Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(7)2024 Mar 24.
Article in English | MEDLINE | ID: mdl-38612448

ABSTRACT

The mechanisms of neuronal cell death in neurodegenerative disease remain incompletely understood, although recent studies have made significant advances. Apoptosis was previously considered to be the only mechanism of neuronal cell death in neurodegenerative diseases. However, recent findings have challenged this dogma, identifying new subtypes of necrotic neuronal cell death. The present review provides an updated summary of necrosis subtypes and discusses their potential roles in neurodegenerative cell death. Among numerous necrosis subtypes, including necroptosis, paraptosis, ferroptosis, and pyroptosis, transcriptional repression-induced atypical cell death (TRIAD) has been identified as a potential mechanism of neuronal cell death. TRIAD is induced by functional deficiency of TEAD-YAP and self-amplifies via the release of HMGB1. TRIAD is a feasible potential mechanism of neuronal cell death in Alzheimer's disease and other neurodegenerative diseases. In addition to induction of cell death, HMGB1 released during TRIAD activates brain inflammatory responses, which is a potential link between neurodegeneration and neuroinflammation.


Subject(s)
HMGB1 Protein , Neurodegenerative Diseases , Humans , Neuroinflammatory Diseases , Necrosis , Cell Death
2.
Commun Biol ; 7(1): 413, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38594382

ABSTRACT

Better understanding of the earliest molecular pathologies of all neurodegenerative diseases is expected to improve human therapeutics. We investigated the earliest molecular pathology of spinocerebellar ataxia type 1 (SCA1), a rare familial neurodegenerative disease that primarily induces death and dysfunction of cerebellum Purkinje cells. Extensive prior studies have identified involvement of transcription or RNA-splicing factors in the molecular pathology of SCA1. However, the regulatory network of SCA1 pathology, especially central regulators of the earliest developmental stages and inflammatory events, remains incompletely understood. Here, we elucidated the earliest developmental pathology of SCA1 using originally developed dynamic molecular network analyses of sequentially acquired RNA-seq data during differentiation of SCA1 patient-derived induced pluripotent stem cells (iPSCs) to Purkinje cells. Dynamic molecular network analysis implicated histone genes and cytokine-relevant immune response genes at the earliest stages of development, and revealed relevance of ISG15 to the following degradation and accumulation of mutant ataxin-1 in Purkinje cells of SCA1 model mice and human patients.


Subject(s)
Induced Pluripotent Stem Cells , Spinocerebellar Ataxias , Animals , Humans , Mice , Cytokines , Induced Pluripotent Stem Cells/pathology , Mice, Transgenic , Purkinje Cells/physiology , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/pathology , Ubiquitins
3.
Commun Med (Lond) ; 3(1): 170, 2023 Nov 28.
Article in English | MEDLINE | ID: mdl-38017287

ABSTRACT

BACKGROUND: Charcot-Marie-Tooth disease type 1A (CMT1A) is one of the most common hereditary peripheral neuropathies caused by duplication of 1.5 Mb genome region including PMP22 gene. We aimed to correct the duplication in human CMT1A patient-derived iPS cells (CMT1A-iPSCs) by genome editing and intended to analyze the effect on Schwann cells differentiated from CMT1A-iPSCs. METHODS: We designed multiple gRNAs targeting a unique sequence present at two sites that sandwich only a single copy of duplicated peripheral myelin protein 22 (PMP22) genes, and selected one of them (gRNA3) from screening their efficiencies by T7E1 mismatch detection assay. AAV2-hSaCas9-gRNAedit was generated by subcloning gRNA3 into pX601-AAV-CMV plasmid, and the genome editing AAV vector was infected to CMT1A-iPSCs or CMT1A-iPSC-derived Schwann cell precursors. The effect of the genome editing AAV vector on myelination was evaluated by co-immunostaining of myelin basic protein (MBP), a marker of mature myelin, and microtubule-associated protein  2(MAP2), a marker of neurites or by electron microscopy. RESULTS: Here we show that infection of CMT1A-iPS cells (iPSCs) with AAV2-hSaCas9-gRNAedit expressing both hSaCas9 and gRNA targeting the tandem repeat sequence decreased PMP22 gene duplication by 20-40%. Infection of CMT1A-iPSC-derived Schwann cell precursors with AAV2-hSaCas9-gRNAedit normalized PMP22 mRNA and PMP22 protein expression levels, and also ameliorated increased apoptosis and impaired myelination in CMT1A-iPSC-derived Schwann cells. CONCLUSIONS: In vivo transfer of AAV2-hSaCas9-gRNAedit to peripheral nerves could be a potential therapeutic modality for CMT1A patient after careful examinations of toxicity including off-target mutations.


