Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Neuron ; 109(14): 2275-2291.e8, 2021 07 21.
Article in English | MEDLINE | ID: mdl-34133945

ABSTRACT

C9orf72 repeat expansions cause inherited amyotrophic lateral sclerosis (ALS)/frontotemporal dementia (FTD) and result in both loss of C9orf72 protein expression and production of potentially toxic RNA and dipeptide repeat proteins. In addition to ALS/FTD, C9orf72 repeat expansions have been reported in a broad array of neurodegenerative syndromes, including Alzheimer's disease. Here we show that C9orf72 deficiency promotes a change in the homeostatic signature in microglia and a transition to an inflammatory state characterized by an enhanced type I IFN signature. Furthermore, C9orf72-depleted microglia trigger age-dependent neuronal defects, in particular enhanced cortical synaptic pruning, leading to altered learning and memory behaviors in mice. Interestingly, C9orf72-deficient microglia promote enhanced synapse loss and neuronal deficits in a mouse model of amyloid accumulation while paradoxically improving plaque clearance. These findings suggest that altered microglial function due to decreased C9orf72 expression directly contributes to neurodegeneration in repeat expansion carriers independent of gain-of-function toxicities.


Subject(s)
Aging/metabolism , Amyloid/metabolism , C9orf72 Protein/metabolism , Microglia/metabolism , Synapses/metabolism , Aging/genetics , Aging/pathology , Amyloid/genetics , Animals , C9orf72 Protein/genetics , DNA Repeat Expansion , Disease Models, Animal , Lysosomes/metabolism , Mice , Mice, Knockout , Synapses/pathology
2.
Cell Syst ; 12(2): 159-175.e9, 2021 02 17.
Article in English | MEDLINE | ID: mdl-33382996

ABSTRACT

Induced pluripotent stem cell (iPSC)-derived neural cultures from amyotrophic lateral sclerosis (ALS) patients can model disease phenotypes. However, heterogeneity arising from genetic and experimental variability limits their utility, impacting reproducibility and the ability to track cellular origins of pathogenesis. Here, we present methodologies using single-cell RNA sequencing (scRNA-seq) analysis to address these limitations. By repeatedly differentiating and applying scRNA-seq to motor neurons (MNs) from healthy, familial ALS, sporadic ALS, and genome-edited iPSC lines across multiple patients, batches, and platforms, we account for genetic and experimental variability toward identifying unified and reproducible ALS signatures. Combining HOX and developmental gene expression with global clustering, we anatomically classified cells into rostrocaudal, progenitor, and postmitotic identities. By relaxing statistical thresholds, we discovered genes in iPSC-MNs that were concordantly dysregulated in postmortem MNs and yielded predictive ALS markers in other human and mouse models. Our approach thus revealed early, convergent, and MN-resolved signatures of ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Induced Pluripotent Stem Cells/metabolism , Motor Neurons/metabolism , Animals , Disease Models, Animal , Humans , Mice
3.
Nature ; 585(7823): 96-101, 2020 09.
Article in English | MEDLINE | ID: mdl-32814898

ABSTRACT

Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are neurodegenerative disorders that overlap in their clinical presentation, pathology and genetic origin. Autoimmune disorders are also overrepresented in both ALS and FTD, but this remains an unexplained epidemiologic observation1-3. Expansions of a hexanucleotide repeat (GGGGCC) in the C9orf72 gene are the most common cause of familial ALS and FTD (C9-ALS/FTD), and lead to both repeat-containing RNA and dipeptide accumulation, coupled with decreased C9orf72 protein expression in brain and peripheral blood cells4-6. Here we show in mice that loss of C9orf72 from myeloid cells alone is sufficient to recapitulate the age-dependent lymphoid hypertrophy and autoinflammation seen in animals with a complete knockout of C9orf72. Dendritic cells isolated from C9orf72-/- mice show marked early activation of the type I interferon response, and C9orf72-/- myeloid cells are selectively hyperresponsive to activators of the stimulator of interferon genes (STING) protein-a key regulator of the innate immune response to cytosolic DNA. Degradation of STING through the autolysosomal pathway is diminished in C9orf72-/- myeloid cells, and blocking STING suppresses hyperactive type I interferon responses in C9orf72-/- immune cells as well as splenomegaly and inflammation in C9orf72-/- mice. Moreover, mice lacking one or both copies of C9orf72 are more susceptible to experimental autoimmune encephalitis, mirroring the susceptibility to autoimmune diseases seen in people with C9-ALS/FTD. Finally, blood-derived macrophages, whole blood and brain tissue from patients with C9-ALS/FTD all show an elevated type I interferon signature compared with samples from people with sporadic ALS/FTD; this increased interferon response can be suppressed with a STING inhibitor. Collectively, our results suggest that patients with C9-ALS/FTD have an altered immunophenotype because their reduced levels of C9orf72 cannot suppress the inflammation mediated by the induction of type I interferons by STING.


