Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Acta Neuropathol ; 147(1): 84, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38750212

ABSTRACT

Amyotrophic Lateral Sclerosis/Parkinsonism-Dementia Complex (ALS/PDC), a rare and complex neurological disorder, is predominantly observed in the Western Pacific islands, including regions of Japan, Guam, and Papua. This enigmatic condition continues to capture medical attention due to affected patients displaying symptoms that parallel those seen in either classical amyotrophic lateral sclerosis (ALS) or Parkinson's disease (PD). Distinctly, postmortem examinations of the brains of affected individuals have shown the presence of α-synuclein aggregates and TDP-43, which are hallmarks of PD and classical ALS, respectively. These observations are further complicated by the detection of phosphorylated tau, accentuating the multifaceted proteinopathic nature of ALS/PDC. The etiological foundations of this disease remain undetermined, and genetic investigations have yet to provide conclusive answers. However, emerging evidence has implicated the contribution of astrocytes, pivotal cells for maintaining brain health, to neurodegenerative onset, and likely to play a significant role in the pathogenesis of ALS/PDC. Leveraging advanced induced pluripotent stem cell technology, our team cultivated multiple astrocyte lines to further investigate the Japanese variant of ALS/PDC (Kii ALS/PDC). CHCHD2 emerged as a significantly dysregulated gene when disease astrocytes were compared to healthy controls. Our analyses also revealed imbalances in the activation of specific pathways: those associated with astrocytic cilium dysfunction, known to be involved in neurodegeneration, and those related to major neurological disorders, including classical ALS and PD. Further in-depth examinations revealed abnormalities in the mitochondrial morphology and metabolic processes of the affected astrocytes. A particularly striking observation was the reduced expression of CHCHD2 in the spinal cord, motor cortex, and oculomotor nuclei of patients with Kii ALS/PDC. In summary, our findings suggest a potential reduction in the support Kii ALS/PDC astrocytes provide to neurons, emphasizing the need to explore the role of CHCHD2 in maintaining mitochondrial health and its implications for the disease.


Subject(s)
Amyotrophic Lateral Sclerosis , Astrocytes , DNA-Binding Proteins , Mitochondrial Proteins , Transcription Factors , Astrocytes/pathology , Astrocytes/metabolism , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Humans , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Mitochondria/pathology , Mitochondria/metabolism , Male , Female , Middle Aged , Aged
2.
Science ; 378(6619): eadc9020, 2022 11 04.
Article in English | MEDLINE | ID: mdl-36378959

ABSTRACT

Astrocytes, a type of glia, are abundant and morphologically complex cells. Here, we report astrocyte molecular profiles, diversity, and morphology across the mouse central nervous system (CNS). We identified shared and region-specific astrocytic genes and functions and explored the cellular origins of their regional diversity. We identified gene networks correlated with astrocyte morphology, several of which unexpectedly contained Alzheimer's disease (AD) risk genes. CRISPR/Cas9-mediated reduction of candidate genes reduced astrocyte morphological complexity and resulted in cognitive deficits. The same genes were down-regulated in human AD, in an AD mouse model that displayed reduced astrocyte morphology, and in other human brain disorders. We thus provide comprehensive molecular data on astrocyte diversity and mechanisms across the CNS and on the molecular basis of astrocyte morphology in health and disease.


Subject(s)
Alzheimer Disease , Astrocytes , Central Nervous System , Transcriptome , Animals , Humans , Mice , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Astrocytes/classification , Astrocytes/metabolism , Astrocytes/ultrastructure , Disease Models, Animal , Central Nervous System/cytology , Central Nervous System/metabolism
3.
Acta Neuropathol Commun ; 9(1): 1, 2021 01 05.
Article in English | MEDLINE | ID: mdl-33402227