Charcot-Marie-Tooth disease type 1A (CMT1A) is a common heritable form of the condition that develops when nerves in the body's extremities, such as the hands, feet and arms, are damaged due to an extra copy of PMP22 gene being incorrectly produced. Currently, no known therapies exist. Here, we developed a method to delete the additional copy of PMP22 gene by 20­40% to prevent overproduction. Our results show that this method can reduce PMP22 protein production, leading to near normal production in patient's nerve cells. Further safety assessments should now be undertaken. If the treatment is safe for patients it could become a therapeutic option for CMT1A patients.

4.
Cell Rep ; 42(8): 112962, 2023 08 29.
Article in English | MEDLINE | ID: mdl-37591248

ABSTRACT

Prion-like protein propagation is considered a common pathogenic mechanism in neurodegenerative diseases. Here we investigate the in vivo propagation pattern and aggregation state of mutant α-synuclein by injecting adeno-associated viral (AAV)-α-synuclein-A53T-EGFP into the mouse olfactory cortex. Comparison of aggregation states in various brain regions at multiple time points after injection using western blot analyses shows that the monomeric state of the mutant/misfolded protein propagates to remote brain regions by 2 weeks and that the propagated proteins aggregate in situ after being incorporated into neurons. Moreover, injection of Alexa 488-labeled α-synuclein-A53T confirms the monomeric propagation at 2 weeks. Super-resolution microscopy shows that both α-synuclein-A53T proteins propagate via the lymphatic system, penetrate perineuronal nets, and reach the surface of neurons. Electron microscopy shows that the propagated mutant/misfolded monomer forms fibrils characteristic of Parkinson's disease after its incorporation into neurons. These findings suggest a mode of propagation different from that of aggregate-dependent propagation.


Subject(s)
Parkinson Disease , alpha-Synuclein , Animals , Mice , alpha-Synuclein/genetics , Brain , Lymphatic System , Blotting, Western , Mutant Proteins
5.
Nat Commun ; 14(1): 9, 2023 01 04.
Article in English | MEDLINE | ID: mdl-36599853

ABSTRACT

Polyglutamine binding protein 5 (PQBP5), also called nucleolar protein 10 (NOL10), binds to polyglutamine tract sequences and is expressed in the nucleolus. Using dynamic imaging of high-speed atomic force microscopy, we show that PQBP5/NOL10 is an intrinsically disordered protein. Super-resolution microscopy and correlative light and electron microscopy method show that PQBP5/NOL10 makes up the skeletal structure of the nucleolus, constituting the granule meshwork in the granular component area, which is distinct from other nucleolar substructures, such as the fibrillar center and dense fibrillar component. In contrast to other nucleolar proteins, which disperse to the nucleoplasm under osmotic stress conditions, PQBP5/NOL10 remains in the nucleolus and functions as an anchor for reassembly of other nucleolar proteins. Droplet and thermal shift assays show that the biophysical features of PQBP5/NOL10 remain stable under stress conditions, explaining the spatial role of this protein. PQBP5/NOL10 can be functionally depleted by sequestration with polyglutamine disease proteins in vitro and in vivo, leading to the pathological deformity or disappearance of the nucleolus. Taken together, these findings indicate that PQBP5/NOL10 is an essential protein needed to maintain the structure of the nucleolus.