Subject(s)
C9orf72 Protein/genetics , C9orf72 Protein/metabolism , Inflammation/metabolism , Inflammation/prevention & control , Membrane Proteins/metabolism , Myeloid Cells/metabolism , Aging/immunology , Amyotrophic Lateral Sclerosis/genetics , Animals , C9orf72 Protein/deficiency , Dendritic Cells/cytology , Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Humans , Inflammation/genetics , Inflammation/immunology , Interferon Type I/biosynthesis , Interferon Type I/immunology , Membrane Proteins/antagonists & inhibitors , Mice , Myeloid Cells/immunology , Neoplasms/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology
4.
J Trauma Acute Care Surg ; 89(5): 955-961, 2020 11.
Article in English | MEDLINE | ID: mdl-32472900

ABSTRACT

BACKGROUND: How recurrent traumatic brain injury (rTBI) alters brain function years after insult is largely unknown. This study aims to characterize the mechanistic cause for long-term brain deterioration following rTBI using a rat model. METHODS: Eighteen Sprague-Dawley wild-type rats underwent bilateral rTBI using a direct skull impact device or sham treatment, once per week for 5 weeks, and were euthanized 56 weeks after the first injury. Weekly rotarod performance measured motor deficits. Beam walk and grip strength were also assessed. Brain tissue were stained and volume was computed using Stereo Investigator's Cavalieri Estimator. The L5 cortical layer proximal to the injury site was microdissected and submitted for sequencing with count analyzed using R "DESeq2" and "GOStats." Brain-derived neurotrophic factor (BDNF) levels were determined using enzyme-linked immunosorbent assay. RESULTS: Rotarod data demonstrated permanent deficits 1 year after rTBI. Decreased beam walk performance and grip strength was noted among rTBI rodents. Recurrent traumatic brain injury led to thinner cortex and thinner corpus callosum, enlarged ventricles, and differential expression of 72 genes (25 upregulated, 47 downregulated) including dysregulation of those associated with TBI (BDNF, NR4A1/2/3, Arc, and Egr) and downregulation in pathways associated with neuroprotection and neuroplasticity. Over the course of the study, BDNF levels decreased in both rTBI and sham rodents, and at each time point, the decrease in BDNF was more pronounced after rTBI. CONCLUSION: Recurrent traumatic brain injury causes significant long-term alteration in brain health leading to permanent motor deficits, cortical and corpus callosum thinning, and expansion of the lateral ventricles. Gene expression and BDNF analysis suggest a significant drop in pathways associated with neuroplasticity and neuroprotection. Although rTBI may not cause immediate neurological abnormalities, continued brain deterioration occurs after the initial trauma in part due to a decline in genes associated with neuroplasticity and neuroprotection.


Subject(s)
Brain Injuries, Traumatic/complications , Brain-Derived Neurotrophic Factor/blood , Brain/pathology , Cognitive Dysfunction/etiology , Animals , Brain-Derived Neurotrophic Factor/metabolism , Cognitive Dysfunction/blood , Cognitive Dysfunction/pathology , Disease Models, Animal , Down-Regulation , Humans , Rats , Recurrence , Time Factors
5.
Immunity ; 49(5): 873-885.e7, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30366765

ABSTRACT

Receptor interacting protein 2 (RIP2) plays a role in sensing intracellular pathogens, but its function in T cells is unclear. We show that RIP2 deficiency in CD4+ T cells resulted in chronic and severe interleukin-17A-mediated inflammation during Chlamydia pneumoniae lung infection, increased T helper 17 (Th17) cell formation in lungs of infected mice, accelerated atherosclerosis, and more severe experimental autoimmune encephalomyelitis. While RIP2 deficiency resulted in reduced conventional Th17 cell differentiation, it led to significantly enhanced differentiation of pathogenic (p)Th17 cells, which was dependent on RORα transcription factor and interleukin-1 but independent of nucleotide oligomerization domain (NOD) 1 and 2. Overexpression of RIP2 resulted in suppression of pTh17 cell differentiation, an effect mediated by its CARD domain, and phenocopied by a cell-permeable RIP2 CARD peptide. Our data suggest that RIP2 has a T cell-intrinsic role in determining the balance between homeostatic and pathogenic Th17 cell responses.


