongases/deficiency , Fatty Acid Elongases/genetics , Fatty Acid Elongases/metabolism , Female , Gene Knockout Techniques , Male , Mice , Mice, Knockout , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neurotoxins/chemistry , Neurotoxins/toxicity
12.
Trends Biochem Sci ; 47(1): 1-2, 2022 01.
Article in English | MEDLINE | ID: mdl-34654581

ABSTRACT

A recent study by Huang et al. unexpectedly uncovered that DAXX moonlights as a booster of protein folding, including counteracting aggregation of tumor suppressor p53. Since p53 aggregation is a common hallmark of cancer, this finding provides a potential pathway to therapeutically reactivate p53 signaling and halt tumor progression.


Subject(s)
Neoplasms , Nuclear Proteins , Co-Repressor Proteins/metabolism , Humans , Molecular Chaperones/metabolism , Nuclear Proteins/metabolism , Protein Folding , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
13.
Mol Cell ; 71(5): 649-651, 2018 09 06.
Article in English | MEDLINE | ID: mdl-30193092

ABSTRACT

In this issue of Molecular Cell, McGurk et al. (2018) identify how poly(ADP-ribose) binding tunes the phase behavior of the ALS disease protein TDP-43, uncovering the molecular events underlying its aggregation in disease and illuminating a novel therapeutic target.


Subject(s)
Amyotrophic Lateral Sclerosis , Poly Adenosine Diphosphate Ribose , DNA-Binding Proteins , Humans , Sugars
14.
Trends Genet ; 38(9): 904-919, 2022 09.
Article in English | MEDLINE | ID: mdl-35487823

ABSTRACT

Motor neurons are a remarkably powerful cell type in the central nervous system. They innervate and control the contraction of virtually every muscle in the body and their dysfunction underlies numerous neuromuscular diseases. Some motor neurons seem resistant to degeneration whereas others are vulnerable. The intrinsic heterogeneity of motor neurons in adult organisms has remained elusive. The development of high-throughput single-cell transcriptomics has changed the paradigm, empowering rapid isolation and profiling of motor neuron nuclei, revealing remarkable transcriptional diversity within the skeletal and autonomic nervous systems. Here, we discuss emerging technologies for defining motor neuron heterogeneity in the adult motor system as well as implications for disease and spinal cord injury. We establish a roadmap for future applications of emerging techniques - such as epigenetic profiling, spatial RNA sequencing, and single-cell somatic mutational profiling to adult motor neurons, which will revolutionize our understanding of the healthy and degenerating adult motor system.


Subject(s)
Amyotrophic Lateral Sclerosis , Adult , Amyotrophic Lateral Sclerosis/genetics , Animals , Disease Models, Animal , Humans , Motor Neurons/metabolism , Spinal Cord , Transcriptome/genetics
15.
Brain ; 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38940350

ABSTRACT

In frontotemporal lobar degeneration (FTLD), pathological protein aggregation in specific brain regions is associated with declines in human-specialized social-emotional and language functions. In most patients, disease protein aggregates contain either TDP-43 (FTLD-TDP) or tau (FTLD-tau). Here, we explored whether FTLD-associated regional degeneration patterns relate to regional gene expression of human accelerated regions (HARs), conserved sequences that have undergone positive selection during recent human evolution. To this end, we used structural neuroimaging from patients with FTLD and human brain regional transcriptomic data from controls to identify genes expressed in FTLD-targeted brain regions. We then integrated primate comparative genomic data to test our hypothesis that FTLD targets brain regions linked to expression levels of recently evolved genes. In addition, we asked whether genes whose expression correlates with FTLD atrophy are enriched for genes that undergo cryptic splicing when TDP-43 function is impaired. We found that FTLD-TDP and FTLD-tau subtypes target brain regions with overlapping and distinct gene expression correlates, highlighting many genes linked to neuromodulatory functions. FTLD atrophy-correlated genes were strongly enriched for HARs. Atrophy-correlated genes in FTLD-TDP showed greater overlap with TDP-43 cryptic splicing genes and genes with more numerous TDP-43 binding sites compared with atrophy-correlated genes in FTLD-tau. Cryptic splicing genes were enriched for HAR genes, and vice versa, but this effect was due to the confounding influence of gene length. Analyses performed at the individual-patient level revealed that the expression of HAR genes and cryptically spliced genes within putative regions of disease onset differed across FTLD-TDP subtypes.