Subject(s)
Cell Nucleolus , Cell Nucleus , Nuclear Proteins , Humans , Cell Nucleolus/metabolism , Cell Nucleus/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Osmotic Pressure/physiology
6.
Nat Commun ; 12(1): 6565, 2021 11 15.
Article in English | MEDLINE | ID: mdl-34782623

ABSTRACT

Brain inflammation generally accompanies and accelerates neurodegeneration. Here we report a microglial mechanism in which polyglutamine binding protein 1 (PQBP1) senses extrinsic tau 3R/4R proteins by direct interaction and triggers an innate immune response by activating a cyclic GMP-AMP synthase (cGAS)-Stimulator of interferon genes (STING) pathway. Tamoxifen-inducible and microglia-specific depletion of PQBP1 in primary culture in vitro and mouse brain in vivo shows that PQBP1 is essential for sensing-tau to induce nuclear translocation of nuclear factor κB (NFκB), NFκB-dependent transcription of inflammation genes, brain inflammation in vivo, and eventually mouse cognitive impairment. Collectively, PQBP1 is an intracellular receptor in the cGAS-STING pathway not only for cDNA of human immunodeficiency virus (HIV) but also for the transmissible neurodegenerative disease protein tau. This study characterises a mechanism of brain inflammation that is common to virus infection and neurodegenerative disorders.


Subject(s)
DNA-Binding Proteins/metabolism , Encephalitis/metabolism , Membrane Proteins/metabolism , Microglia/metabolism , Nucleotidyltransferases/metabolism , Animals , Brain , DNA-Binding Proteins/genetics , Encephalitis/immunology , Female , HIV , Humans , Immunity, Innate , Male , Membrane Glycoproteins , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/drug effects , NF-kappa B/metabolism , Neurodegenerative Diseases , Nucleotidyltransferases/genetics , Tamoxifen/pharmacology
7.
Commun Biol ; 4(1): 1175, 2021 10 11.
Article in English | MEDLINE | ID: mdl-34635772

ABSTRACT

DNA damage is increased in Alzheimer's disease (AD), while the underlying mechanisms are unknown. Here, we employ comprehensive phosphoproteome analysis, and identify abnormal phosphorylation of 70 kDa subunit of Ku antigen (Ku70) at Ser77/78, which prevents Ku70-DNA interaction, in human AD postmortem brains. The abnormal phosphorylation inhibits accumulation of Ku70 to the foci of DNA double strand break (DSB), impairs DNA damage repair and eventually causes transcriptional repression-induced atypical cell death (TRIAD). Cells under TRIAD necrosis reveal senescence phenotypes. Extracellular high mobility group box 1 (HMGB1) protein, which is released from necrotic or hyper-activated neurons in AD, binds to toll-like receptor 4 (TLR4) of neighboring neurons, and activates protein kinase C alpha (PKCα) that executes Ku70 phosphorylation at Ser77/78. Administration of human monoclonal anti-HMGB1 antibody to post-symptomatic AD model mice decreases neuronal DSBs, suppresses secondary TRIAD necrosis of neurons, prevents escalation of neurodegeneration, and ameliorates cognitive symptoms. TRIAD shares multiple features with senescence. These results discover the HMGB1-Ku70 axis that accounts for the increase of neuronal DNA damage and secondary enhancement of TRIAD, the cell death phenotype of senescence, in AD.


Subject(s)
Alzheimer Disease/pathology , DNA Damage , DNA Repair , HMGB1 Protein/physiology , Ku Autoantigen/metabolism , Signal Transduction/genetics , Animals , HMGB1 Protein/genetics , Mice , Mice, Transgenic , Phosphorylation
8.
Commun Biol ; 4(1): 961, 2021 08 12.
Article in English | MEDLINE | ID: mdl-34385591

ABSTRACT

Multiple gene mutations cause familial frontotemporal lobar degeneration (FTLD) while no single gene mutations exists in sporadic FTLD. Various proteins aggregate in variable regions of the brain, leading to multiple pathological and clinical prototypes. The heterogeneity of FTLD could be one of the reasons preventing development of disease-modifying therapy. We newly develop a mathematical method to analyze chronological changes of PPI networks with sequential big data from comprehensive phosphoproteome of four FTLD knock-in (KI) mouse models (PGRNR504X-KI, TDP43N267S-KI, VCPT262A-KI and CHMP2BQ165X-KI mice) together with four transgenic mouse models of Alzheimer's disease (AD) and with APPKM670/671NL-KI mice at multiple time points. The new method reveals the common core pathological network across FTLD and AD, which is shared by mouse models and human postmortem brains. Based on the prediction, we performed therapeutic intervention of the FTLD models, and confirmed amelioration of pathologies and symptoms of four FTLD mouse models by interruption of the core molecule HMGB1, verifying the new mathematical method to predict dynamic molecular networks.