Subject(s)
Cell Differentiation/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Th17 Cells/cytology , Th17 Cells/metabolism , Animals , Atherosclerosis , Biomarkers , Caspase Activation and Recruitment Domain , Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/mortality , Gene Expression , Immunophenotyping , Inflammation/genetics , Inflammation/metabolism , Interleukin-17/biosynthesis , Interleukin-1beta , Mice , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Receptor-Interacting Protein Serine-Threonine Kinase 2 , Receptor-Interacting Protein Serine-Threonine Kinases/chemistry , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
6.
J Vis Exp ; (132)2018 02 27.
Article in English | MEDLINE | ID: mdl-29553509

ABSTRACT

The accumulation of misfolded proteins is central to pathology in Huntington's disease (HD) and many other neurodegenerative disorders. Specifically, a key pathological feature of HD is the aberrant accumulation of mutant HTT (mHTT) protein into high molecular weight complexes and intracellular inclusion bodies composed of fragments and other proteins. Conventional methods to measure and understand the contributions of various forms of mHTT-containing aggregates include fluorescence microscopy, western blot analysis, and filter trap assays. However, most of these methods are conformation specific, and therefore may not resolve the full state of mHTT protein flux due to the complex nature of aggregate solubility and resolution. For the identification of aggregated mHTT and various modified forms and complexes, separation and solubilization of the cellular aggregates and fragments is mandatory. Here we describe a method to isolate and visualize soluble mHTT, monomers, oligomers, fragments, and an insoluble high molecular weight (HMW) accumulated mHTT species. HMW mHTT tracks with disease progression, corresponds with mouse behavior readouts, and has been beneficially modulated by certain therapeutic interventions1. This approach can be used with mouse brain, peripheral tissues, and cell culture but may be adapted to other model systems or disease contexts.


Subject(s)
Dose Fractionation, Radiation , Huntingtin Protein/genetics , Huntington Disease/drug therapy , Animals , Humans , Huntingtin Protein/metabolism , Mice , Models, Biological
7.
Appl Neuropsychol Adult ; 24(6): 532-539, 2017.
Article in English | MEDLINE | ID: mdl-27541806

ABSTRACT

The 3s Spreadsheet Test (3S Test) is a cancellation test with high stimulus density and low target-to-distractor ratio, to assess stimulus-centered and viewer-centered spatial neglect. Twenty-five stroke survivors with left-sided neglect and 68 age-matched healthy controls took the 3S Test. Patients also took the Apples Test, a validated cancellation test. No patient's accuracy on the 3S Test was in the normal range. A total of 91.7% of the patients had an abnormal start; 52.6% of the patients took abnormally long time to complete the 3S Test. The 3S and Apples Tests agreed poorly on both the viewer-centered and stimulus-centered neglect classifications (κ = .25 and .05, respectively), but consistently identified 8 of the 25 patients (32%) as having both forms of neglect while each isolated form of neglect was present in one patient (4%). Lesion data were consistent with previous studies. These findings suggest that the 3S Test is a sensitive cancellation test for assessing viewer-centered and stimulus-centered spatial neglect. We are currently developing the 3S Test version 2 to refine the test stimuli and the scoring procedure.


Subject(s)
Neuropsychological Tests/standards , Perceptual Disorders/diagnosis , Space Perception/physiology , Stroke/diagnosis , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Perceptual Disorders/etiology , Stroke/complications
8.
Nat Neurosci ; 19(9): 1256-67, 2016 09.
Article in English | MEDLINE | ID: mdl-27428653

ABSTRACT

Modeling amyotrophic lateral sclerosis (ALS) with human induced pluripotent stem cells (iPSCs) aims to reenact embryogenesis, maturation and aging of spinal motor neurons (spMNs) in vitro. As the maturity of spMNs grown in vitro compared to spMNs in vivo remains largely unaddressed, it is unclear to what extent this in vitro system captures critical aspects of spMN development and molecular signatures associated with ALS. Here, we compared transcriptomes among iPSC-derived spMNs, fetal spinal tissues and adult spinal tissues. This approach produced a maturation scale revealing that iPSC-derived spMNs were more similar to fetal spinal tissue than to adult spMNs. Additionally, we resolved gene networks and pathways associated with spMN maturation and aging. These networks enriched for pathogenic familial ALS genetic variants and were disrupted in sporadic ALS spMNs. Altogether, our findings suggest that developing strategies to further mature and age iPSC-derived spMNs will provide more effective iPSC models of ALS pathology.


Subject(s)
Aging , Amyotrophic Lateral Sclerosis/metabolism , Motor Neurons/cytology , Neurogenesis/physiology , Amyotrophic Lateral Sclerosis/physiopathology , Gene Expression , Gene Regulatory Networks , Humans , Induced Pluripotent Stem Cells/physiology , Neurogenesis/genetics
9.
Neuron ; 90(3): 507-20, 2016 05 04.
Article in English | MEDLINE | ID: mdl-27146268

ABSTRACT

The disruption of protein quality control networks is central to pathology in Huntington's disease (HD) and other neurodegenerative disorders. The aberrant accumulation of insoluble high-molecular-weight protein complexes containing the Huntingtin (HTT) protein and SUMOylated protein corresponds to disease manifestation. We previously identified an HTT-selective E3 SUMO ligase, PIAS1, that regulates HTT accumulation and SUMO modification in cells. Here we investigated whether PIAS1 modulation in neurons alters HD-associated phenotypes in vivo. Instrastriatal injection of a PIAS1-directed miRNA significantly improved behavioral phenotypes in rapidly progressing mutant HTT (mHTT) fragment R6/2 mice. PIAS1 reduction prevented the accumulation of mHTT and SUMO- and ubiquitin-modified proteins, increased synaptophysin levels, and normalized key inflammatory markers. In contrast, PIAS1 overexpression exacerbated mHTT-associated phenotypes and aberrant protein accumulation. These results confirm the association between aberrant accumulation of expanded polyglutamine-dependent insoluble protein species and pathogenesis, and they link phenotypic benefit to reduction of these species through PIAS1 modulation.


Subject(s)
Brain/metabolism , Huntington Disease/genetics , Mutation/genetics , Nerve Tissue Proteins/genetics , Neurons/metabolism , Protein Inhibitors of Activated STAT/genetics , Small Ubiquitin-Related Modifier Proteins/genetics , Animals , Disease Models, Animal , Humans , Huntingtin Protein/genetics , Mice , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Phenotype
10.
Neuron ; 88(5): 892-901, 2015 Dec 02.
Article in English | MEDLINE | ID: mdl-26637796

ABSTRACT

Noncoding expansions of a hexanucleotide repeat (GGGGCC) in the C9orf72 gene are the most common cause of familial amyotrophic lateral sclerosis and frontotemporal dementia. Here we report transgenic mice carrying a bacterial artificial chromosome (BAC) containing the full human C9orf72 gene with either a normal allele (15 repeats) or disease-associated expansion (∼100-1,000 repeats; C9-BACexp). C9-BACexp mice displayed pathologic features seen in C9orf72 expansion patients, including widespread RNA foci and repeat-associated non-ATG (RAN) translated dipeptides, which were suppressed by antisense oligonucleotides targeting human C9orf72. Nucleolin distribution was altered, supporting that either C9orf72 transcripts or RAN dipeptides promote nucleolar dysfunction. Despite early and widespread production of RNA foci and RAN dipeptides in C9-BACexp mice, behavioral abnormalities and neurodegeneration were not observed even at advanced ages, supporting the hypothesis that RNA foci and RAN dipeptides occur presymptomatically and are not sufficient to drive neurodegeneration in mice at levels seen in patients.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Brain/pathology , DNA Repeat Expansion/genetics , Frontotemporal Dementia/pathology , Proteins/genetics , Spinal Cord/pathology , Age Factors , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Brain/metabolism , C9orf72 Protein , Cells, Cultured , Chromosomes, Artificial, Bacterial/genetics , Chromosomes, Artificial, Bacterial/metabolism , Disease Models, Animal , Frontotemporal Dementia/genetics , Frontotemporal Dementia/physiopathology , Glutamic Acid/pharmacology , Humans , Mice , Mice, Transgenic , Motor Activity/genetics , Muscle Strength/genetics , Neuromuscular Junction/genetics , Neuromuscular Junction/pathology , Neurons/drug effects , Psychomotor Performance/physiology , Spinal Cord/metabolism
11.
Sci Transl Med ; 5(208): 208ra149, 2013 Oct 23.
Article in English | MEDLINE | ID: mdl-24154603

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a severe neurodegenerative condition characterized by loss of motor neurons in the brain and spinal cord. Expansions of a hexanucleotide repeat (GGGGCC) in the noncoding region of the C9ORF72 gene are the most common cause of the familial form of ALS (C9-ALS), as well as frontotemporal lobar degeneration and other neurological diseases. How the repeat expansion causes disease remains unclear, with both loss of function (haploinsufficiency) and gain of function (either toxic RNA or protein products) proposed. We report a cellular model of C9-ALS with motor neurons differentiated from induced pluripotent stem cells (iPSCs) derived from ALS patients carrying the C9ORF72 repeat expansion. No significant loss of C9ORF72 expression was observed, and knockdown of the transcript was not toxic to cultured human motor neurons. Transcription of the repeat was increased, leading to accumulation of GGGGCC repeat-containing RNA foci selectively in C9-ALS iPSC-derived motor neurons. Repeat-containing RNA foci colocalized with hnRNPA1 and Pur-α, suggesting that they may be able to alter RNA metabolism. C9-ALS motor neurons showed altered expression of genes involved in membrane excitability including DPP6, and demonstrated a diminished capacity to fire continuous spikes upon depolarization compared to control motor neurons. Antisense oligonucleotides targeting the C9ORF72 transcript suppressed RNA foci formation and reversed gene expression alterations in C9-ALS motor neurons. These data show that patient-derived motor neurons can be used to delineate pathogenic events in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , DNA Repeat Expansion/genetics , Induced Pluripotent Stem Cells/pathology , Motor Neurons/metabolism , Motor Neurons/pathology , Proteins/genetics , RNA/metabolism , C9orf72 Protein , Exons/genetics , Gene Knockdown Techniques , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Motor Neurons/drug effects , Oligonucleotides, Antisense/pharmacology , RNA/biosynthesis , RNA/genetics , Transcription, Genetic/drug effects
12.
Cell Rep ; 4(2): 362-75, 2013 Jul 25.
Article in English | MEDLINE | ID: mdl-23871671

ABSTRACT

A key feature in Huntington disease (HD) is the accumulation of mutant Huntingtin (HTT) protein, which may be regulated by posttranslational modifications. Here, we define the primary sites of SUMO modification in the amino-terminal domain of HTT, show modification downstream of this domain, and demonstrate that HTT is modified by the stress-inducible SUMO-2. A systematic study of E3 SUMO ligases demonstrates that PIAS1 is an E3 SUMO ligase for both HTT SUMO-1 and SUMO-2 modification and that reduction of dPIAS in a mutant HTT Drosophila model is protective. SUMO-2 modification regulates accumulation of insoluble HTT in HeLa cells in a manner that mimics proteasome inhibition and can be modulated by overexpression and acute knockdown of PIAS1. Finally, the accumulation of SUMO-2-modified proteins in the insoluble fraction of HD postmortem striata implicates SUMO-2 modification in the age-related pathogenic accumulation of mutant HTT and other cellular proteins that occurs during HD progression.


Subject(s)
Huntington Disease/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Protein Inhibitors of Activated STAT/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Aged , Aged, 80 and over , Amino Acid Sequence , Animals , Catalytic Domain , Drosophila , Female , HeLa Cells , Humans , Huntingtin Protein , Huntington Disease/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Molecular Sequence Data , Mutation , Protein Inhibitors of Activated STAT/genetics , Protein Processing, Post-Translational , Small Ubiquitin-Related Modifier Proteins/genetics , Transfection , Ubiquitin-Protein Ligases/metabolism
13.
J Cell Biol ; 187(7): 1083-99, 2009 Dec 28.
Article in English | MEDLINE | ID: mdl-20026656

ABSTRACT

Expansion of the polyglutamine repeat within the protein Huntingtin (Htt) causes Huntington's disease, a neurodegenerative disease associated with aging and the accumulation of mutant Htt in diseased neurons. Understanding the mechanisms that influence Htt cellular degradation may target treatments designed to activate mutant Htt clearance pathways. We find that Htt is phosphorylated by the inflammatory kinase IKK, enhancing its normal clearance by the proteasome and lysosome. Phosphorylation of Htt regulates additional post-translational modifications, including Htt ubiquitination, SUMOylation, and acetylation, and increases Htt nuclear localization, cleavage, and clearance mediated by lysosomal-associated membrane protein 2A and Hsc70. We propose that IKK activates mutant Htt clearance until an age-related loss of proteasome/lysosome function promotes accumulation of toxic post-translationally modified mutant Htt. Thus, IKK activation may modulate mutant Htt neurotoxicity depending on the cell's ability to degrade the modified species.


Subject(s)
I-kappa B Kinase/physiology , Lysosomes/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Animals , Brain/metabolism , Cell Line , Humans , Huntingtin Protein , Mice , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/chemistry , Nuclear Proteins/analysis , Nuclear Proteins/chemistry , Phosphorylation , Protein Structure, Tertiary , Rats , Rats, Sprague-Dawley , Solubility , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL
...