16.
J Neurosci ; 43(2): 333-345, 2023 01 11.
Article in English | MEDLINE | ID: mdl-36446586

ABSTRACT

Hexanucleotide repeat expansion (HRE) within C9orf72 is the most common genetic cause of frontotemporal dementia (FTD). Thalamic atrophy occurs in both sporadic and familial FTD but is thought to distinctly affect HRE carriers. Separately, emerging evidence suggests widespread derepression of transposable elements (TEs) in the brain in several neurodegenerative diseases, including C9orf72 HRE-mediated FTD (C9-FTD). Whether TE activation can be measured in peripheral blood and how the reduction in peripheral C9orf72 expression observed in HRE carriers relates to atrophy and clinical impairment remain unknown. We used FreeSurfer software to assess the effects of C9orf72 HRE and clinical diagnosis (n = 78 individuals, male and female) on atrophy of thalamic nuclei. We also generated a novel, human, whole-blood RNA-sequencing dataset to determine the relationships among peripheral C9orf72 expression, TE activation, thalamic atrophy, and clinical severity (n = 114 individuals, male and female). We confirmed global thalamic atrophy and reduced C9orf72 expression in HRE carriers. Moreover, we identified disproportionate atrophy of the right mediodorsal lateral nucleus in HRE carriers and showed that C9orf72 expression associated with clinical severity, independent of thalamic atrophy. Strikingly, we found global peripheral activation of TEs, including the human endogenous LINE-1 element L1HS L1HS levels were associated with atrophy of multiple pulvinar nuclei, a thalamic region implicated in C9-FTD. Integration of peripheral transcriptomic and neuroimaging data from human HRE carriers revealed atrophy of specific thalamic nuclei, demonstrated that C9orf72 levels relate to clinical severity, and identified marked derepression of TEs, including L1HS, which predicted atrophy of FTD-relevant thalamic nuclei.SIGNIFICANCE STATEMENT Pathogenic repeat expansion in C9orf72 is the most frequent genetic cause of FTD and amyotrophic lateral sclerosis (ALS; C9-FTD/ALS). The clinical, neuroimaging, and pathologic features of C9-FTD/ALS are well characterized, whereas the intersections of transcriptomic dysregulation and brain structure remain largely unexplored. Herein, we used a novel radiogenomic approach to examine the relationship between peripheral blood transcriptomics and thalamic atrophy, a neuroimaging feature disproportionately impacted in C9-FTD/ALS. We confirmed reduction of C9orf72 in blood and found broad dysregulation of transposable elements-genetic elements typically repressed in the human genome-in symptomatic C9orf72 expansion carriers, which associated with atrophy of thalamic nuclei relevant to FTD. C9orf72 expression was also associated with clinical severity, suggesting that peripheral C9orf72 levels capture disease-relevant information.


Subject(s)
Amyotrophic Lateral Sclerosis , Frontotemporal Dementia , Humans , Male , Female , Amyotrophic Lateral Sclerosis/genetics , Frontotemporal Dementia/diagnostic imaging , Frontotemporal Dementia/genetics , Frontotemporal Dementia/pathology , C9orf72 Protein/genetics , DNA Transposable Elements , Atrophy
17.
Am J Hum Genet ; 107(3): 445-460, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32750315

ABSTRACT

Tandem repeats are proposed to contribute to human-specific traits, and more than 40 tandem repeat expansions are known to cause neurological disease. Here, we characterize a human-specific 69 bp variable number tandem repeat (VNTR) in the last intron of WDR7, which exhibits striking variability in both copy number and nucleotide composition, as revealed by long-read sequencing. In addition, greater repeat copy number is significantly enriched in three independent cohorts of individuals with sporadic amyotrophic lateral sclerosis (ALS). Each unit of the repeat forms a stem-loop structure with the potential to produce microRNAs, and the repeat RNA can aggregate when expressed in cells. We leveraged its remarkable sequence variability to align the repeat in 288 samples and uncover its mechanism of expansion. We found that the repeat expands in the 3'-5' direction, in groups of repeat units divisible by two. The expansion patterns we observed were consistent with duplication events, and a replication error called template switching. We also observed that the VNTR is expanded in both Denisovan and Neanderthal genomes but is fixed at one copy or fewer in non-human primates. Evaluating the repeat in 1000 Genomes Project samples reveals that some repeat segments are solely present or absent in certain geographic populations. The large size of the repeat unit in this VNTR, along with our multiplexed sequencing strategy, provides an unprecedented opportunity to study mechanisms of repeat expansion, and a framework for evaluating the roles of VNTRs in human evolution and disease.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Amyotrophic Lateral Sclerosis/genetics , Evolution, Molecular , Tandem Repeat Sequences/genetics , Aged , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyotrophic Lateral Sclerosis/pathology , DNA Repeat Expansion/genetics , Female , Gene Expression Regulation/genetics , Humans , Male , Minisatellite Repeats/genetics , Phenotype , Species Specificity
18.
Nature ; 544(7650): 367-371, 2017 04 20.
Article in English | MEDLINE | ID: mdl-28405022