Subject(s)
Alzheimer Disease/etiology , Disease Models, Animal , Frontotemporal Lobar Degeneration/etiology , Alzheimer Disease/pathology , Animals , Frontotemporal Lobar Degeneration/pathology , Humans , Mice , Mice, Transgenic , Models, Theoretical
9.
ACS Chem Neurosci ; 12(16): 3015-3027, 2021 08 18.
Article in English | MEDLINE | ID: mdl-34319089

ABSTRACT

Tau aggregation is a central hallmark of tauopathies such as frontotemporal lobar degeneration and progressive supranuclear palsy as well as of Alzheimer's disease, and it has been a target for therapeutic development. Herein, we unexpectedly found that hepta-histidine (7H), an inhibitor of the interaction between Ku70 and Huntingtin proteins, suppresses aggregation of Tau-R3 peptides in vitro. Addition of the trans-activator of transcription (TAT) sequence (YGRKKRRQRRR) derived from the TAT protein to 7H increased its permeability into cells, and TAT-7H treatment of iPS cell-derived neurons carrying Tau or APP mutations suppressed Tau phosphorylation. These results indicate that 7H is a promising lead compound for developing anti-aggregation drugs against Tau-related neurodegenerative diseases including Alzheimer's disease (AD).


Subject(s)
Alzheimer Disease , Frontotemporal Lobar Degeneration , Tauopathies , Alzheimer Disease/drug therapy , Histidine , Humans , tau Proteins
10.
Life Sci Alliance ; 4(7)2021 07.
Article in English | MEDLINE | ID: mdl-34130995

ABSTRACT

The early-stage pathologies of frontotemporal lobal degeneration (FTLD) remain largely unknown. In VCPT262A-KI mice carrying VCP gene mutation linked to FTLD, insufficient DNA damage repair in neural stem/progenitor cells (NSCs) activated DNA-PK and CDK1 that disabled MCM3 essential for the G1/S cell cycle transition. Abnormal neural exit produced neurons carrying over unrepaired DNA damage and induced early-stage transcriptional repression-induced atypical cell death (TRIAD) necrosis accompanied by the specific markers pSer46-MARCKS and YAP. In utero gene therapy expressing normal VCP or non-phosphorylated mutant MCM3 rescued DNA damage, neuronal necrosis, cognitive function, and TDP43 aggregation in adult neurons of VCPT262A-KI mice, whereas similar therapy in adulthood was less effective. The similar early-stage neuronal necrosis was detected in PGRNR504X-KI, CHMP2BQ165X-KI, and TDPN267S-KI mice, and blocked by embryonic treatment with AAV-non-phospho-MCM3. Moreover, YAP-dependent necrosis occurred in neurons of human FTLD patients, and consistently pSer46-MARCKS was increased in cerebrospinal fluid (CSF) and serum of these patients. Collectively, developmental stress followed by early-stage neuronal necrosis is a potential target for therapeutics and one of the earliest general biomarkers for FTLD.


Subject(s)
Frontotemporal Lobar Degeneration/pathology , Neural Stem Cells/metabolism , Valosin Containing Protein/metabolism , Animals , Cell Cycle , Cell Lineage/genetics , Cells, Cultured , DNA Damage/genetics , DNA Damage/physiology , DNA-Binding Proteins/metabolism , Frontotemporal Lobar Degeneration/cerebrospinal fluid , Frontotemporal Lobar Degeneration/genetics , Gene Expression/genetics , Gene Expression Regulation/genetics , Mice , Mice, Inbred C57BL , Mutation , Necrosis/metabolism , Necrosis/pathology , Neural Stem Cells/pathology , Neurons/metabolism , Valosin Containing Protein/genetics
11.
Nat Commun ; 11(1): 507, 2020 01 24.
Article in English | MEDLINE | ID: mdl-31980612