ABSTRACT

Microglia-mediated neuroinflammation has been implicated in the pathogenesis of Alzheimer's disease (AD). Although microglia in aging and neurodegenerative disease model mice show a loss of homeostatic phenotype and activation of disease-associated microglia (DAM), a correlation between those phenotypes and the degree of neuronal cell loss has not been clarified. In this study, we performed RNA sequencing of microglia isolated from three representative neurodegenerative mouse models, AppNL-G-F/NL-G-F with amyloid pathology, rTg4510 with tauopathy, and SOD1G93A with motor neuron disease by magnetic activated cell sorting. In parallel, gene expression patterns of the human precuneus with early Alzheimer's change (n = 11) and control brain (n = 14) were also analyzed by RNA sequencing. We found that a substantial reduction of homeostatic microglial genes in rTg4510 and SOD1G93A microglia, whereas DAM genes were uniformly upregulated in all mouse models. The reduction of homeostatic microglial genes was correlated with the degree of neuronal cell loss. In human precuneus with early AD pathology, reduced expression of genes related to microglia- and oligodendrocyte-specific markers was observed, although the expression of DAM genes was not upregulated. Our results implicate a loss of homeostatic microglial function in the progression of AD and other neurodegenerative diseases. Moreover, analyses of human precuneus also suggest loss of microglia and oligodendrocyte functions induced by early amyloid pathology in human.


Subject(s)
Alzheimer Disease/genetics , Amyotrophic Lateral Sclerosis/genetics , Microglia/metabolism , Parietal Lobe/metabolism , Tauopathies/genetics , Transcriptome , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Case-Control Studies , Homeostasis/genetics , Humans , Mice , Mice, Transgenic , Microglia/pathology , Parietal Lobe/pathology , RNA-Seq , Superoxide Dismutase/genetics , Tauopathies/metabolism , Tauopathies/pathology
4.
Mol Brain ; 13(1): 147, 2020 11 13.
Article in English | MEDLINE | ID: mdl-33183323

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by cognitive decline with accumulation of amyloid beta (Aß) and neurofibrillary tangles that usually begins 15-30 years before clinical diagnosis. Rodent models that recapitulate aggressive Aß and/or the pathology of neurofibrillary tangles are essential for AD research. Accordingly, non-invasive early detection systems in these animal models are required to evaluate the phenotypic changes, elucidate the mechanism of disease progression, and facilitate development of novel therapeutic approaches. Although many behavioral tests efficiently reveal cognitive impairments at the later stage of the disease in AD models, it has been challenging to detect such impairments at the early stage. To address this issue, we subjected 4-6-month-old male AppNL-G-F/NL-G-F knock-in (App-KI) mice to touchscreen-based location discrimination (LD), different object-location paired-associate learning (dPAL), and reversal learning tests, and compared the results with those of the classical Morris water maze test. These tests are mainly dependent on the brain regions prone to Aß accumulation at the earliest stages of the disease. At 4-6 months, considered to represent the early stage of disease when mice exhibit initial deposition of Aß and slight gliosis, the classical Morris water maze test revealed no difference between groups, whereas touchscreen-based LD and dPAL tasks revealed significant impairments in task performance. Our report is the first to confirm that a systematic touchscreen-based behavioral test battery can sensitively detect the early stage of cognitive decline in an AD-linked App-KI mouse model. This system could be applied in future translational research.


Subject(s)
Alzheimer Disease/complications , Amyloid beta-Protein Precursor/metabolism , Cognitive Dysfunction/complications , Discrimination Learning , Gene Knock-In Techniques , Paired-Associate Learning , Task Performance and Analysis , Alzheimer Disease/physiopathology , Animals , Astrocytes/pathology , Cognitive Dysfunction/physiopathology , Disease Models, Animal , Maze Learning , Mice, Inbred C57BL , Mice, Transgenic , Microglia/pathology , Neurogenesis , Neuroglia/metabolism , Neuroglia/pathology , Plaque, Amyloid/complications , Plaque, Amyloid/pathology , Spatial Memory
5.
Mol Brain ; 13(1): 8, 2020 01 20.
Article in English | MEDLINE | ID: mdl-31959210

ABSTRACT

Abnormal accumulation of TAR DNA-binding protein 43 (TDP-43), a DNA/RNA binding protein, is a pathological signature of amyotrophic lateral sclerosis (ALS). Missense mutations in the TARDBP gene are also found in inherited and sporadic ALS, indicating that dysfunction in TDP-43 is causative for ALS. To model TDP-43-linked ALS in rodents, we generated TDP-43 knock-in mice with inherited ALS patient-derived TDP-43M337V mutation. Homozygous TDP-43M337V mice developed normally without exhibiting detectable motor dysfunction and neurodegeneration. However, splicing of mRNAs regulated by TDP-43 was deregulated in the spinal cords of TDP-43M337V mice. Together with the recently reported TDP-43 knock-in mice with ALS-linked mutations, our finding indicates that ALS patient-derived mutations in the TARDBP gene at a carboxyl-terminal domain of TDP-43 may cause a gain of splicing function by TDP-43, however, were insufficient to induce robust neurodegeneration in mice.


