636 results on '"Takaomi C. Saido"'
Search Results
602. Cerebrospinal fluid neprilysin is reduced in prodromal Alzheimer's disease.
- Author
-
Masahiro Maruyama, Makoto Higuchi, Yoshie Takaki, Yukio Matsuba, Haruko Tanji, Miyako Nemoto, Naoki Tomita, Toshifumi Matsui, Nobuhisa Iwata, Hiroaki Mizukami, Shin‐Ichi Muramatsu, Keiya Ozawa, Takaomi C. Saido, Hiroyuki Arai, and Hidetada Sasaki
- Published
- 2005
603. Involvement of proteases in glycosyltransferase secretion: Alzheimer's β-secretase-dependent cleavage and a following processing by an aminopeptidase.
- Author
-
Shinobu Kitazume, Minoru Suzuki, Takaomi C. Saido, and Yasuhiro Hashimoto
- Published
- 2004
- Full Text
- View/download PDF
604. The crucial role of caspase-9 in the disease progression of a transgenic ALS mouse model.
- Author
-
Haruhisa Inoue, Kayoko Tsukita, Takuji Iwasato, Yasuyuki Suzuki, Masanori Tomioka, Minako Tateno, Masahiro Nagao, Akihiro Kawata, Takaomi C. Saido, Masayuki Miura, Hidemi Misawa, Shigeyoshi Itohara, and Ryosuke Takahashi
- Subjects
GENETIC mutation ,PEROXIDES ,AMYOTROPHIC lateral sclerosis ,THERAPEUTICS - Abstract
Mutant copper/zinc superoxide dismutase (SOD1)-overexpressing transgenic mice, a mouse model for familial amyotrophic lateral sclerosis (ALS), provides an excellent resource for developing novel therapies for ALS. Several observations suggest that mitochondria-dependent apoptotic signaling, including caspase-9 activation, may play an important role in mutant SOD1-related neurodegeneration. To elucidate the role of caspase-9 in ALS, we examined the effects of an inhibitor of X chromosome-linked inhibitor of apoptosis (XIAP), a mammalian inhibitor of caspase-3, -7 and -9, and p35, a baculoviral broad caspase inhibitor that does not inhibit caspase-9. When expressed in spinal motor neurons of mutant SOD1 mice using transgenic techniques, XIAP attenuated disease progression without delaying onset. In contrast, p35 delayed onset without slowing disease progression. Moreover, caspase-9 was activated in spinal motor neurons of human ALS subjects. These data strongly suggest that caspase-9 plays a crucial role in disease progression of ALS and constitutes a promising therapeutic target. [ABSTRACT FROM AUTHOR]
- Published
- 2003
- Full Text
- View/download PDF
605. Analysis of the processing of Alzheimer amyloid precursor protein in culture cells by cDNA transfection
- Author
-
Takaomi C. Saido, Seiichi Kawashima, Yumiko Saito, Kei Maruyama, Yoshihiro Kumagae, Wakako Harigaya, and Mihoko Usami
- Subjects
Pharmacology ,biology ,Chemistry ,Cdna transfection ,Amyloid precursor protein ,biology.protein ,Molecular biology - Published
- 1993
- Full Text
- View/download PDF
606. Activation of calpain at the cell membrane and its physiological implications
- Author
-
Takaomi C. Saido, Koichi Suzuki, Yoshiko Akita, Akiko Hata, Ohiogo Ohno, and Hiroshi Kawasaki
- Subjects
Pharmacology ,Cell membrane ,medicine.anatomical_structure ,biology ,Chemistry ,biology.protein ,medicine ,Calpain ,Cell biology - Published
- 1991
- Full Text
- View/download PDF
607. Induction of increased calcium uptake in liposomes having membrane proteins of chicken erythrocytes by S-adenosylmethionine
- Author
-
Fusao Makishima, Toshiaki Osawa, Takaomi C. Saido, and Satoshi Toyoshima
- Subjects
S-Adenosylmethionine ,Cell Membrane Permeability ,Erythrocytes ,Biophysics ,chemistry.chemical_element ,Calcium ,Biology ,Methylation ,Biochemistry ,Animals ,Molecular Biology ,Phospholipids ,chemistry.chemical_classification ,Liposome ,Erythrocyte Membrane ,Peripheral membrane protein ,Membrane Proteins ,Biological Transport ,Cell Biology ,Aminoisobutyric acid ,Enzyme ,Membrane ,Membrane protein ,chemistry ,Liposomes ,Chickens - Abstract
Liposomes having membrane proteins of chicken erythrocytes were prepared and the effect of S-adenosylmethionine on 45Ca2+ uptake into the liposomes was investigated. S-Adenosylmethionine, a donor of methyl groups in enzymatic methylation, induced an increase of 45Ca2+ uptake into the proteoliposomes with membrane proteins but not into the liposomes without membrane proteins. Increased release of 45Ca2+ from the inside of the proteoliposomes was also induced by S-adenosylmethionine. These increases of uptake and release of 45Ca2+ were inhibited by S-adenosylhomocystein, an inhibitor of enzymatic methylation. Furthermore, membrane proteins from chicken erythrocytes showed protein and phospholipid methyltransferase activities. The uptake of other materials, 3-0-[methyl-3H]glucose, α-[1-14C]aminoisobutyric acid, 42K+ and 54Mn2+, into the proteoliposomes was not increased by S-adenosylmethionine. These results suggest that enzymatic methylation of membrane components may have an important role in the regulation of calcium transport in the chicken erythrocyte membrane and this regulation is rather specific for calcium.
- Published
- 1983
- Full Text
- View/download PDF
608. Cognitive deficits in single App knock-in mouse models
- Author
-
Yuki Kobayashi, Takaomi C. Saido, Takashi Saito, Shigeyoshi Itohara, Akira Masuda, and Naomi Kogo
- Subjects
0301 basic medicine ,Genetically modified mouse ,Male ,Cognitive Neuroscience ,Mice, Transgenic ,Experimental and Cognitive Psychology ,Disease ,IntelliCage ,03 medical and health sciences ,Amyloid beta-Protein Precursor ,Executive Function ,Mice ,Behavioral Neuroscience ,0302 clinical medicine ,Cognition ,Alzheimer Disease ,Behavior monitoring ,Amyloid precursor protein ,medicine ,Animals ,Learning ,Cognitive Dysfunction ,Knock in mouse ,Spatial Memory ,biology ,Behavior, Animal ,Pathophysiology ,Disease Models, Animal ,030104 developmental biology ,Compulsive behavior ,biology.protein ,Female ,Sex ,medicine.symptom ,Psychology ,Neuroscience ,Alzheimer’s disease ,030217 neurology & neurosurgery ,Model mouse - Abstract
Transgenic mouse models of Alzheimer’s disease (AD) with nonphysiologic overexpression of amyloid precursor protein (APP) exhibit various unnatural symptoms/dysfunctions. To overcome this issue, mice with single humanized App knock-in (KI) carrying Swedish (NL), Beyreuther/Iberian (F), and Arctic (G) mutations in different combinations were recently developed. The validity of these mouse models of AD from a behavioral viewpoint, however, has not been extensively evaluated. Thus, using an automated behavior monitoring system, we analyzed various behavioral domains, including executive function, and learning and memory. The App-KI mice carrying NL-G-F mutations showed clear deficits in spatial memory and flexible learning, enhanced compulsive behavior, and reduced attention performance. Mice carrying NL-F mutations exhibited modest abnormalities. The NL-G-F mice had a greater and more rapid accumulation of Aβ deposits and glial responses. These findings reveal that single pathologic App-KI is sufficient to produce deficits in broad cognitive domains and that App-KI mouse lines with different levels of pathophysiology are useful models of AD.
- Full Text
- View/download PDF
609. New era of calpain research Discovery of tissue-specific calpains
- Author
-
Koichi Suzuki, Hiroyuki Sorimachi, and Takaomi C. Saido
- Subjects
Cysteine Endopeptidases ,Evolution ,Protein subunit ,Molecular Sequence Data ,Biophysics ,Biochemistry ,Isozyme ,Substrate Specificity ,Species Specificity ,Structural Biology ,Molecular diversity ,Gene expression ,Genetics ,medicine ,Animals ,Humans ,Tissue specific ,Amino Acid Sequence ,Molecular Biology ,Peptide sequence ,Phylogeny ,biology ,Calpain ,Muscles ,Stomach ,Skeletal muscle ,Cell Biology ,Novel species ,medicine.anatomical_structure ,biology.protein ,Tissue-specific expression - Abstract
The recent discovery of several new calpain species other than the two species thus far studied reveals that calpain, especially the calpain large subunit, constitutes a family comprising at least six members that can be classified into ubiquitous (μ-, m- and μm-types) and tissue-specific (p94 or nCL-1 specific for skeletal muscle, and nCL-2 and -2' specific for stomach) calpains. The newly identified tissue-specific calpains have various characteristics distinct from conventional calpains in structure, manner of expression, and enzyme activity. Unique features of tissuespecific calpains are discussed together with the evolutionary view of the calpain large subunit.
- Full Text
- View/download PDF
610. Protein kinase C isoforms in muscle cells and their regulation by phorbol ester and calpain
- Author
-
Neil E. Forsberg, Bor rung Ou, Dong hyun Hong, Takaomi C. Saido, Jianya Huan, Jan ying Yeh, and Peter R. Cheeke
- Subjects
Phorbol ester ,Molecular Sequence Data ,Down-Regulation ,Biology ,chemistry.chemical_compound ,Protein kinase C ,Endopeptidases ,Phorbol Esters ,medicine ,Animals ,Myocyte ,Amino Acid Sequence ,Molecular Biology ,Cells, Cultured ,Myogenesis ,Calpain ,Muscles ,PKCS ,Muscle cell ,Skeletal muscle ,Cell Biology ,Molecular biology ,Rats ,Cell biology ,Isoenzymes ,Cytosol ,enzymes and coenzymes (carbohydrates) ,medicine.anatomical_structure ,chemistry ,Phorbol ,biology.protein ,biological phenomena, cell phenomena, and immunity ,Signal Transduction ,Subcellular Fractions - Abstract
Objectives were to identify the PKC isoforms in cultured muscle cells, to examine roles of Ca(2+)-dependent proteinases (calpains) in processing of various muscle PKC isozymes and to obtain a mechanistic description of the processing of PKCs by examining the temporal relationships between phorbol ester-dependent translocation of muscle PKCs and calpains between cytosolic and membrane compartments. Using six isoform (alpha, beta, gamma, delta, epsilon, zeta)-specific polyclonal antibodies, PKC alpha, delta and zeta were detected in rat skeletal muscle and in L8 myoblasts and myotubes. PKC alpha and zeta were primarily localized in the cytosolic fraction of L8 myotubes whereas PKC delta was more abundant in the membrane fraction. Phorbol ester (TPA) caused rapid depletion of myotube PKC alpha and PKC alpha and PKC delta isoforms from the cytosolic compartment and rapid appearance of these isoforms in the membrane fraction. However, long-term exposure of myotubes to TPA eventually caused down-regulation of PKCs in the membrane compartment. Down-regulation of PKCs in the membrane fraction was partially blocked by calpain inhibitor II. However, the rapid TPA-dependent cytosolic depletion of PKCs was unaffected by calpain inhibitor. This suggests that calpains may be responsible for membrane-associated down-regulation of PKCs but not for cytosolic depletion. In the final study we assessed the effects of phorbol ester on compartmentation of m-calpain with PKCs in muscle cells. Like the PKCs, TPA caused rapid association of m-calpain with the membrane fraction followed by down-regulation. This demonstrates that phorbol esters cause translocation of both PKCs and calpains to membranes where processing of PKCs may occur via the limited proteolysis exerted by calpains.
- Full Text
- View/download PDF
611. Staurosporine-related compounds, K252a and UCN-01, inhibit both cPKC and nPKC
- Author
-
Keiko Mizuno, Tatsuya Tamaoki, Shigeo Ohno, Takaomi C. Saido, and Koichi Suzuki
- Subjects
medicine.drug_class ,Biophysics ,Carbazoles ,PKC inhibitor ,Inhibitory postsynaptic potential ,Biochemistry ,Isozyme ,Indole Alkaloids ,Substrate Specificity ,chemistry.chemical_compound ,Novel PKC ,Alkaloids ,Structural Biology ,Protein kinase C ,Genetics ,medicine ,Staurosporine ,Animals ,Molecular Biology ,chemistry.chemical_classification ,biology ,Cell Biology ,Protein kinase inhibitor ,Molecular biology ,Isoenzymes ,Enzyme ,chemistry ,Enzyme inhibitor ,biology.protein ,Cattle ,K252a ,medicine.drug - Abstract
The potent inhibitors of protein kinase C (PKC), H7, staurosporine, and staurosporine derivatives, were examined for their inhibitory effects on novel PKC (nPKC) isozymes delta and epsilon. H7 and staurosporine, usually used as selective inhibitors of PKC, showed similar inhibitory effects on cPKC (a mixture of cPKC alpha, beta, and gamma) and nPKC delta and epsilon. The inhibitory effects of K252a, a non-selective protein kinase inhibitor, on cPKC was 3.2- and 22-fold higher than those on nPKC epsilon and delta, respectively. The staurosporine derivatives UCN-01 and UCN-01-Me also showed selective inhibition of cPKC.
- Full Text
- View/download PDF
612. Protein-O-carboxylmethyltransferase from cytosol and membranes of chicken erythrocytes
- Author
-
Takaomi C. Saido, Satoshi Toyoshima, and Toshiaki Osawa
- Subjects
Erythrocytes ,Size-exclusion chromatography ,Biochemistry ,Chromatography, Affinity ,Gel permeation chromatography ,Cytosol ,Affinity chromatography ,Animals ,Protein Methyltransferases ,Molecular Biology ,chemistry.chemical_classification ,Gel electrophoresis ,Chromatography ,Chemistry ,Erythrocyte Membrane ,Substrate (chemistry) ,General Medicine ,Protein O-Methyltransferase ,Fetuin ,Molecular Weight ,Kinetics ,Enzyme ,Chromatography, Gel ,Chickens - Abstract
Cytosolic protein-O-carboxylmethyltransferase was purified more than 4,000-fold in specific activity and membrane-associated protein-O-carboxylmethyltransferase carboxymethylase about 900-fold from chicken erythrocytes by use of a combination of affinity chromatography on immobilized S-adenosyl-L-homocysteine and gel filtration on Sephacryl S-200 (Pharmacia), together with 3-((3-cholamidopropyl)-dimethylammonio)-1-propane-sulfonate as a detergent to solubilize the membrane-associated enzyme. The two enzymes were characterized by examining the dependence of their activity on pH and on concentration of S-adenosyl-L-methionine using fetuin as an exogenous methyl-acceptor substrate, and were found to differ somewhat. The cytosolic enzyme had a pH optimum of 6.0 with an apparent Km value of 2.1 microM for S-adenosyl-L-methionine, whereas corresponding values for the membrane-associated enzyme were 6.5 and 0.71 microM. This report deals with the biochemical differences between purified cytosolic and membrane-associated protein carboxymethylase from the same cell source.
- Published
- 1987
613. The calpain proteolytic system in neonatal hypoxic-ischemia
- Author
-
Henrik Hagberg, Takaomi C. Saido, Elsa Bona, Tomio Ono, Klas Blomgren, Amanda McRae, Anna Elmered, and Seiichi Kawashima
- Subjects
Male ,medicine.medical_specialty ,Proteases ,Transcription, Genetic ,Proteolysis ,Endogeny ,Nerve Tissue Proteins ,Cysteine Proteinase Inhibitors ,Inhibitory postsynaptic potential ,General Biochemistry, Genetics and Molecular Biology ,Functional Laterality ,White matter ,Cytosol ,History and Philosophy of Science ,Internal medicine ,medicine ,Animals ,Rats, Wistar ,Hypoxia, Brain ,Calpastatin ,DNA Primers ,Cerebral Cortex ,medicine.diagnostic_test ,biology ,Chemistry ,Calpain ,General Neuroscience ,Calcium-Binding Proteins ,Microfilament Proteins ,Rats ,Blot ,Endocrinology ,medicine.anatomical_structure ,Animals, Newborn ,Ischemic Attack, Transient ,biology.protein ,Female ,Carrier Proteins - Abstract
Neonatal rats were subjected to transient cerebral hypoxic-ischemia (HI, unilateral occlusion of the common carotid artery +7.70% O2 for 100 min) and allowed to recover for up to 14 days. Calpain caseinolytic activity was found to increase in both hemispheres for at least 20 hr. Hypoxic exposure per se increased the activity of calpains, more pronounced in a membrane-associated fraction, probably through interaction with cellular components, whereas HI introduced a loss of activity, most likely through consumption and loss of proteases. Consecutive tissue sections were stained with antibodies against calpastatin, alpha-fodrin, the 150-kDa breakdown product of alpha-fodrin (FBDP, marker of calpain proteolysis) or microtubule-associated protein 2 (MAP-2, marker of dendrosomatic neuronal injury). Areas with brain injury displayed a distinct loss of MAP-2, which clearly delineated the infarct. FBDP accumulated in injured and borderline regions ipsilaterally, and a less conspicuous, transient increase in FBDP also occurred in the contralateral hemisphere, especially in the white matter. The cytosolic fraction (CF) and the membrane and microsomal fraction (MMF) of cortical tissue were subjected to Western blotting and stained with antibodies against calpain, calpastatin and the 150-kDa breakdown product of alpha-fodrin (FBDP). Calpain immunoreactivity decreased bilaterally in the CF during the insult (62-68% of controls) and remained significantly lower during early recovery, whereas the MMF showed no significant changes. This translocation of calpains coincided with the appearance of FBDP in the ipsilateral, HI hemisphere, displaying a significantly higher level of FBDP from immediately after the insult until at least 1 day of recovery (204-292% of controls). No significant changes in FBDP were found in the contralateral, undamaged hemisphere, despite translocation of calpains in both hemispheres, a prerequisite for calpain activation. This discrepancy may be related to changes in the endogenous inhibitor, calpastatin. Calpastatin protein was found to decrease during and shortly after HI in the ipsilateral, but not the contralateral, hemisphere. The inhibitory activity of calpastatin also tended to decrease after HI, indicating that a reduction of calpastatin may be necessary for extensive calpain activation to occur. The mRNA of m-calpain increased in the HI hemisphere 48 hr after the insult (167%, p < 0.001), a time point when the protein was also increased. In summary, our findings indicate that calpains are activated during HI and in the early phase of reperfusion after HI, preceding neuronal death.
614. Styrene 7,8-oxide induces caspase activation and regular DNA fragmentation in neuronal cells
- Author
-
Roshan Tofighi, Elisabetta Daré, Diana Poli, Sandra Ceccatelli, Takashi Momoi, Maria Vittoria Vettori, Antonio Mutti, and Takaomi C. Saido
- Subjects
Cell Membrane Permeability ,DNA damage ,Poly ADP ribose polymerase ,Immunoblotting ,DNA Fragmentation ,chemistry.chemical_compound ,Styrene oxide ,Humans ,Coloring Agents ,Molecular Biology ,Cells, Cultured ,Caspase ,Neurons ,Staining and Labeling ,biology ,General Neuroscience ,Microfilament Proteins ,Osmolar Concentration ,Calpain ,Trypan Blue ,Molecular biology ,Enzyme Activation ,chemistry ,Cell culture ,Apoptosis ,Caspases ,Culture Media, Conditioned ,biology.protein ,Epoxy Compounds ,DNA fragmentation ,Neurology (clinical) ,Tumor Suppressor Protein p53 ,Carrier Proteins ,Developmental Biology - Abstract
Neurobehavioral changes have been described in workers occupationally exposed to styrene vapors. Alterations of neurotransmitters and loss of neurons have been observed in brains of styrene-exposed rats. However, the mechanisms of neuronal damage are not yet clearly understood. We have characterized the cellular alterations induced by the main reactive intermediate of styrene metabolism, styrene 7,8-oxide (SO) in the human neuroblastoma SK-N-MC cell line and primary culture of rat cerebellar granule cells (CGC). SK-N-MC cells exposed to SO (0.3-1 mM) displayed apoptotic morphology, together with chromatin condensation and DNA cleavage into high molecular weight fragments of regular size. These features were accompanied by the activation of class II caspases, as detected with the DEVD assay, by following the cleavage of the caspase-substrate poly (ADP-ribose) polymerase (PARP) and by detection of the active fragment of caspase-3. Pre-incubation of the cells with the caspase inhibitor z-VAD-fmk reduced the cellular damage induced by SO, suggesting that caspases play an important role in SO toxicity. Increased proteolysis by class II caspases was detected also in primary culture of CGC exposed to SO. In addition, the presence of the 150-kDa cleavage product of alpha-fodrin suggests a possible activation of calpains in SK-N-MC cells. Moreover, SO did not affect the level of expression of the p53 protein, even though it is known to cause DNA damage. The identified intracellular pathways affected by SO exposure provides end-points that can be used in future studies for the evaluation of the neurotoxic effect of styrene in vivo.
615. Activation of calpain I converts excitotoxic neuron death into a caspase-independent cell death
- Author
-
Monika Poppe, Takaomi C. Saido, Aaron J. Krohn, C. Marc Luetjens, Greg M. Cole, Jochen H. M. Prehn, Silke Lankiewicz, and Nguyen Truc Bui
- Subjects
Programmed cell death ,N-Methylaspartate ,Apoptosis ,Cytochrome c Group ,Hippocampus ,Biochemistry ,medicine ,Animals ,Staurosporine ,Molecular Biology ,Cells, Cultured ,Caspase ,Neurons ,biology ,Calpain ,Cytochrome c ,Glutamate receptor ,Cell Biology ,Molecular biology ,Mitochondria ,Rats ,Cell biology ,Enzyme Activation ,Microscopy, Fluorescence ,Caspases ,biology.protein ,Neuron death ,medicine.drug - Abstract
Glutamate receptor overactivation contributes to neuron death after stroke, trauma, and epileptic seizures. Exposure of cultured rat hippocampal neurons to the selective glutamate receptor agonist N-methyl-d-aspartate (300 microm, 5 min) or to the apoptosis-inducing protein kinase inhibitor staurosporine (300 nm) induced a delayed neuron death. In both cases, neuron death was preceded by the mitochondrial release of the pro-apoptotic factor cytochrome c. Unlike staurosporine, the N-methyl-d-aspartate-induced release of cytochrome c did not lead to significant activation of caspase-3, the main caspase involved in the execution of neuronal apoptosis. In contrast, activation of the Ca(2+)-activated neutral protease calpain I was readily detectable after the exposure to N-methyl-d-aspartate. In a neuronal cell-free apoptosis system, calpain I prevented the ability of cytochrome c to activate the caspase cascade by inhibiting the processing of procaspase-3 and -9 into their active subunits. In the hippocampal neuron cultures, the inhibition of calpain activity restored caspase-3-like protease activity after an exposure to N-methyl-d-aspartate. Our data demonstrate the existence of signal transduction pathways that prevent the entry of cells into a caspase-dependent cell death program after the mitochondrial release of cytochrome c.
616. Comprehensive behavioral phenotyping of calpastatin-knockout mice
- Author
-
Nobuhisa Iwata, Takaomi C. Saido, Tsuyoshi Miyakawa, Keizo Takao, Ryuichi Nakajima, Shu-Ming Huang, and Jiro Takano
- Subjects
Reflex, Startle ,Pain ,Endogeny ,Anxiety ,Motor Activity ,lcsh:RC346-429 ,Rotarod performance test ,Mice ,Cellular and Molecular Neuroscience ,Calcium-binding protein ,Conditioning, Psychological ,Avoidance Learning ,Animals ,Maze Learning ,Social Behavior ,Molecular Biology ,lcsh:Neurology. Diseases of the nervous system ,Swimming ,Calpastatin ,Mice, Knockout ,Behavior, Animal ,biology ,Research ,Calcium-Binding Proteins ,Calpain ,Fear ,Sensory Gating ,Startle reaction ,Phenotype ,Rotarod Performance Test ,Knockout mouse ,Exploratory Behavior ,biology.protein ,Psychology ,Neuroscience ,Intracellular - Abstract
Background Calpastatin is an endogenous inhibitor of calpain, intracellular calcium-activated protease. It has been suggested to be involved in molecular mechanisms of long-term plasticity and excitotoxic pathways. However, functions of calpastatin in vivo are still largely unknown. To examine the physiological roles of calpastatin, we subjected calpastatin-knockout mice to a comprehensive behavioral test battery. Results Calpastatin-knockout mice showed decreased locomotor activity under stressful environments, and decreased acoustic startle response, but we observed no significant change in hippocampus-dependent memory function. Conclusion These results suggest that calpastatin is likely to be more closely associated with affective rather than cognitive aspects of brain function.
617. Muscle-specific calpain, p94, is degraded by autolysis immediately after translation, resulting in disappearance from muscle
- Author
-
Shoichi Ishiura, Koichi Suzuki, Hiroyuki Sorimachi, Hiroshi Kawasaki, Hideo Sugita, K.-I. Arahata, Masayuki Miyasaka, Noriko Toyama-Sorimachi, and Takaomi C. Saido
- Subjects
Macromolecular Substances ,Protein subunit ,Molecular Sequence Data ,Biology ,Kidney ,Transfection ,Biochemistry ,Cell Line ,Complementary DNA ,Gene expression ,Chlorocebus aethiops ,medicine ,Serine ,Myocyte ,Animals ,Amino Acid Sequence ,Cysteine ,RNA, Messenger ,Molecular Biology ,Sequence Deletion ,Messenger RNA ,COS cells ,Base Sequence ,Calpain ,Muscles ,Skeletal muscle ,Cell Biology ,Molecular biology ,Immunohistochemistry ,Recombinant Proteins ,Rats ,Molecular Weight ,medicine.anatomical_structure ,Oligodeoxyribonucleotides ,Protein Biosynthesis ,biology.protein ,Mutagenesis, Site-Directed - Abstract
We previously identified a third type of the calpain large subunit named p94 as a cDNA whose mRNA is expressed exclusively in skeletal muscle at levels approximately 10-fold more abundant than those of the conventional calpain subunit. Rat skeletal muscle fractions were screened by two anti-peptide antibodies raised against two specific sequences in p94, but the p94 protein could not be found. To examine this apparent discrepancy between the amounts of mRNA and protein, wild-type p94 was expressed in COS cells. Although p94 mRNA was expressed normally in COS cells, only very small amounts of the protein and its presumed degradation products were detected by the antibodies described above. A series of COOH-terminal deletion mutants was constructed and expressed in COS cells and L8 cells, a rat myoblast cell line. When IS2, one of the specific regions of p94, was completely eliminated, the truncated p94 proteins were expressed normally, and the amount of the expressed proteins was at least 100-fold higher than with wild-type p94. Moreover, when site-directed mutagenesis was introduced to change the presumed active-site cysteine of p94 to serine or alanine, the mutated p94 proteins were highly expressed like the IS2-deleted mutants. These results indicate the following. 1) The mRNA for p94 is normally transcribed in COS, L8, and muscle cells; 2) the p94 protein becomes active in the cytosol immediately after translation; 3) the p94 protein virtually disappears from cells by autocatalytic degradation; and 4) the p94-specific IS2 region plays an important role in this degradation. In vitro translation experiments support this idea. Furthermore, p94 shows nuclear localization when expressed in COS cells. The physiological function of p94 in muscle is discussed on the basis of the analysis of these transfectants.
618. Synapse Loss and Microglial Activation Precede Tangles in a P301S Tauopathy Mouse Model
- Author
-
Takaomi C. Saido, Tetsuya Suhara, Shu-Ming Huang, John Q. Trojanowski, Bin Zhang, Jun Maeda, Makoto Higuchi, Yasumasa Yoshiyama, Virginia M.-Y. Lee, and Nobuhisa Iwata
- Subjects
Aging ,Neuroscience(all) ,Transgene ,Blotting, Western ,HUMDISEASE ,tau Proteins ,Hippocampal formation ,Biology ,Amyloid Neuropathies ,Microgliosis ,Microtubules ,MOLNEURO ,Tacrolimus ,Tangle ,Synapse ,Mice ,Atrophy ,medicine ,Animals ,Humans ,Gliosis ,Phosphorylation ,Neuroinflammation ,General Neuroscience ,Brain ,Neurofibrillary Tangles ,Macrophage Activation ,Amygdala ,medicine.disease ,Immunohistochemistry ,Microscopy, Electron ,Solubility ,Spinal Cord ,Mossy Fibers, Hippocampal ,Nerve Degeneration ,Synapses ,Microglia ,Tauopathy ,Neuroscience ,Immunosuppressive Agents - Abstract
SummaryFilamentous tau inclusions are hallmarks of Alzheimer's disease (AD) and related tauopathies, but earlier pathologies may herald disease onset. To investigate this, we studied wild-type and P301S mutant human tau transgenic (Tg) mice. Filamentous tau lesions developed in P301S Tg mice at 6 months of age, and progressively accumulated in association with striking neuron loss as well as hippocampal and entorhinal cortical atrophy by 9-12 months of age. Remarkably, hippocampal synapse loss and impaired synaptic function were detected in 3 month old P301S Tg mice before fibrillary tau tangles emerged. Prominent microglial activation also preceded tangle formation. Importantly, immunosuppression of young P301S Tg mice with FK506 attenuated tau pathology and increased lifespan, thereby linking neuroinflammation to early progression of tauopathies. Thus, hippocampal synaptic pathology and microgliosis may be the earliest manifestations of neurodegenerative tauopathies, and abrogation of tau-induced microglial activation could retard progression of these disorders.
- Full Text
- View/download PDF
619. Cystatin F (Cst7) drives sex-dependent changes in microglia in an amyloid-driven model of Alzheimer’s disease
- Author
-
Michael JD Daniels, Lucas Lefevre, Stefan Szymkowiak, Alice Drake, Laura McCulloch, Makis Tzioras, Jack Barrington, Owen R Dando, Xin He, Mehreen Mohammad, Hiroki Sasaguri, Takashi Saito, Takaomi C Saido, Tara L Spires-Jones, and Barry W McColl
- Subjects
microglia ,Alzheimer's disease ,lysosome ,sexual dimorphism ,phagocytosis ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Microglial endolysosomal (dys)function is strongly implicated in neurodegenerative disease. Transcriptomic studies show that a microglial state characterised by a set of genes involved in endolysosomal function is induced in both mouse Alzheimer’s disease (AD) models and human AD brain, and that the emergence of this state is emphasised in females. Cst7 (encoding cystatin F) is among the most highly upregulated genes in these microglia. However, despite such striking and robust upregulation, the function of Cst7 in neurodegenerative disease is not understood. Here, we crossed Cst7-/- mice with the AppNL-G-F mouse to test the role of Cst7 in a model of amyloid-driven AD. Surprisingly, we found that Cst7 plays a sexually dimorphic role regulating microglia in this model. In females, Cst7-/-AppNL-G-F microglia had greater endolysosomal gene expression, lysosomal burden, and amyloid beta (Aβ) burden in vivo and were more phagocytic in vitro. However, in males, Cst7-/-AppNL-G-F microglia were less inflammatory and had a reduction in lysosomal burden but had no change in Aβ burden. Overall, our study reveals functional roles for one of the most commonly upregulated genes in microglia across disease models, and the sex-specific profiles of Cst7-/--altered microglial disease phenotypes. More broadly, the findings raise important implications for AD including crucial questions on sexual dimorphism in neurodegenerative disease and the interplay between endolysosomal and inflammatory pathways in AD pathology.
- Published
- 2023
- Full Text
- View/download PDF
620. Brain p3‐Alcβ peptide restores neuronal viability impaired by Alzheimer's amyloid β‐peptide
- Author
-
Saori Hata, Haruka Saito, Takeharu Kakiuchi, Dai Fukumoto, Shigeyuki Yamamoto, Kensaku Kasuga, Ayano Kimura, Koichi Moteki, Ruriko Abe, Shungo Adachi, Shoich Kinoshita, Kumiko Yoshizawa‐Kumagaye, Hideki Nishio, Takashi Saito, Takaomi C Saido, Tohru Yamamoto, Masaki Nishimura, Hidenori Taru, Yuriko Sobu, Hiroyuki Ohba, Shingo Nishiyama, Norihiro Harada, Takeshi Ikeuchi, Hideo Tsukada, Yasuomi Ouchi, and Toshiharu Suzuki
- Subjects
AD therapy ,alcadein ,Alzheimer's disease (AD) ,mitochondria ,PET imaging ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract We propose a new therapeutic strategy for Alzheimer's disease (AD). Brain peptide p3‐Alcβ37 is generated from the neuronal protein alcadein β through cleavage of γ‐secretase, similar to the generation of amyloid β (Aβ) derived from Aβ‐protein precursor/APP. Neurotoxicity by Aβ oligomers (Aβo) is the prime cause prior to the loss of brain function in AD. We found that p3‐Alcβ37 and its shorter peptide p3‐Alcβ9‐19 enhanced the mitochondrial activity of neurons and protected neurons against Aβo‐induced toxicity. This is due to the suppression of the Aβo‐mediated excessive Ca2+ influx into neurons by p3‐Alcβ. Successful transfer of p3‐Alcβ9‐19 into the brain following peripheral administration improved the mitochondrial viability in the brain of AD mice model, in which the mitochondrial activity is attenuated by increasing the neurotoxic human Aβ42 burden, as revealed through brain PET imaging to monitor mitochondrial function. Because mitochondrial dysfunction is common in the brain of AD patients alongside increased Aβ and reduced p3‐Alcβ37 levels, the administration of p3‐Alcβ9‐19 may be a promising treatment for restoring, protecting, and promoting brain functions in patients with AD.
- Published
- 2023
- Full Text
- View/download PDF
621. AAV‐mediated delivery of an anti‐BACE1 VHH alleviates pathology in an Alzheimer's disease model
- Author
-
Marika Marino, Lujia Zhou, Melvin Y Rincon, Zsuzsanna Callaerts‐Vegh, Jens Verhaert, Jérôme Wahis, Eline Creemers, Lidia Yshii, Keimpe Wierda, Takashi Saito, Catherine Marneffe, Iryna Voytyuk, Yessica Wouters, Maarten Dewilde, Sandra I Duqué, Cécile Vincke, Yona Levites, Todd E Golde, Takaomi C Saido, Serge Muyldermans, Adrian Liston, Bart De Strooper, and Matthew G Holt
- Subjects
AAV ,Alzheimer’s disease ,anti‐BACE1 ,VHH ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract Single domain antibodies (VHHs) are potentially disruptive therapeutics, with important biological value for treatment of several diseases, including neurological disorders. However, VHHs have not been widely used in the central nervous system (CNS), largely because of their restricted blood–brain barrier (BBB) penetration. Here, we propose a gene transfer strategy based on BBB‐crossing adeno‐associated virus (AAV)‐based vectors to deliver VHH directly into the CNS. As a proof‐of‐concept, we explored the potential of AAV‐delivered VHH to inhibit BACE1, a well‐characterized target in Alzheimer’s disease. First, we generated a panel of VHHs targeting BACE1, one of which, VHH‐B9, shows high selectivity for BACE1 and efficacy in lowering BACE1 activity in vitro. We further demonstrate that a single systemic dose of AAV‐VHH‐B9 produces positive long‐term (12 months plus) effects on amyloid load, neuroinflammation, synaptic function, and cognitive performance, in the AppNL‐G‐F Alzheimer’s mouse model. These results constitute a novel therapeutic approach for neurodegenerative diseases, which is applicable to a range of CNS disease targets.
- Published
- 2022
- Full Text
- View/download PDF
622. Microglia and CD206+ border-associated mouse macrophages maintain their embryonic origin during Alzheimer’s disease
- Author
-
Xiaoting Wu, Takashi Saito, Takaomi C Saido, Anna M Barron, and Christiane Ruedl
- Subjects
brain ,macrophages ,microglia ,fate-mapping ,border-associated macrophages ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Brain microglia and border-associated macrophages (BAMs) display distinct spatial, developmental, and phenotypic features. Although at steady state, the origins of distinct brain macrophages are well-documented, the dynamics of their replenishment in neurodegenerative disorders remain elusive, particularly for activated CD11c+ microglia and BAMs. In this study, we conducted a comprehensive fate-mapping analysis of murine microglia and BAMs and their turnover kinetics during Alzheimer’s disease (AD) progression. We used a novel inducible AD mouse model to investigate the contribution of bone marrow (BM) cells to the pool of fetal-derived brain macrophages during the development of AD. We demonstrated that microglia remain a remarkably stable embryonic-derived population even during the progression of AD pathology, indicating that neither parenchymal macrophage subpopulation originates from, nor is replenished by, BM-derived cells. At the border-associated brain regions, bona fide CD206+ BAMs are minimally replaced by BM-derived cells, and their turnover rates are not accelerated by AD. In contrast, all other myeloid cells are swiftly replenished by BM progenitors. This information further elucidates the turnover kinetics of these cells not only at steady state, but also in neurodegenerative diseases, which is crucial for identifying potential novel therapeutic targets.
- Published
- 2021
- Full Text
- View/download PDF
623. NHE6 depletion corrects ApoE4-mediated synaptic impairments and reduces amyloid plaque load
- Author
-
Theresa Pohlkamp, Xunde Xian, Connie H Wong, Murat S Durakoglugil, Gordon Chandler Werthmann, Takaomi C Saido, Bret M Evers, Charles L White III, Jade Connor, Robert E Hammer, and Joachim Herz
- Subjects
ApoE ,Alzheimer's ,NHE6 ,neurodegeneration ,endosome ,trafficking ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Apolipoprotein E4 (ApoE4) is the most important and prevalent risk factor for late-onset Alzheimer’s disease (AD). The isoelectric point of ApoE4 matches the pH of the early endosome (EE), causing its delayed dissociation from ApoE receptors and hence impaired endolysosomal trafficking, disruption of synaptic homeostasis, and reduced amyloid clearance. We have shown that enhancing endosomal acidification by inhibiting the EE-specific sodium-hydrogen exchanger 6 (NHE6) restores vesicular trafficking and normalizes synaptic homeostasis. Remarkably and unexpectedly, loss of NHE6 (encoded by the gene Slc9a6) in mice effectively suppressed amyloid deposition even in the absence of ApoE4, suggesting that accelerated acidification of EEs caused by the absence of NHE6 occludes the effect of ApoE on amyloid plaque formation. NHE6 suppression or inhibition may thus be a universal, ApoE-independent approach to prevent amyloid buildup in the brain. These findings suggest a novel therapeutic approach for the prevention of AD by which partial NHE6 inhibition reverses the ApoE4-induced endolysosomal trafficking defect and reduces plaque load.
- Published
- 2021
- Full Text
- View/download PDF
624. Fibrillar Aβ triggers microglial proteome alterations and dysfunction in Alzheimer mouse models
- Author
-
Laura Sebastian Monasor, Stephan A Müller, Alessio Vittorio Colombo, Gaye Tanrioever, Jasmin König, Stefan Roth, Arthur Liesz, Anna Berghofer, Anke Piechotta, Matthias Prestel, Takashi Saito, Takaomi C Saido, Jochen Herms, Michael Willem, Christian Haass, Stefan F Lichtenthaler, and Sabina Tahirovic
- Subjects
Alzheimer's disease ,microglia ,proteomic signatures ,neuroinflammation ,phagocytosis ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Microglial dysfunction is a key pathological feature of Alzheimer's disease (AD), but little is known about proteome-wide changes in microglia during the course of AD and their functional consequences. Here, we performed an in-depth and time-resolved proteomic characterization of microglia in two mouse models of amyloid β (Aβ) pathology, the overexpression APPPS1 and the knock-in APP-NL-G-F (APP-KI) model. We identified a large panel of Microglial Aβ Response Proteins (MARPs) that reflect heterogeneity of microglial alterations during early, middle and advanced stages of Aβ deposition and occur earlier in the APPPS1 mice. Strikingly, the kinetic differences in proteomic profiles correlated with the presence of fibrillar Aβ, rather than dystrophic neurites, suggesting that fibrillar Aβ may trigger the AD-associated microglial phenotype and the observed functional decline. The identified microglial proteomic fingerprints of AD provide a valuable resource for functional studies of novel molecular targets and potential biomarkers for monitoring AD progression or therapeutic efficacy.
- Published
- 2020
- Full Text
- View/download PDF
625. Amyloid β induces interneuron-specific changes in the hippocampus of APPNL-F mice.
- Author
-
Katalin E Sos, Márton I Mayer, Virág T Takács, Abel Major, Zsuzsanna Bardóczi, Barnabas M Beres, Tamás Szeles, Takashi Saito, Takaomi C Saido, István Mody, Tamás F Freund, and Gábor Nyiri
- Subjects
Medicine ,Science - Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by cognitive decline and amyloid-beta (Aβ) depositions generated by the proteolysis of amyloid precursor protein (APP) in the brain. In APPNL-F mice, APP gene was humanized and contains two familial AD mutations, and APP-unlike other mouse models of AD-is driven by the endogenous mouse APP promoter. Similar to people without apparent cognitive dysfunction but with heavy Aβ plaque load, we found no significant decline in the working memory of adult APPNL-F mice, but these mice showed decline in the expression of normal anxiety. Using immunohistochemistry and 3D block-face scanning electron microscopy, we found no changes in GABAA receptor positivity and size of somatic and dendritic synapses of hippocampal interneurons. We did not find alterations in the level of expression of perineuronal nets around parvalbumin (PV) interneurons or in the density of PV- or somatostatin-positive hippocampal interneurons. However, in contrast to other investigated cell types, PV interneuron axons were occasionally mildly dystrophic around Aβ plaques, and the synapses of PV-positive axon initial segment (AIS)-targeting interneurons were significantly enlarged. Our results suggest that PV interneurons are highly resistant to amyloidosis in APPNL-F mice and amyloid-induced increase in hippocampal pyramidal cell excitability may be compensated by PV-positive AIS-targeting cells. Mechanisms that make PV neurons more resilient could therefore be exploited in the treatment of AD for mitigating Aβ-related inflammatory effects on neurons.
- Published
- 2020
- Full Text
- View/download PDF
626. Correction: T-type calcium channel enhancer SAK3 promotes dopamine and serotonin releases in the hippocampus in naive and amyloid precursor protein knock-in mice.
- Author
-
Shuo Wang, Yasushi Yabuki, Kazuya Matsuo, Jing Xu, Hisanao Izumi, Kenji Sakimura, Takashi Saito, Takaomi C Saido, and Kohji Fukunaga
- Subjects
Medicine ,Science - Abstract
[This corrects the article DOI: 10.1371/journal.pone.0206986.].
- Published
- 2019
- Full Text
- View/download PDF
627. Humanization of the entire murine Mapt gene provides a murine model of pathological human tau propagation.
- Author
-
Takashi Saito, Naomi Mihira, Yukio Matsuba, Hiroki Sasaguri, Shoko Hashimoto, Sneha Narasimhan, Bin Zhang, Shigeo Murayama, Makoto Higuchi, Lee, Virginia M. Y., Trojanowski, John Q., and Takaomi C. Saido
- Subjects
- *
NEUROFIBRILLARY tangles , *AMYLOID beta-protein precursor , *TAU proteins , *MICROTUBULE-associated proteins , *HUMAN behavior , *ALZHEIMER'S disease - Abstract
In cortical regions of brains from individuals with preclinical or clinical Alzheimer's disease (AD), extracellular β-amyloid (Aβ) deposition precedes the aggregation of pathological intracellular tau (the product of the gene microtubule-associated protein tau (MAPT)). To our knowledge, current mouse models of tauopathy reconstitute tau pathology by overexpressing mutant human tau protein. Here, through a homologous recombination approach that replaced the entire murine Mapt gene with the human ortholog, we developed knock-in mice with humanized Mapt to create an in vivoplatform for studying human tauopathy. Of note, the humanized Mapt expressed all six tau isoforms present in humans. We next cross-bred the MAPT knock-in mice with single amyloid precursor protein (App) knock-in mice to investigate the Aβ-tau axis in AD etiology. The double-knock-in mice exhibited higher tau phosphorylation than did single MAPT knock-in mice but initially lacked apparent tauopathy and neurodegeneration, as observed in the single App knock-in mice. We further observed that tau humanization significantly accelerates cell-to-cell propagation of AD brain-derived pathological tau both in the absence and presence of Aβ-amyloidosis. In the presence of Aβ-amyloidosis, tau accumulation was intensified and closely associated with dystrophic neurites, consistently showing that Aβ-amyloidosis affects tau pathology. Our results also indicated that the pathological human tau interacts better with human tau than with murine tau, suggesting species-specific differences between these orthologous pathogenic proteins. We propose that the MAPT knock-in mice will make it feasible to investigate the behaviors and characteristics of human tau in an animal model. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
628. T-type calcium channel enhancer SAK3 promotes dopamine and serotonin releases in the hippocampus in naive and amyloid precursor protein knock-in mice.
- Author
-
Shuo Wang, Yasushi Yabuki, Kazuya Matsuo, Jing Xu, Hisanao Izumi, Kenji Sakimura, Takashi Saito, Takaomi C Saido, and Kohji Fukunaga
- Subjects
Medicine ,Science - Abstract
T-type calcium channels in the brain mediate the pathophysiology of epilepsy, pain, and sleep. Recently, we developed a novel therapeutic candidate, SAK3 (ethyl 8'-methyl-2',4-dioxo-2-(piperidin-1-yl)-2'H-spiro[cyclopentane-1,3'-imidazo[1,2-a] pyridine]-2-ene-3-carboxylate), for Alzheimer's disease (AD). The cognitive improvement by SAK3 is closely associated with enhanced acetylcholine (ACh) release in the hippocampus. Since monoamines such as dopamine (DA), noradrenaline (NA), and serotonin (5-HT) are also involved in hippocampus-dependent learning and psychomotor behaviors in mice, we investigated the effects of SAK3 on these monoamine releases in the mouse brain. Oral administration of SAK3 (0.5 mg/kg, p.o.) significantly promoted DA and 5-HT releases in the naive mouse hippocampal CA1 region but not in the medial prefrontal cortex (mPFC), while SAK3 did not affect NA release in either brain region. The T-type calcium channel-specific inhibitor, NNC 55-0396 (1 μM) significantly antagonized SAK3-enhanced DA and 5-HT releases in the hippocampus. Interestingly, the α7 nicotinic ACh receptor (nAChR) antagonist, methyllycaconitine (1 nM) significantly inhibited DA release, and the α4 nAChR antagonist, dihydro-β-erythroidine (100 μM) significantly blocked both DA and 5-HT releases following SAK3 (0.5 mg/kg, p.o.) administration in the hippocampus. SAK3 did not alter basal monoamine contents both in the mPFC and hippocampus. SAK3 (0.5 mg/kg, p.o.) administration also significantly elevated DA and 5-HT releases in the hippocampal CA1 region of amyloid-precursor protein (APP)NL-GF knock-in (KI) mice. Moreover, hippocampal DA and 5-HT contents were significantly decreased in APPNL-GF KI mice. Taken together, our data suggest that SAK3 promotes monoamine DA and 5-HT releases by enhancing the T-type calcium channel and nAChR in the mouse hippocampus.
- Published
- 2018
- Full Text
- View/download PDF
629. An aberrant sugar modification of BACE1 blocks its lysosomal targeting in Alzheimer's disease
- Author
-
Yasuhiko Kizuka, Shinobu Kitazume, Reiko Fujinawa, Takashi Saito, Nobuhisa Iwata, Takaomi C Saido, Miyako Nakano, Yoshiki Yamaguchi, Yasuhiro Hashimoto, Matthias Staufenbiel, Hiroyuki Hatsuta, Shigeo Murayama, Hiroshi Manya, Tamao Endo, and Naoyuki Taniguchi
- Subjects
Alzheimer's disease (AD) ,amyloid‐β (Aβ) ,BACE1 ,bisecting GlcNAc ,GnT‐III (Mgat3) ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract The β‐site amyloid precursor protein cleaving enzyme‐1 (BACE1), an essential protease for the generation of amyloid‐β (Aβ) peptide, is a major drug target for Alzheimer's disease (AD). However, there is a concern that inhibiting BACE1 could also affect several physiological functions. Here, we show that BACE1 is modified with bisecting N‐acetylglucosamine (GlcNAc), a sugar modification highly expressed in brain, and demonstrate that AD patients have higher levels of bisecting GlcNAc on BACE1. Analysis of knockout mice lacking the biosynthetic enzyme for bisecting GlcNAc, GnT‐III (Mgat3), revealed that cleavage of Aβ‐precursor protein (APP) by BACE1 is reduced in these mice, resulting in a decrease in Aβ plaques and improved cognitive function. The lack of this modification directs BACE1 to late endosomes/lysosomes where it is less colocalized with APP, leading to accelerated lysosomal degradation. Notably, other BACE1 substrates, CHL1 and contactin‐2, are normally cleaved in GnT‐III‐deficient mice, suggesting that the effect of bisecting GlcNAc on BACE1 is selective to APP. Considering that GnT‐III‐deficient mice remain healthy, GnT‐III may be a novel and promising drug target for AD therapeutics.
- Published
- 2015
- Full Text
- View/download PDF
630. Endoplasmic reticulum stress responses in mouse models of Alzheimer’s disease: Overexpression paradigm versus knockin paradigm.
- Author
-
Shoko Hashimoto, Ayano Ishii, Naoko Kamano, Naoto Watamura, Takashi Saito, Toshio Ohshima, Makoto Yokosuka, and Takaomi C. Saido
- Subjects
- *
GENETICS of Alzheimer's disease , *ANIMAL models of Alzheimer's disease , *GENETIC overexpression , *LABORATORY mice , *ETIOLOGY of Alzheimer's disease - Abstract
Endoplasmic reticulum (ER) stress is believed to play an important role in the etiology of Alzheimer's disease (AD). The accumulation of misfolded proteins and perturbation of intracellular calcium homeostasis are thought to underlie the induction of ER stress, resulting in neuronal dysfunction and cell death. Several reports have shown an increased ER stress response in amyloid precursor protein (APP) and presenilin1 (PS1) double-transgenic (Tg) AD mouse models. However, whether the ER stress observed in these mouse models is actually caused by AD pathology remains unclear. APP and PS1 contain one and nine transmembrane domains, respectively, for which it has been postulated that overexpressed membrane proteins can become wedged in a misfolded configuration in ER membranes, thereby inducing nonspecific ER stress. Here, we used an App-knockin (KI) AD mouse model that accumulates amyloid-β (Aβ) peptide without overexpressing APP to investigate whether the ER stress response is heightened because of Aβ pathology. Thorough examinations indicated that no ER stress responses arose in App-KI or single APP-Tg mice. These results suggest that PS1 overexpression or mutation induced a nonspecific ER stress response that was independent of Aβ pathology in the double-Tg mice. Moreover, we observed no ER stress in a mouse model of tauopathy (P301S-Tau-Tg mice) at various ages, suggesting that ER stress is also not essential in tau pathology–induced neurodegeneration. We conclude that the role of ER stress in AD pathogenesis needs to be carefully addressed in future studies. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
631. Time-course global proteome analyses reveal an inverse correlation between Aβ burden and immunoglobulin M levels in the APPNL-F mouse model of Alzheimer disease.
- Author
-
Hansen Wang, Declan Williams, Jennifer Griffin, Takashi Saito, Takaomi C Saido, Paul E Fraser, Ekaterina Rogaeva, and Gerold Schmitt-Ulms
- Subjects
Medicine ,Science - Abstract
Alzheimer disease (AD) stands out amongst highly prevalent diseases because there is no effective treatment nor can the disease be reliably diagnosed at an early stage. A hallmark of AD is the accumulation of aggregation-prone amyloid β peptides (Aβ), the main constituent of amyloid plaques. To identify Aβ-dependent changes to the global proteome we used the recently introduced APPNL-F mouse model of AD, which faithfully recapitulates the Aβ pathology of the disease, and a workflow that interrogated the brain proteome of these mice by quantitative mass spectrometry at three different ages. The elevated Aβ burden in these mice was observed to cause almost no changes to steady-state protein levels of the most abundant >2,500 brain proteins, including 12 proteins encoded by well-confirmed AD risk loci. The notable exception was a striking reduction in immunoglobulin heavy mu chain (IGHM) protein levels in homozygote APPNL-F/NL-F mice, relative to APPNL-F/wt littermates. Follow-up experiments revealed that IGHM levels generally increase with age in this model. Although discovered with brain samples, the relative IGHM depletion in APPNL-F/NL-F mice was validated to manifest systemically in the blood, and did not extend to other blood proteins, including immunoglobulin G. Results presented are consistent with a cause-effect relationship between the excessive accumulation of Aβ and the selective depletion of IGHM levels, which may be of relevance for understanding the etiology of the disease and ongoing efforts to devise blood-based AD diagnostics.
- Published
- 2017
- Full Text
- View/download PDF
632. Neuron-specific methylome analysis reveals epigenetic regulation and tau-related dysfunction of BRCA1 in Alzheimer's disease.