ABSTRACT

The timing and characteristics of neuronal death in Alzheimer's disease (AD) remain largely unknown. Here we examine AD mouse models with an original marker, myristoylated alanine-rich C-kinase substrate phosphorylated at serine 46 (pSer46-MARCKS), and reveal an increase of neuronal necrosis during pre-symptomatic phase and a subsequent decrease during symptomatic phase. Postmortem brains of mild cognitive impairment (MCI) rather than symptomatic AD patients reveal a remarkable increase of necrosis. In vivo imaging reveals instability of endoplasmic reticulum (ER) in mouse AD models and genome-edited human AD iPS cell-derived neurons. The level of nuclear Yes-associated protein (YAP) is remarkably decreased in such neurons under AD pathology due to the sequestration into cytoplasmic amyloid beta (Aß) aggregates, supporting the feature of YAP-dependent necrosis. Suppression of early-stage neuronal death by AAV-YAPdeltaC reduces the later-stage extracellular Aß burden and cognitive impairment, suggesting that preclinical/prodromal YAP-dependent neuronal necrosis represents a target for AD therapeutics.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Cell Cycle Proteins/metabolism , Transcription Factors/metabolism , Alzheimer Disease/cerebrospinal fluid , Amyloid beta-Peptides/metabolism , Animals , Cell Nucleus/metabolism , Cognitive Dysfunction/cerebrospinal fluid , Cognitive Dysfunction/pathology , Computer Simulation , Disease Models, Animal , Endoplasmic Reticulum/pathology , Endoplasmic Reticulum/ultrastructure , Female , HMGB1 Protein/cerebrospinal fluid , Humans , Induced Pluripotent Stem Cells/metabolism , Lysophospholipids/metabolism , Male , Mice, Transgenic , Necrosis , Neurons/metabolism , Neurons/pathology , Signal Transduction , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Time-Lapse Imaging , YAP-Signaling Proteins
12.
Mol Psychiatry ; 23(10): 2090-2110, 2018 10.
Article in English | MEDLINE | ID: mdl-30283027

ABSTRACT

Early-phase pathologies of Alzheimer's disease (AD) are attracting much attention after clinical trials of drugs designed to remove beta-amyloid (Aß) aggregates failed to recover memory and cognitive function in symptomatic AD patients. Here, we show that phosphorylation of serine/arginine repetitive matrix 2 (SRRM2) at Ser1068, which is observed in the brains of early phase AD mouse models and postmortem end-stage AD patients, prevents its nuclear translocation by inhibiting interaction with T-complex protein subunit α. SRRM2 deficiency in neurons destabilized polyglutamine binding protein 1 (PQBP1), a causative gene for intellectual disability (ID), greatly affecting the splicing patterns of synapse-related genes, as demonstrated in a newly generated PQBP1-conditional knockout model. PQBP1 and SRRM2 were downregulated in cortical neurons of human AD patients and mouse AD models, and the AAV-PQBP1 vector recovered RNA splicing, the synapse phenotype, and the cognitive decline in the two mouse models. Finally, the kinases responsible for the phosphorylation of SRRM2 at Ser1068 were identified as ERK1/2 (MAPK3/1). These results collectively reveal a new aspect of AD pathology in which a phosphorylation signal affecting RNA splicing and synapse integrity precedes the formation of extracellular Aß aggregates and may progress in parallel with tau phosphorylation.


Subject(s)
Alzheimer Disease/genetics , Carrier Proteins/genetics , Nuclear Proteins/genetics , RNA-Binding Proteins/genetics , Active Transport, Cell Nucleus , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Brain/metabolism , Carrier Proteins/metabolism , Cognition , DNA-Binding Proteins , Disease Models, Animal , Humans , Induced Pluripotent Stem Cells , Intellectual Disability/genetics , MAP Kinase Signaling System , Mice , Mice, Knockout , Mice, Transgenic , Mitogen-Activated Protein Kinases/metabolism , Neurons/metabolism , Nuclear Proteins/metabolism , Phosphorylation , Primary Cell Culture , RNA Splicing , RNA-Binding Proteins/metabolism , tau Proteins/metabolism
13.
eNeuro ; 5(4)2018.
Article in English | MEDLINE | ID: mdl-30225354

ABSTRACT

Phosphorylation of myristoylated alanine-rich C kinase substrate (MARCKS) reflects neurite degeneration at the early stage of Alzheimer's disease (AD), before extracellular Aß aggregates are histologically detectable. Here, we demonstrate that similar changes in MARCKS occur in Parkinson's disease (PD) and dementia with Lewy bodies (DLB) pathologies in both mouse models and human patients. The increase in the level of pSer46-MARCKS began before α-synuclein aggregate formation, at a time when human α-Syn-BAC-Tg/GBA-hetero-KO mice exhibited no symptoms, and was sustained during aging, consistent with the pattern in human postmortem brains. The results strongly imply a common mechanism of pre-aggregation neurite degeneration in AD and PD/DLB pathologies.


Subject(s)
Aging/metabolism , Alzheimer Disease , Lewy Body Disease , Myristoylated Alanine-Rich C Kinase Substrate/metabolism , Neurites/pathology , Parkinson Disease , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Disease Models, Animal , Female , Humans , Lewy Body Disease/metabolism , Lewy Body Disease/pathology , Male , Mice , Mice, Transgenic , Parkinson Disease/metabolism , Parkinson Disease/pathology , Phosphorylation
14.
Nat Commun ; 9(1): 433, 2018 01 30.
Article in English | MEDLINE | ID: mdl-29382817

ABSTRACT

Mutations in the progranulin (PGRN) gene cause a tau pathology-negative and TDP43 pathology-positive form of frontotemporal lobar degeneration (FTLD-TDP). We generated a knock-in mouse harboring the R504X mutation (PGRN-KI). Phosphoproteomic analysis of this model revealed activation of signaling pathways connecting PKC and MAPK to tau prior to TDP43 aggregation and cognitive impairments, and identified PKCα as the kinase responsible for the early-stage tau phosphorylation at Ser203. Disinhibition of Gas6 binding to Tyro3 due to PGRN reduction results in activation of PKCα via PLCγ, inducing tau phosphorylation at Ser203, mislocalization of tau to dendritic spines, and spine loss. Administration of a PKC inhibitor, B-Raf inhibitor, or knockdown of molecules in the Gas6-Tyro3-tau axis rescues spine loss and cognitive impairment of PGRN-KI mice. Collectively, these results suggest that targeting of early-stage and aggregation-independent tau signaling represents a promising therapeutic strategy for this disease.


Subject(s)
Frontotemporal Lobar Degeneration/etiology , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , tau Proteins/metabolism , Animals , Disease Models, Animal , Frontotemporal Lobar Degeneration/metabolism , Gene Knock-In Techniques , Granulins , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Phospholipase C gamma/metabolism , Phosphorylation , Progranulins , Protein Kinase C-alpha/metabolism , Proto-Oncogene Proteins B-raf/metabolism
15.
Nat Commun ; 8(1): 1864, 2017 11 30.
Article in English | MEDLINE | ID: mdl-29192206

ABSTRACT

YAP and its neuronal isoform YAPdeltaC are implicated in various cellular functions. We found that expression of YAPdeltaC during development, but not adulthood, rescued neurodegeneration phenotypes of mutant ataxin-1 knock-in (Atxn1-KI) mice. YAP/YAPdeltaC interacted with RORα via the second WW domain and served as co-activators of its transcriptional activity. YAP/YAPdeltaC formed a transcriptional complex with RORα on cis-elements of target genes and regulated their expression. Both normal and mutant Atxn1 interacted with YAP/YAPdeltaC, but only mutant Atxn1 depleted YAP/YAPdeltaC from the RORα complex to suppress transcription on short timescales. Over longer periods, mutant Atxn1 also decreased RORα in vivo. Genetic supplementation of YAPdeltaC restored the RORα and YAP/YAPdeltaC levels, recovered YAP/YAPdeltaC in the RORα complex and normalized target gene transcription in Atxn1-KI mice in vivo. Collectively, our data suggest that functional impairment of YAP/YAPdeltaC by mutant Atxn1 during development determines the adult pathology of SCA1 by suppressing RORα-mediated transcription.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Ataxin-1/genetics , Cerebellum/metabolism , Gene Expression Regulation, Developmental/genetics , Neurons/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Phosphoproteins/metabolism , Spinocerebellar Ataxias/genetics , Animals , Cell Cycle Proteins , Cerebellum/cytology , Disease Models, Animal , Gene Knock-In Techniques , Male , Mice , Phenotype , Protein Isoforms , Rotarod Performance Test , Spinocerebellar Ataxias/physiopathology , YAP-Signaling Proteins
16.
Sci Rep ; 6: 33861, 2016 Sep 22.
Article in English | MEDLINE | ID: mdl-27653664

ABSTRACT

We identified drug seeds for treating Huntington's disease (HD) by combining in vitro single molecule fluorescence spectroscopy, in silico molecular docking simulations, and in vivo fly and mouse HD models to screen for inhibitors of abnormal interactions between mutant Htt and physiological Ku70, an essential DNA damage repair protein in neurons whose function is known to be impaired by mutant Htt. From 19,468 and 3,010,321 chemicals in actual and virtual libraries, fifty-six chemicals were selected from combined in vitro-in silico screens; six of these were further confirmed to have an in vivo effect on lifespan in a fly HD model, and two chemicals exerted an in vivo effect on the lifespan, body weight and motor function in a mouse HD model. Two oligopeptides, hepta-histidine (7H) and Angiotensin III, rescued the morphological abnormalities of primary neurons differentiated from iPS cells of human HD patients. For these selected drug seeds, we proposed a possible common structure. Unexpectedly, the selected chemicals enhanced rather than inhibited Htt aggregation, as indicated by dynamic light scattering analysis. Taken together, these integrated screens revealed a new pathway for the molecular targeted therapy of HD.

17.
Sci Rep ; 6: 31895, 2016 08 25.
Article in English | MEDLINE | ID: mdl-27557632

ABSTRACT

Alzheimer's disease (AD) is the most common neurodegenerative disease, but it remains an intractable condition. Its pathogenesis is predominantly attributed to the aggregation and transmission of two molecules, Aß and tau; however, other pathological mechanisms are possible. Here, we reveal that phosphorylation of MARCKS, a submembrane protein that regulates the stability of the actin network, occurs at Ser46 prior to aggregation of Aß and is sustained throughout the course of AD in human and mouse brains. Furthermore, HMGB1 released from necrotic or hyperexcitatory neurons binds to TLR4, triggers the specific phosphorylation of MARCKS via MAP kinases, and induces neurite degeneration, the classical hallmark of AD pathology. Subcutaneous injection of a newly developed monoclonal antibody against HMGB1 strongly inhibits neurite degeneration even in the presence of Aß plaques and completely recovers cognitive impairment in a mouse model. HMGB1 and Aß mutually affect polymerization of the other molecule, and the therapeutic effects of the anti-HMGB1 monoclonal antibody are mediated by Aß-dependent and Aß-independent mechanisms. We propose that HMGB1 is a critical pathogenic molecule promoting AD pathology in parallel with Aß and tau and a new key molecular target of preclinical antibody therapy to delay the onset of AD.


Subject(s)
Alzheimer Disease/metabolism , HMGB1 Protein/metabolism , Myristoylated Alanine-Rich C Kinase Substrate/metabolism , Neurites/pathology , Toll-Like Receptor 4/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacology , Disease Models, Animal , HMGB1 Protein/antagonists & inhibitors , Humans , Mice , Mitogen-Activated Protein Kinases/metabolism , Molecular Targeted Therapy , Myristoylated Alanine-Rich C Kinase Substrate/chemistry , Neurites/drug effects , Neurites/metabolism , Phosphorylation/drug effects , Serine/metabolism , tau Proteins/metabolism
18.
Hum Mol Genet ; 25(21): 4749-4770, 2016 11 01.
Article in English | MEDLINE | ID: mdl-28171658

ABSTRACT

Neuronal cell death in neurodegenerative diseases is not fully understood. Here we report that mutant huntingtin (Htt), a causative gene product of Huntington's diseases (HD) selectively induces a new form of necrotic cell death, in which endoplasmic reticulum (ER) enlarges and cell body asymmetrically balloons and finally ruptures. Pharmacological and genetic analyses revealed that the necrotic cell death is distinct from the RIP1/3 pathway-dependent necroptosis, but mediated by a functional deficiency of TEAD/YAP-dependent transcription. In addition, we revealed that a cell cycle regulator, Plk1, switches the balance between TEAD/YAP-dependent necrosis and p73/YAP-dependent apoptosis by shifting the interaction partner of YAP from TEAD to p73 through YAP phosphorylation at Thr77. In vivo ER imaging with two-photon microscopy detects similar ER enlargement, and viral vector-mediated delivery of YAP as well as chemical inhibitors of the Hippo pathway such as S1P recover the ER instability and necrosis in HD model mice. Intriguingly S1P completely stops the decline of motor function of HD model mice even after the onset of symptom. Collectively, we suggest approaches targeting the signalling pathway of TEAD/YAP-transcription-dependent necrosis (TRIAD) could lead to a therapeutic development against HD.


Subject(s)
Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Necrosis/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Cycle Proteins , Cell Death , DNA-Binding Proteins/genetics , Endoplasmic Reticulum/metabolism , Humans , Huntington Disease/metabolism , Mice , Mice, Inbred C57BL , Necrosis/genetics , Neurons/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Phosphorylation , Primary Cell Culture , Protein Binding , Signal Transduction , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
19.
Hum Mol Genet ; 25(20): 4432-4447, 2016 10 15.
Article in English | MEDLINE | ID: mdl-28173122

ABSTRACT

DNA damage and repair is a critical domain of many neurodegenerative diseases. In this study, we focused on RpA1, a candidate key molecule in polyQ disease pathologies, and tested the therapeutic effect of adeno-associated virus (AAV) vector expressing RpA1 on mutant Ataxin-1 knock-in (Atxn1-KI) mice. We found significant effects on motor functions, normalized DNA damage markers (γH2AX and 53BP1), and improved Purkinje cell morphology; effects that lasted for 50 weeks following AAV-RpA1 infection. In addition, we confirmed that AAV-RpA1 indirectly recovered multiple cellular functions such as RNA splicing, transcription and cell cycle as well as abnormal morphology of dendrite and dendritic spine of Purkinje cells in Atxn1-KI mice. All these results suggested a possibility of gene therapy with RpA1 for SCA1.


Subject(s)
Ataxin-1/genetics , DNA Repair , Mutation , Replication Protein A/metabolism , Spinocerebellar Ataxias/metabolism , Animals , Cell Cycle , DNA/metabolism , DNA Damage , Dependovirus , Disease Models, Animal , Gene Knock-In Techniques , Genetic Therapy , Mice , Purkinje Cells/metabolism , Purkinje Cells/pathology , Purkinje Cells/physiology , RNA/metabolism , RNA Splicing , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/pathology , Spinocerebellar Ataxias/physiopathology , Transcription, Genetic
20.
Sci Rep ; 5: 12115, 2015 Jul 14.
Article in English | MEDLINE | ID: mdl-26169250

ABSTRACT

We developed a new technique to observe macroautophagy in the brain in vivo, and examined whether fasting induced macroautophagy in neurons and how the induction was different between Alzheimer's disease (AD) model and control mice. Lentivirus for EGFP-LC3 injected into the brain successfully visualized autophagosome in living neurons by two-photon microscopy. The time-lapse imaging revealed that fasting increased the number, size and signal intensity of autophagosome in neurons. In AD model mice, these parameters of autophagosome were higher at the basal levels before starvation, and increased more rapidly by fasting than in control mice. However, metabolism of exogenous labeled Aß evaluated by the new technique suggested that the activated macroautophagy was insufficient to degrade the intracellular Aß increased by enhanced uptake from extracellular space after fasting. Ordinary immunohistochemistry also revealed that fasting increased intracellular accumulation of endogenous Aß, triggered cell dysfunction but did not mostly decrease extracellular Aß accumulation. Moreover, we unexpectedly discovered a circadian rhythm of basal level of macroautophagy. These results revealed new aspects of neuronal autophagy in normal/AD states and indicated usefulness of our method for evaluating autophagy functions in vivo.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Autophagy , Fasting/metabolism , Neurons/metabolism , Alzheimer Disease/pathology , Animals , Brain/metabolism , Circadian Rhythm , Disease Models, Animal , Extracellular Space/metabolism , Intracellular Space/metabolism , Mice , Microscopy, Fluorescence , Neurons/cytology , Proteolysis
SELECTION OF CITATIONS
SEARCH DETAIL
...