Subject(s)
Alternative Splicing/physiology , DNA-Binding Proteins/genetics , Mutation, Missense , Point Mutation , Alternative Splicing/genetics , Amyotrophic Lateral Sclerosis/genetics , Animals , Base Sequence , Brain/metabolism , CRISPR-Cas Systems , DNA-Binding Proteins/physiology , Exons/genetics , Gene Knock-In Techniques , Humans , Mice , RNA, Messenger/metabolism , Spinal Cord/metabolism
6.
Acta Neuropathol Commun ; 7(1): 118, 2019 07 25.
Article in English | MEDLINE | ID: mdl-31345270

ABSTRACT

Intracellular mislocalization of TAR DNA-binding protein 43 (TDP-43), a nuclear DNA/RNA-binding protein involved in RNA metabolism, is a pathological hallmark of amyotrophic lateral sclerosis (ALS). Although the aggregation-prone, TDP-43 C-terminal domain is widely considered as a key component of TDP-43 pathology in ALS, recent studies including ours suggest that TDP-43 N-terminal fragments (TDP-∆C) may also contribute to the motor dysfunction in ALS. However, the specific pathological functions of TDP-43 N-terminal fragments in mice have not been elucidated. Here, we established TDP-∆C knock-in mice missing a part of exon 6 of murine Tardbp gene, which encodes the C-terminal region of TDP-43. Homozygous TDP-∆C mice showed embryonic lethality, indicating that the N-terminal domain of TDP-43 alone is not sufficient for normal development. In contrast, heterozygous TDP-∆C mice developed normally but exhibited age-dependent mild motor dysfunction with a loss of C-boutons, large cholinergic synaptic terminals on spinal α-motor neurons. TDP-∆C protein broadly perturbed gene expression in the spinal cords of aged heterozygous TDP-∆C mice, including downregulation of Notch1 mRNA. Moreover, the level of Notch1 mRNA was suppressed both by TDP-43 depletion and TDP-∆C expression in Neuro2a cells. Decreased Notch1 mRNA expression in aged TDP-∆C mice was associated with the age-dependent motor dysfunction and loss of Akt surviving signal. Our findings indicate that the N-terminal region of TDP-43 derived from TDP-∆C induces the age-dependent motor dysfunction associated with impaired Notch1-Akt axis in mice.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , DNA-Binding Proteins/deficiency , Proto-Oncogene Proteins c-akt/biosynthesis , Receptor, Notch1/biosynthesis , Signal Transduction/physiology , Age Factors , Amyotrophic Lateral Sclerosis/genetics , Animals , Cell Line, Tumor , Cells, Cultured , DNA-Binding Proteins/genetics , Gene Knock-In Techniques , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-akt/genetics , Receptor, Notch1/genetics
7.
Cell Death Differ ; 25(12): 2130-2146, 2018 12.
Article in English | MEDLINE | ID: mdl-29568058

ABSTRACT

There is compelling evidence that glial-immune interactions contribute to the progression of neurodegenerative diseases. The adaptive immune response has been implicated in disease processes of amyotrophic lateral sclerosis (ALS), but it remains unknown if innate immune signaling also contributes to ALS progression. Here we report that deficiency of the innate immune adaptor TIR domain-containing adaptor inducing interferon-ß (TRIF), which is essential for certain Toll-like receptor (TLR) signaling cascades, significantly shortens survival time and accelerates disease progression of ALS mice. While myeloid differentiation factor 88 (MyD88) is also a crucial adaptor for most TLR signaling pathways, MyD88 deficiency had only a marginal impact on disease course. Moreover, TRIF deficiency reduced the number of natural killer (NK), NK-T-lymphocytes, and CD8-T cells infiltrating into the spinal cord of ALS mice, but experimental modulation of these populations did not substantially influence survival time. Instead, we found that aberrantly activated astrocytes expressing Mac2, p62, and apoptotic markers were accumulated in the lesions of TRIF-deficient ALS mice, and that the number of aberrantly activated astrocytes was negatively correlated with survival time. These findings suggest that TRIF pathway plays an important role in protecting a microenvironment surrounding motor neurons by eliminating aberrantly activated astrocytes.


Subject(s)
Adaptor Proteins, Vesicular Transport/deficiency , Adaptor Proteins, Vesicular Transport/immunology , Amyotrophic Lateral Sclerosis/pathology , Astrocytes/pathology , Immunity, Innate , Adaptor Proteins, Vesicular Transport/genetics , Amyotrophic Lateral Sclerosis/immunology , Animals , Astrocytes/immunology , Disease Progression , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
8.
J Neurochem ; 145(1): 80-89, 2018 04.
Article in English | MEDLINE | ID: mdl-29282717

ABSTRACT

Cystatin C (CysC) is a major protein component of Bunina bodies, which are a pathological hallmark observed in the remaining motor neurons of patients with amyotrophic lateral sclerosis (ALS). Dominant mutations in the SOD1 gene, encoding Cu/Zn superoxide dismutase (SOD1), are causative for a subset of inherited ALS cases. Our previous study showed that CysC exerts a neuroprotective effect against mutant SOD1-mediated toxicity in vitro; however, in vivo evidence of the beneficial effects mediated by CysC remains obscure. Here we examined the therapeutic potential of recombinant human CysC in vivo using a mouse model of ALS in which the ALS-linked mutated SOD1 gene is expressed (SOD1G93A mice). Intracerebroventricular administration of CysC during the early symptomatic SOD1G93A mice extended their survival times. Administered CysC was predominantly distributed in ventral horn neurons including motor neurons, and induced autophagy through AMP-activated kinase activation to reduce the amount of insoluble mutant SOD1 species. Moreover, PGC-1α, a disease modifier of ALS, was restored by CysC through AMP-activated kinase activation. Finally, the administration of CysC also promoted aggregation of CysC in motor neurons, which is similar to Bunina bodies. Taken together, our findings suggest that CysC represents a promising therapeutic candidate for ALS.


Subject(s)
Amyotrophic Lateral Sclerosis , Cystatin C/pharmacology , Motor Neurons/drug effects , Neuroprotective Agents/pharmacology , Animals , Autophagy/drug effects , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Transgenic , Mutation , Recombinant Proteins/pharmacology , Superoxide Dismutase-1/genetics
9.
Protein Sci ; 26(3): 484-496, 2017 03.
Article in English | MEDLINE | ID: mdl-27977888

ABSTRACT

Misfolding of mutant Cu/Zn-superoxide dismutase (SOD1) is a pathological hallmark in a familial form of amyotrophic lateral sclerosis. Pathogenic mutations have been proposed to monomerize SOD1 normally adopting a homodimeric configuration and then trigger abnormal oligomerization of SOD1 proteins. Despite this, a misfolded conformation of SOD1 leading to the oligomerization at physiological conditions still remains ambiguous. Here, we show that, around the body temperature (∼37°C), mutant SOD1 maintains a dimeric configuration but lacks most of its secondary structures. Also, such an abnormal SOD1 dimer with significant structural disorder was prone to irreversibly forming the oligomers crosslinked via disulfide bonds. The disulfide-crosslinked oligomers of SOD1 were detected in the spinal cords of the diseased mice expressing mutant SOD1. We hence propose an alternative pathway of mutant SOD1 misfolding that is responsible for oligomerization in the pathologies of the disease.


Subject(s)
Amyotrophic Lateral Sclerosis , Protein Folding , Protein Multimerization , Superoxide Dismutase-1 , Amyotrophic Lateral Sclerosis/enzymology , Amyotrophic Lateral Sclerosis/genetics , Animals , Disease Models, Animal , Disulfides/chemistry , Disulfides/metabolism , Humans , Mice , Mice, Transgenic , Superoxide Dismutase-1/chemistry , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism
10.
EMBO Mol Med ; 8(12): 1421-1437, 2016 12.
Article in English | MEDLINE | ID: mdl-27821430

ABSTRACT

A homozygous mutation in the gene for sigma 1 receptor (Sig1R) is a cause of inherited juvenile amyotrophic lateral sclerosis (ALS16). Sig1R localizes to the mitochondria-associated membrane (MAM), which is an interface of mitochondria and endoplasmic reticulum. However, the role of the MAM in ALS is not fully elucidated. Here, we identified a homozygous p.L95fs mutation of Sig1R as a novel cause of ALS16. ALS-linked Sig1R variants were unstable and incapable of binding to inositol 1,4,5-triphosphate receptor type 3 (IP3R3). The onset of mutant Cu/Zn superoxide dismutase (SOD1)-mediated ALS disease in mice was accelerated when Sig1R was deficient. Moreover, either deficiency of Sig1R or accumulation of mutant SOD1 induced MAM disruption, resulting in mislocalization of IP3R3 from the MAM, calpain activation, and mitochondrial dysfunction. Our findings indicate that a loss of Sig1R function is causative for ALS16, and collapse of the MAM is a common pathomechanism in both Sig1R- and SOD1-linked ALS Furthermore, our discovery of the selective enrichment of IP3R3 in motor neurons suggests that integrity of the MAM is crucial for the selective vulnerability in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Endoplasmic Reticulum/physiology , Mitochondrial Membranes/physiology , Receptors, sigma/genetics , Animals , Child , Female , Humans , Mice , Superoxide Dismutase-1/genetics , Sigma-1 Receptor
11.
Acta Neuropathol Commun ; 4: 15, 2016 Feb 18.
Article in English | MEDLINE | ID: mdl-26891847

ABSTRACT

INTRODUCTION: Increasing evidence implicates the role of the cell types surrounding motor neurons, such as interneurons and glial cells, in non-cell autonomous neurodegeneration of amyotrophic lateral sclerosis (ALS). C-boutons, the large cholinergic synapses that innervate spinal α-motor neurons to control their excitability, are progressively lost from motor neurons in both human ALS and mutant Cu/Zn superoxide dismutase 1 (SOD1)-ALS mice. Neuregulin-1 (NRG1), a trophic factor implicated in neural development, transmission, and synaptic plasticity, has been reported to localize in the synapse of C-boutons. However, the roles of NRG1 in maintenance of motor neuron health and activity, as well as the functional consequences of its alteration in motor neuron disease, are not fully understood. RESULTS: NRG1 was localized to the post-synaptic face of C-boutons and its expression was significantly lost in SOD1-ALS mice and human ALS patients. Losses of NRG1 expression and C-boutons occurred almost contemporaneously in SOD1-ALS mice. In addition, expressions of ErbB3 and ErbB4, receptors for NRG1, were reduced in the motor neurons of SOD1-ALS mice. Furthermore, viral-mediated delivery of type III-NRG1 to the spinal cord restored the number of C-boutons and extended the survival time of SOD1-ALS mice. CONCLUSIONS: These results suggest that maintenance of NRG1-ErbB4/3 axis by supplementation of NRG1 confers neuroprotection in motor neuron disease, partly through the maintenance of C-boutons of spinal motor neurons.


Subject(s)
Amyotrophic Lateral Sclerosis , Motor Neurons/pathology , Neuregulin-1/metabolism , Neuroprotection/physiology , Presynaptic Terminals/metabolism , Spinal Cord/pathology , Adult , Age Factors , Aged , Aged, 80 and over , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Disease Models, Animal , Female , Gene Expression Regulation/genetics , Humans , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Motor Neurons/metabolism , Mutation/genetics , Nerve Tissue Proteins/metabolism , Postmortem Changes , Receptor, ErbB-3/metabolism , Shab Potassium Channels/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , Vesicular Acetylcholine Transport Proteins/metabolism
12.
Neurobiol Dis ; 86: 16-28, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26607786

ABSTRACT

Proteotoxicity of misfolded, disease-causing proteins is deeply implicated in the pathomechanisms for neurodegenerative diseases including copper-zinc superoxide dismutase (SOD1)-linked amyotrophic lateral sclerosis (ALS). However, the precise cellular quality control (QC) mechanisms against aggregation of misfolded mutant SOD1 proteins remain elusive. Here, we found that the Mahogunin ring finger-1 (MGRN1) E3 ubiquitin ligase, which catalyzes mono-ubiquitination to the substrate, was dysregulated in the cellular and mouse models of ALS and that it preferentially interacted with various mutant forms of SOD1. Intriguingly, the motor neurons of presymptomatic ALS mice have diminished MGRN1 cytoplasmic distribution. MGRN1 was partially recruited to mutant SOD1 inclusions where they were positive for p62 and Lamp2. Moreover, overexpression of MGRN1 reduced mutant SOD1 aggregation and alleviated its proteotoxic effects on cells. Taken together, our findings suggest that MGRN1 contributes to the clearance of toxic mutant SOD1 inclusions likely through autophagic pathway, and, most likely, the sequestration of MGRN1 sensitizes motor neurons to degeneration in the ALS mouse model. Furthermore, the present study identifies the MGRN1-mediated protein QC mechanism as a novel therapeutic target in neurodegenerative diseases.


Subject(s)
Amyotrophic Lateral Sclerosis/enzymology , Amyotrophic Lateral Sclerosis/pathology , Motor Neurons/metabolism , Motor Neurons/pathology , Superoxide Dismutase/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , COS Cells , Cell Survival , Chlorocebus aethiops , Disease Models, Animal , HEK293 Cells , Humans , Mice , Mice, Transgenic , Spinal Cord/metabolism , Spinal Cord/pathology , Superoxide Dismutase/genetics , Superoxide Dismutase-1
15.
Cell Rep ; 11(4): 592-604, 2015 Apr 28.
Article in English | MEDLINE | ID: mdl-25892237

ABSTRACT

Neuroinflammation, which includes both neuroprotective and neurotoxic reactions by activated glial cells and infiltrated immune cells, is involved in the pathomechanism of amyotrophic lateral sclerosis (ALS). However, the cytokines that regulate the neuroprotective inflammatory response in ALS are not clear. Here, we identify transforming growth factor-ß1 (TGF-ß1), which is upregulated in astrocytes of murine and human ALS, as a negative regulator of neuroprotective inflammatory response. We demonstrate that astrocyte-specific overproduction of TGF-ß1 in SOD1(G93A) mice accelerates disease progression in a non-cell-autonomous manner, with reduced IGF-I production in deactivated microglia and fewer T cells with an IFN-γ-dominant milieu. Moreover, expression levels of endogenous TGF-ß1 in SOD1(G93A) mice negatively correlate with lifespan. Furthermore, pharmacological administration of a TGF-ß signaling inhibitor after disease onset extends survival time of SOD1(G93A) mice. These findings indicate that astrocytic TGF-ß1 determines disease progression and is critical to the pathomechanism of ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Astrocytes/metabolism , T-Lymphocytes/metabolism , Transforming Growth Factor beta1/metabolism , Amyotrophic Lateral Sclerosis/genetics , Animals , Cells, Cultured , Interferon-gamma/metabolism , Mice , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , Transforming Growth Factor beta1/antagonists & inhibitors , Transforming Growth Factor beta1/genetics
16.
Mol Brain ; 7: 62, 2014 Aug 29.
Article in English | MEDLINE | ID: mdl-25167838

ABSTRACT

BACKGROUND: Dominant mutations in superoxide dismutase 1 (SOD1) cause degeneration of motor neurons in a subset of inherited amyotrophic lateral sclerosis (ALS). The pathogenetic process mediated by misfolded and/or aggregated mutant SOD1 polypeptides is hypothesized to be suppressed by protein refolding. This genetic study is aimed to test whether mutant SOD1-mediated ALS pathology recapitulated in mice could be alleviated by overexpressing a longevity-related deacetylase SIRT1 whose substrates include a transcription factor heat shock factor 1 (HSF1), the master regulator of the chaperone system. RESULTS: We established a line of transgenic mice that chronically overexpress SIRT1 in the brain and spinal cord. While inducible HSP70 (HSP70i) was upregulated in the spinal cord of SIRT1 transgenic mice (PrP-Sirt1), no neurological and behavioral alterations were detected. To test hypothetical benefits of SIRT1 overexpression, we crossbred PrP-Sirt1 mice with two lines of ALS model mice: A high expression line that exhibits a severe phenotype (SOD1G93A-H) or a low expression line with a milder phenotype (SOD1G93A-L). The Sirt1 transgene conferred longer lifespan without altering the time of symptomatic onset in SOD1G93A-L. Biochemical analysis of the spinal cord revealed that SIRT1 induced HSP70i expression through deacetylation of HSF1 and that SOD1G93A-L/PrP-Sirt1 double transgenic mice contained less insoluble SOD1 than SOD1G93A-L mice. Parallel experiments showed that Sirt1 transgene could not rescue a more severe phenotype of SOD1G93A-H transgenic mice partly because their HSP70i level had peaked out. CONCLUSIONS: The genetic supplementation of SIRT1 can ameliorate a mutant SOD1-linked ALS mouse model partly through the activation of the HSF1/HSP70i chaperone system. Future studies shall include testing potential benefits of pharmacological enhancement of the deacetylation activity of SIRT1 after the onset of the symptom.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , DNA-Binding Proteins/metabolism , HSP70 Heat-Shock Proteins/metabolism , Sirtuin 1/metabolism , Superoxide Dismutase/genetics , Transcription Factors/metabolism , Acetylation , Animals , Behavior, Animal , Disease Models, Animal , Gene Dosage , Heat Shock Transcription Factors , Humans , Longevity , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Promoter Regions, Genetic/genetics , Protein Folding , Spinal Cord/pathology , Superoxide Dismutase-1 , Up-Regulation
17.
Rinsho Shinkeigaku ; 54(12): 1128-31, 2014.
Article in Japanese | MEDLINE | ID: mdl-25672727

ABSTRACT

Neuroinflammation, characterized by activated astrocytes and microglia, infiltrated T cells, and the subsequent production of inflammatory mediators, is a pathological hallmark of amyotrophic lateral sclerosis (ALS). Since microglia produce proinflammatory cytokines and other neurotoxic or protective molecules and astrocytes reduce their glutamate uptake, these functional changes in glial cells are considered to play important roles in neurodegeneration. However, what regulates the neuroprotective response in neuroinflammation of ALS has not been clarified. In this regard, we identify astrocyte-derived transforming growth factor-ß1 (TGF-ß1) as a negative regulator of neuroprotective inflammatory response by microglia and T cells. Understanding the pathomechanism of neuroinflammation of ALS leads to the development of new therapies that target glial cells and T cells.


Subject(s)
Amyotrophic Lateral Sclerosis/etiology , Animals , Disease Progression , Inflammation , Mice , Microglia/physiology
18.
Neurobiol Aging ; 34(4): 1310.e11-23, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23040663

ABSTRACT

Protein aggregation and ordered fibrillar amyloid deposition inside and outside of the central nervous system cells is the common pathologic hallmark of most aging-related neurodegenerative disorders. Dominant mutations in the gene encoding superoxide dismutase 1 (SOD1) protein are linked to familial amyotrophic lateral sclerosis (ALS), a neurodegenerative disease characterized by progressive degeneration of motor neurons, leading to muscle paralysis and death. The major histochemical hallmark in the remaining motor neurons of ALS is the intracellular accumulation of ubiquitinated inclusions consisting of insoluble aberrant protein aggregates. However, the molecular pathomechanisms underlying the process have been elusive. Here for the first time, we report that E6-AP, a homologous to E6-AP C terminus-type E3 ubiquitin ligase depleted in ALS mouse models before neurodegeneration. E6-AP coimmunoprecipitates with the SOD1 protein and is predominantly mislocalized in mutant SOD1-containing inclusion bodies. Overexpression of E6-AP increases the ubiquitination and facilitates degradation of SOD1 proteins. Finally, we show that the overexpression of E6-AP suppresses the aggregation and cell death mediated by mutated SOD1 proteins and cellular protective effect is more prominent when E6-AP is overexpressed along with Hsp70. These data suggest that enhancing the activity of E6-AP ubiquitin ligase might be a viable therapeutic strategy to eliminate mutant SOD1-mediated toxicity in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Inclusion Bodies/metabolism , Neurons/metabolism , Superoxide Dismutase/metabolism , Ubiquitin-Protein Ligases/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Cell Aggregation , Cell Survival , Cells, Cultured , Mice , Mice, Transgenic , Superoxide Dismutase-1
19.
Magn Reson Med Sci ; 7(3): 163-7, 2008.
Article in English | MEDLINE | ID: mdl-18827459

ABSTRACT

We investigated 2 Japanese siblings with a complicated form of familial spastic paraplegia. Cranial magnetic resonance (MR) imaging revealed marked thinning of the corpus callosum. Diffusion tensor imaging (DTI) showed microstructural changes in the thalamus, basal ganglia, and cerebral white matter, and single photon emission computed tomography (SPECT) using 99mTc-ethylcysteinate dimer showed very similar findings. DTI and SPECT effectively revealed global changes not revealed by conventional MR imaging.


Subject(s)
Cognition Disorders/diagnosis , Corpus Callosum/pathology , Diffusion Magnetic Resonance Imaging/methods , Paraplegia/congenital , Paraplegia/diagnosis , Adolescent , Adult , Cognition Disorders/genetics , Humans , Male , Reproducibility of Results , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...