- Author
-
Tatsuo Mano, Kenichi Nagata, Takashi Nonaka, Airi Tarutani, Tomohiro Imamura, Tadafumi Hashimoto, Taro Bannai, Kagari Koshi-Mano, Takeyuki Tsuchida, Ryo Ohtomo, Junko Takahashi-Fujigasaki, Satoshi Yamashita, Yasumasa Ohyagi, Ryo Yamasaki, Shoji Tsuji, Akira Tamaoka, Takeshi Ikeuchi, Takaomi C. Saido, Takeshi Iwatsubo, and Toshikazu Ushijima
- Subjects
- *
ALZHEIMER'S disease , *TAU proteins , *AMYLOID beta-protein , *EPIGENETICS , *AMYLOID beta-protein precursor , *NEURONS - Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disease characterized by pathology of accumulated amyloid β (Aβ) and phosphorylated tau proteins in the brain. Postmortem degradation and cellular complexity within the brain have limited approaches to molecularly define the causal relationship between pathological features and neuronal dysfunction in AD. To overcome these limitations, we analyzed the neuron-specific DNA methylome of postmortem brain samples from AD patients, which allowed differentially hypomethylated region of the BRCA1 promoter to be identified. Expression of BRCA1 was significantly up-regulated in AD brains, consistent with its hypomethylation. BRCA1 protein levels were also elevated in response to DNA damage induced by Aβ. BRCA1 became mislocalized to the cytoplasm and highly insoluble in a tau-dependent manner, resulting in DNA fragmentation in both in vitro cellular and in vivo mouse models. BRCA1 dysfunction under Aβ burden is consistent with concomitant deterioration of genomic integrity and synaptic plasticity. The Brca1 promoter region of AD model mice brain was similarly hypomethylated, indicating an epigenetic mechanism underlying BRCA1 regulation in AD. Our results suggest deterioration of DNA integrity as a central contributing factor in AD pathogenesis. Moreover, these data demonstrate the technical feasibility of using neuron-specific DNA methylome analysis to facilitate discovery of etiological candidates in sporadic neurodegenerative diseases. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
633. Involvement of calpains in adult neurogenesis: implications for stroke
- Author
-
Vanessa Mendes Machado, Maria Inês Morte, Bruno Pereira Carreira, Maria Manuela Azevedo, Jiro eTakano, Nobuhisa eIwata, Takaomi C Saido, Hannelore eAsmussen, Alan Rick Horwitz, Caetana Monteiro Carvalho, and Inês Maria Araújo
- Subjects
Hippocampus ,Neurogenesis ,Stroke ,Migration ,proliferation ,Subventricular zone ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Calpains are ubiquitous proteases involved in cell proliferation, adhesion and motility. In the brain, calpains have been associated with neuronal damage in both acute and neurodegenerative disorders, but their physiological function in the nervous system remains elusive. During brain ischemia, there is a large increase in the levels of intracellular calcium, leading to the activation of calpains. Inhibition of these proteases has been shown to reduce neuronal death in a variety of stroke models. On the other hand, after stroke, neural stem cells increase their proliferation and newly formed neuroblasts migrate towards the site of injury. However, the process of forming new neurons after injury is not efficient and finding ways to improve it may help with recovery after lesion. Understanding the role of calpains in the process of neurogenesis may therefore open a new window for the treatment of stroke. We investigated the involvement of calpains in neural stem cell proliferation and neuroblast migration in two highly neurogenic regions in the mouse brain, the dentate gyrus and the subventricular zone. We used mice that lack calpastatin, the endogenous calpain inhibitor, and calpains were also modulated directly, using calpeptin, a pharmacological calpain inhibitor. Calpastatin deletion impaired both neural stem cell proliferation and neuroblast migration. Calpain inhibition increased neural stem cell proliferation, migration speed and migration distance in cells from the subventricular zone. Overall, our work suggests that calpains are important for neurogenesis and warrant further research on how they influence the formation of new neurons. Prospective therapies targeting calpain activity not only may afford neuroprotection following stroke, but also benefit the formation and survival of new neurons.
- Published
- 2015
- Full Text
- View/download PDF
634. Dutch, Flemish, Italian, and Arctic mutations of APP and resistance of Aβ to physiologically relevant proteolytic degradation.
- Author
-
Satoshi Tsubuki, Yoshie Takaki, and Takaomi C Saido
- Subjects
- *
AMYLOIDOSIS , *PROTEIN metabolism disorders , *PEPTIDES , *LYMPHOPROLIFERATIVE disorders - Abstract
The Dutch, Flemish, Italian, and Arctic mutations in the amyloid precursor protein (APP) gene encode changes within the sequence of the amyloid β peptide (Aβ) and cause presenile cerebral amyloid angiopathy, cerebral parenchymal amyloidosis, or both. These disorders are caused by accumulation of Aβ, with no evidence of increased Aβ production. Our results showed that these mutations in Aβ make it resistant to proteolytic degradation by neprilysin, the peptidase with the most important role in catabolism of Aβ in the brain. These mutations in Aβ could thus be pathogenic not only by facilitating fibrillogenesis but also by extending the half-life of Aβ in the brain. [ABSTRACT FROM AUTHOR]
- Published
- 2003
- Full Text
- View/download PDF
635. Calpain-dependent Cleavage of N-cadherin Is Involved in the Progression of Post-myocardial Infarction Remodeling.
- Author
-
Yoko Kudo-Sakamoto, Hiroshi Akazawa, Kaoru Ito, Jiro Takano, Masamichi Yano, Chizuru Yabumoto, Atsuhiko T. Naito, Toru Oka, Jong-Kook Lee, Yasushi Sakata, Jun-ichi Suzuki, Takaomi C. Saido, and Issei Komuro
- Subjects
- *
CALPAIN , *PROTEOLYSIS , *PROTEOLYTIC enzymes , *CELL death , *MYOCARDIAL infarction - Abstract
Enzymatic proteolysis by calpains, Ca2+-dependent intracellular cysteine proteases, has been implicated in pathological processes such as cellular degeneration or death. Here, we investigated the role of calpain activation in the hearts subjected to myocardial infarction. We produced myocardial infarction in Cast-/- mice deficient for calpastatin, the specific endogenous inhibitory protein for calpains, and Cast+/+ mice. The activity of cardiac calpains in Cast+/+ mice was not elevated within 1 day but showed a gradual elevation after 7 days following myocardial infarction, which was further pronounced in Cast-/- mice. Although the prevalence of cardiomyocyte death was indistinguishable between Cast-/- and Cast+/+ mice, Cast-/- mice exhibited profound contractile dysfunction and chamber dilatation and showed a significant reduction in survival rate after myocardial infarction as compared with Cast+/+ mice. Notably, immunofluorescence revealed that at 28 days after myocardial infarction, calpains were activated in cardiomyocytes exclusively at the border zone and that Cast-/- mice showed higher intensity and a broader extent of calpain activation at the border zone than Cast+/+ mice. In the border zone of Cast-/- mice, pronounced activation of calpains was associated with a decrease in N-cadherin expression and up-regulation of molecular markers for cardiac hypertrophy and fibrosis. In cultured rat neonatal cardiomyocytes, calpain activation by treatment with ionomycin induced cleavage of N-cadherin and decreased expression levels of β-catenin and connexin 43, which was attenuated by calpain inhibitor. These results thus demonstrate that activation of calpains disassembles cell-cell adhesion at intercalated discs by degrading N-cadherin and thereby promotes left ventricular remodeling after myocardial infarction. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
636. Anti-Aβ drug screening platform using human iPS cell-derived neurons for the treatment of Alzheimer's disease.
- Author
-
Naoki Yahata, Masashi Asai, Shiho Kitaoka, Kazutoshi Takahashi, Isao Asaka, Hiroyuki Hioki, Takeshi Kaneko, Kei Maruyama, Takaomi C Saido, Tatsutoshi Nakahata, Takashi Asada, Shinya Yamanaka, Nobuhisa Iwata, and Haruhisa Inoue
- Subjects
Medicine ,Science - Abstract
BackgroundAlzheimer's disease (AD) is a neurodegenerative disorder that causes progressive memory and cognitive decline during middle to late adult life. The AD brain is characterized by deposition of amyloid β peptide (Aβ), which is produced from amyloid precursor protein by β- and γ-secretase (presenilin complex)-mediated sequential cleavage. Induced pluripotent stem (iPS) cells potentially provide an opportunity to generate a human cell-based model of AD that would be crucial for drug discovery as well as for investigating mechanisms of the disease.Methodology/principal findingsWe differentiated human iPS (hiPS) cells into neuronal cells expressing the forebrain marker, Foxg1, and the neocortical markers, Cux1, Satb2, Ctip2, and Tbr1. The iPS cell-derived neuronal cells also expressed amyloid precursor protein, β-secretase, and γ-secretase components, and were capable of secreting Aβ into the conditioned media. Aβ production was inhibited by β-secretase inhibitor, γ-secretase inhibitor (GSI), and an NSAID; however, there were different susceptibilities to all three drugs between early and late differentiation stages. At the early differentiation stage, GSI treatment caused a fast increase at lower dose (Aβ surge) and drastic decline of Aβ production.Conclusions/significanceThese results indicate that the hiPS cell-derived neuronal cells express functional β- and γ-secretases involved in Aβ production; however, anti-Aβ drug screening using these hiPS cell-derived neuronal cells requires sufficient neuronal differentiation.
- Published
- 2011
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.