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a rapidly progressing neurodegenerative disease that is characterized by motor neuron loss and that leads to paralysis and death 2-5 years after disease onset. Nearly all patients with ALS have aggregates of the RNA-binding protein TDP-43 in their brains and spinal cords, and rare mutations in the gene encoding TDP-43 can cause ALS. There are no effective TDP-43-directed therapies for ALS or related TDP-43 proteinopathies, such as frontotemporal dementia. Antisense oligonucleotides (ASOs) and RNA-interference approaches are emerging as attractive therapeutic strategies in neurological diseases. Indeed, treatment of a rat model of inherited ALS (caused by a mutation in Sod1) with ASOs against Sod1 has been shown to substantially slow disease progression. However, as SOD1 mutations account for only around 2-5% of ALS cases, additional therapeutic strategies are needed. Silencing TDP-43 itself is probably not appropriate, given its critical cellular functions. Here we present a promising alternative therapeutic strategy for ALS that involves targeting ataxin-2. A decrease in ataxin-2 suppresses TDP-43 toxicity in yeast and flies, and intermediate-length polyglutamine expansions in the ataxin-2 gene increase risk of ALS. We used two independent approaches to test whether decreasing ataxin-2 levels could mitigate disease in a mouse model of TDP-43 proteinopathy. First, we crossed ataxin-2 knockout mice with TDP-43 (also known as TARDBP) transgenic mice. The decrease in ataxin-2 reduced aggregation of TDP-43, markedly increased survival and improved motor function. Second, in a more therapeutically applicable approach, we administered ASOs targeting ataxin-2 to the central nervous system of TDP-43 transgenic mice. This single treatment markedly extended survival. Because TDP-43 aggregation is a component of nearly all cases of ALS, targeting ataxin-2 could represent a broadly effective therapeutic strategy.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/therapy , Ataxin-2/deficiency , DNA-Binding Proteins/metabolism , Longevity , Oligonucleotides, Antisense/therapeutic use , Protein Aggregation, Pathological/therapy , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Ataxin-2/genetics , Central Nervous System/metabolism , Cytoplasmic Granules/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Disease Progression , Female , Gene Knockdown Techniques , Humans , Male , Mice , Mice, Knockout , Mice, Transgenic , Motor Skills/physiology , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/genetics , Protein Aggregation, Pathological/genetics , Stress, Physiological , Survival Analysis
19.
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
20.
Nature ; 539(7628): 207-216, 2016 11 10.
Article in English | MEDLINE | ID: mdl-27830778

ABSTRACT

Parkinson's disease is a debilitating, age-associated movement disorder. A central aspect of the pathophysiology of Parkinson's disease is the progressive demise of midbrain dopamine neurons and their axonal projections, but the underlying causes of this loss are unclear. Advances in genetics and experimental model systems have illuminated an important role for defects in intracellular transport pathways to lysosomes. The accumulation of altered proteins and damaged mitochondria, particularly at axon terminals, ultimately might overwhelm the capacity of intracellular disposal mechanisms. Cell-extrinsic mechanisms, including inflammation and prion-like spreading, are proposed to have both protective and deleterious functions in Parkinson's disease.


Subject(s)
Parkinson Disease , Animals , Biological Transport/genetics , Endocytosis , Glucosylceramidase/genetics , Glucosylceramidase/metabolism , Humans , Inflammation/metabolism , Inflammation/pathology , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Lysosomes/metabolism , Models, Biological , Molecular Targeted Therapy , Neurons/metabolism , Neurons/pathology , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology , Parkinson Disease/therapy , Prions/metabolism , Synaptic Vesicles/metabolism , alpha-Synuclein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL