Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Nat Neurosci ; 25(4): 433-445, 2022 04.
Article in English | MEDLINE | ID: mdl-35361972

ABSTRACT

The noncoding genome is substantially larger than the protein-coding genome but has been largely unexplored by genetic association studies. Here, we performed region-based rare variant association analysis of >25,000 variants in untranslated regions of 6,139 amyotrophic lateral sclerosis (ALS) whole genomes and the whole genomes of 70,403 non-ALS controls. We identified interleukin-18 receptor accessory protein (IL18RAP) 3' untranslated region (3'UTR) variants as significantly enriched in non-ALS genomes and associated with a fivefold reduced risk of developing ALS, and this was replicated in an independent cohort. These variants in the IL18RAP 3'UTR reduce mRNA stability and the binding of double-stranded RNA (dsRNA)-binding proteins. Finally, the variants of the IL18RAP 3'UTR confer a survival advantage for motor neurons because they dampen neurotoxicity of human induced pluripotent stem cell (iPSC)-derived microglia bearing an ALS-associated expansion in C9orf72, and this depends on NF-κB signaling. This study reveals genetic variants that protect against ALS by reducing neuroinflammation and emphasizes the importance of noncoding genetic association studies.


Subject(s)
Amyotrophic Lateral Sclerosis , Induced Pluripotent Stem Cells , Interleukin-18 Receptor beta Subunit/genetics , 3' Untranslated Regions/genetics , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Interleukin-18 Receptor beta Subunit/metabolism , Motor Neurons/metabolism
2.
Neuron ; 110(6): 992-1008.e11, 2022 03 16.
Article in English | MEDLINE | ID: mdl-35045337

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a complex disease that leads to motor neuron death. Despite heritability estimates of 52%, genome-wide association studies (GWASs) have discovered relatively few loci. We developed a machine learning approach called RefMap, which integrates functional genomics with GWAS summary statistics for gene discovery. With transcriptomic and epigenetic profiling of motor neurons derived from induced pluripotent stem cells (iPSCs), RefMap identified 690 ALS-associated genes that represent a 5-fold increase in recovered heritability. Extensive conservation, transcriptome, network, and rare variant analyses demonstrated the functional significance of candidate genes in healthy and diseased motor neurons and brain tissues. Genetic convergence between common and rare variation highlighted KANK1 as a new ALS gene. Reproducing KANK1 patient mutations in human neurons led to neurotoxicity and demonstrated that TDP-43 mislocalization, a hallmark pathology of ALS, is downstream of axonal dysfunction. RefMap can be readily applied to other complex diseases.


Subject(s)
Amyotrophic Lateral Sclerosis , Induced Pluripotent Stem Cells , Adaptor Proteins, Signal Transducing/genetics , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Cell Death/genetics , Cytoskeletal Proteins/genetics , Genome-Wide Association Study , Humans , Induced Pluripotent Stem Cells/pathology , Motor Neurons/pathology
4.
Cell Rep ; 33(9): 108456, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33264630

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is an incurable neurodegenerative disease. CAV1 and CAV2 organize membrane lipid rafts (MLRs) important for cell signaling and neuronal survival, and overexpression of CAV1 ameliorates ALS phenotypes in vivo. Genome-wide association studies localize a large proportion of ALS risk variants within the non-coding genome, but further characterization has been limited by lack of appropriate tools. By designing and applying a pipeline to identify pathogenic genetic variation within enhancer elements responsible for regulating gene expression, we identify disease-associated variation within CAV1/CAV2 enhancers, which replicate in an independent cohort. Discovered enhancer mutations reduce CAV1/CAV2 expression and disrupt MLRs in patient-derived cells, and CRISPR-Cas9 perturbation proximate to a patient mutation is sufficient to reduce CAV1/CAV2 expression in neurons. Additional enrichment of ALS-associated mutations within CAV1 exons positions CAV1 as an ALS risk gene. We propose CAV1/CAV2 overexpression as a personalized medicine target for ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Caveolin 1/genetics , Animals , Caveolin 1/metabolism , Genetic Predisposition to Disease , Genetic Variation , Genome , Humans
5.
Mol Cell ; 80(5): 876-891.e6, 2020 12 03.
Article in English | MEDLINE | ID: mdl-33217318

ABSTRACT

Stress granules (SGs) are cytoplasmic assemblies of proteins and non-translating mRNAs. Whereas much has been learned about SG formation, a major gap remains in understanding the compositional changes SGs undergo during normal disassembly and under disease conditions. Here, we address this gap by proteomic dissection of the SG temporal disassembly sequence using multi-bait APEX proximity proteomics. We discover 109 novel SG proteins and characterize distinct SG substructures. We reveal dozens of disassembly-engaged proteins (DEPs), some of which play functional roles in SG disassembly, including small ubiquitin-like modifier (SUMO) conjugating enzymes. We further demonstrate that SUMOylation regulates SG disassembly and SG formation. Parallel proteomics with amyotrophic lateral sclerosis (ALS)-associated C9ORF72 dipeptides uncovered attenuated DEP recruitment during SG disassembly and impaired SUMOylation. Accordingly, SUMO activity ameliorated C9ORF72-ALS-related neurodegeneration in Drosophila. By dissecting the SG spatiotemporal proteomic landscape, we provide an in-depth resource for future work on SG function and reveal basic and disease-relevant mechanisms of SG disassembly.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , C9orf72 Protein/metabolism , Cytoplasmic Granules/metabolism , Drosophila Proteins/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Sumoylation , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , C9orf72 Protein/genetics , Cell Line, Tumor , Cytoplasmic Granules/genetics , Cytoplasmic Granules/pathology , Dipeptides/genetics , Dipeptides/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster , Humans , Mice , Proteomics , Small Ubiquitin-Related Modifier Proteins/genetics
6.
Sci Transl Med ; 11(523)2019 12 18.
Article in English | MEDLINE | ID: mdl-31852800

ABSTRACT

Motor neuron-specific microRNA-218 (miR-218) has recently received attention because of its roles in mouse development. However, miR-218 relevance to human motor neuron disease was not yet explored. Here, we demonstrate by neuropathology that miR-218 is abundant in healthy human motor neurons. However, in amyotrophic lateral sclerosis (ALS) motor neurons, miR-218 is down-regulated and its mRNA targets are reciprocally up-regulated (derepressed). We further identify the potassium channel Kv10.1 as a new miR-218 direct target that controls neuronal activity. In addition, we screened thousands of ALS genomes and identified six rare variants in the human miR-218-2 sequence. miR-218 gene variants fail to regulate neuron activity, suggesting the importance of this small endogenous RNA for neuronal robustness. The underlying mechanisms involve inhibition of miR-218 biogenesis and reduced processing by DICER. Therefore, miR-218 activity in motor neurons may be susceptible to failure in human ALS, suggesting that miR-218 may be a potential therapeutic target in motor neuron disease.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , MicroRNAs/metabolism , Neuropathology/methods , Amyotrophic Lateral Sclerosis/genetics , Animals , Ether-A-Go-Go Potassium Channels/genetics , Ether-A-Go-Go Potassium Channels/metabolism , Humans , Mice , MicroRNAs/genetics , Motor Neurons/metabolism , Neurons/metabolism
7.
Brain Res ; 1647: 105-111, 2016 09 15.
Article in English | MEDLINE | ID: mdl-26778173

ABSTRACT

The genetics of the neurodegenerative diseases amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) turn our attention to RNA metabolism, primarily because many of the identified diseases-associated genes encode for RNA-binding proteins. microRNAs (miRNAs) are endogenous noncoding RNAs that play critical roles in maintaining brain integrity. The current review sheds light on miRNA dysregulation in neurodegenerative diseases, focusing on FTD-ALS. We propose that miRNAs are susceptible to fail when protein factors that are critical for miRNA biogenesis malfunction. Accordingly, potential insufficiencies of the 'microprocessor' complex, the nucleo-cytoplasmic export of miRNA precursors or their processing by Dicer were recently reported. Furthermore, specific miRNAs are involved in the regulation of pathways that are essential for neuronal survival or function. Any change in the expression of these specific miRNAs or in their ability to recognize their target sequences will have negative consequences. Taken together, recent reports strengthens the hypothesis that dysregulation of miRNAs might play an important role in the pathogenesis of neurodegenerative diseases, and highlights the miRNA biogenesis machinery as an interesting target for therapeutic interventions for ALS as well as FTD. This article is part of a Special Issue entitled SI:RNA Metabolism in Disease.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Frontotemporal Dementia/metabolism , MicroRNAs/biosynthesis , Amyotrophic Lateral Sclerosis/genetics , Animals , DEAD-box RNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Humans , MicroRNAs/genetics , Nuclear Pore/metabolism , RNA Processing, Post-Transcriptional , RNA-Binding Protein FUS/metabolism , Ribonuclease III/metabolism
8.
EMBO J ; 34(21): 2633-51, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26330466

ABSTRACT

Interest in RNA dysfunction in amyotrophic lateral sclerosis (ALS) recently aroused upon discovering causative mutations in RNA-binding protein genes. Here, we show that extensive down-regulation of miRNA levels is a common molecular denominator for multiple forms of human ALS. We further demonstrate that pathogenic ALS-causing mutations are sufficient to inhibit miRNA biogenesis at the Dicing step. Abnormalities of the stress response are involved in the pathogenesis of neurodegeneration, including ALS. Accordingly, we describe a novel mechanism for modulating microRNA biogenesis under stress, involving stress granule formation and re-organization of DICER and AGO2 protein interactions with their partners. In line with this observation, enhancing DICER activity by a small molecule, enoxacin, is beneficial for neuromuscular function in two independent ALS mouse models. Characterizing miRNA biogenesis downstream of the stress response ties seemingly disparate pathways in neurodegeneration and further suggests that DICER and miRNAs affect neuronal integrity and are possible therapeutic targets.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , MicroRNAs/biosynthesis , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/pathology , Animals , Base Sequence , Cytoplasmic Granules/metabolism , DEAD-box RNA Helicases/metabolism , Down-Regulation , Drug Evaluation, Preclinical , Enoxacin/pharmacology , Female , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/genetics , Motor Neurons/metabolism , RNA Interference , RNA Processing, Post-Transcriptional , Ribonuclease III/metabolism , Stress, Physiological , Superoxide Dismutase/genetics , Superoxide Dismutase-1
9.
EMBO J ; 34(9): 1143-4, 2015 May 05.
Article in English | MEDLINE | ID: mdl-25736376
10.
J Neurosci ; 34(45): 15070-82, 2014 11 05.
Article in English | MEDLINE | ID: mdl-25378171

ABSTRACT

Activation of the stress response in the presence of diverse challenges requires numerous adaptive molecular and cellular changes. To identify specific microRNA molecules that are altered following chronic stress, mice were subjected to the chronic social defeat procedure. The amygdala from these mice was collected and a screen for microRNAs that were recruited to the RNA-induced silencing complex and differentially expressed between the stressed and unstressed mice was conducted. One of the microRNAs that were significantly altered was microRNA-19b (miR-19b). Bioinformatics analysis revealed the adrenergic receptor ß-1 (Adrb1) as a potential target for this microRNA with multiple conserved seed sites. Consistent with its putative regulation by miR-19b, Adrb1 levels were reduced in the basolateral amygdala (BLA) following chronic stress. In vitro studies using luciferase assays showed a direct effect of miR-19b on Adrb1 levels, which were not evident when miR-19b seed sequences at the Adrb1 transcript were mutated. To assess the role of miR-19b in memory stabilization, previously attributed to BLA-Adrb1, we constructed lentiviruses designed to overexpress or knockdown miR-19b. Interestingly, adult mice injected bilaterally with miR-19b into the BLA showed lower freezing time relative to control in the cue fear conditioning test, and deregulation of noradrenergic circuits, consistent with downregulation of Adrb1 levels. Knockdown of endogenous BLA-miR-19b levels resulted in opposite behavioral and noradrenergic profile with higher freezing time and increase 3-methoxy-4-hydroxyphenylglycol/noradrenaline ratio. These findings suggest a key role for miR-19b in modulating behavioral responses to chronic stress and Adrb1 as an important target of miR-19b in stress-linked brain regions.


Subject(s)
Amygdala/metabolism , Argonaute Proteins/metabolism , MicroRNAs/metabolism , Receptors, Adrenergic, beta-1/metabolism , Stress, Psychological/metabolism , Amygdala/physiopathology , Animals , Argonaute Proteins/genetics , Conditioning, Classical , Freezing Reaction, Cataleptic , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , MicroRNAs/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Adrenergic, beta-1/genetics , Stress, Psychological/physiopathology
11.
J Neurosci ; 34(31): 10141-55, 2014 Jul 30.
Article in English | MEDLINE | ID: mdl-25080578

ABSTRACT

Monocyte-derived macrophages (mo-MΦs) and T cells have been shown to contribute to spinal cord repair. Recently, the remote brain choroid plexus epithelium (CP) was identified as a portal for monocyte recruitment, and its activation for leukocyte trafficking was found to be IFN-γ-dependent. Here, we addressed how the need for effector T cells can be reconciled with the role of inflammation-resolving immune cells in the repair process. Using an acute spinal cord injury model, we show that in mice deficient in IFN-γ-producing T cells, the CP was not activated, and recruitment of inflammation-resolving mo-MΦ to the spinal cord parenchyma was limited. We further demonstrate that mo-MΦ locally regulated recruitment of thymic-derived Foxp3(+) regulatory T (Treg) cells to the injured spinal cord parenchyma at the subacute/chronic phase. Importantly, an ablation protocol that resulted in reduced Tregs at this stage interfered with tissue remodeling, in contrast to Treg transient ablation, restricted to the 4 d period before the injury, which favored repair. The enhanced functional recovery observed following such a controlled decrease of Tregs suggests that reduced systemic immunosuppression at the time of the insult can enhance CNS repair. Overall, our data highlight a dynamic immune cell network needed for repair, acting in discrete compartments and stages, and involving effector and regulatory T cells, interconnected by mo-MΦ. Any of these populations may be detrimental to the repair process if their level or activity become dysregulated. Accordingly, therapeutic interventions must be both temporally and spatially controlled.


Subject(s)
Nerve Regeneration/immunology , Recovery of Function/physiology , Spinal Cord Injuries/physiopathology , T-Lymphocytes, Regulatory/physiology , Animals , Antigens, CD/metabolism , CD11c Antigen/genetics , CX3C Chemokine Receptor 1 , Diphtheria Toxin/pharmacology , Disease Models, Animal , Forkhead Transcription Factors/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Macrophages/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin-Oligodendrocyte Glycoprotein/immunology , Nerve Regeneration/genetics , Peptide Fragments/immunology , Receptors, Chemokine/genetics , Recovery of Function/genetics , T-Box Domain Proteins/deficiency , T-Box Domain Proteins/genetics , Vaccination
SELECTION OF CITATIONS
SEARCH DETAIL
...