Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Xigui Chen is active.

Publication


Featured researches published by Xigui Chen.


Human Molecular Genetics | 2015

Comprehensive phosphoproteome analysis unravels the core signaling network that initiates the earliest synapse pathology in preclinical Alzheimer's disease brain

Kazuhiko Tagawa; Hidenori Homma; Ayumu Saito; Kyota Fujita; Xigui Chen; Seiya Imoto; Tsutomu Oka; Hikaru Ito; Kazumi Motoki; Chisato Yoshida; Hiroyuki Hatsuta; Shigeo Murayama; Takeshi Iwatsubo; Satoru Miyano; Hitoshi Okazawa

Using a high-end mass spectrometry, we screened phosphoproteins and phosphopeptides in four types of Alzheimers disease (AD) mouse models and human AD postmortem brains. We identified commonly changed phosphoproteins in multiple models and also determined phosphoproteins related to initiation of amyloid beta (Aβ) deposition in the mouse brain. After confirming these proteins were also changed in and human AD brains, we put the proteins on experimentally verified protein-protein interaction databases. Surprisingly, most of the core phosphoproteins were directly connected, and they formed a functional network linked to synaptic spine formation. The change of the core network started at a preclinical stage even before histological Aβ deposition. Systems biology analyses suggested that phosphorylation of myristoylated alanine-rich C-kinase substrate (MARCKS) by overactivated kinases including protein kinases C and calmodulin-dependent kinases initiates synapse pathology. Two-photon microscopic observation revealed recovery of abnormal spine formation in the AD model mice by targeting a core protein MARCKS or by inhibiting candidate kinases, supporting our hypothesis formulated based on phosphoproteome analysis.


Embo Molecular Medicine | 2015

HMGB1 facilitates repair of mitochondrial DNA damage and extends the lifespan of mutant ataxin-1 knock-in mice

Hikaru Ito; Kyota Fujita; Kazuhiko Tagawa; Xigui Chen; Hidenori Homma; Toshikazu Sasabe; Jun Shimizu; Shigeomi Shimizu; Takuya Tamura; Shin-ichi Muramatsu; Hitoshi Okazawa

Mutant ataxin‐1 (Atxn1), which causes spinocerebellar ataxia type 1 (SCA1), binds to and impairs the function of high‐mobility group box 1 (HMGB1), a crucial nuclear protein that regulates DNA architectural changes essential for DNA damage repair and transcription. In this study, we established that transgenic or virus vector‐mediated complementation with HMGB1 ameliorates motor dysfunction and prolongs lifespan in mutant Atxn1 knock‐in (Atxn1‐KI) mice. We identified mitochondrial DNA damage repair by HMGB1 as a novel molecular basis for this effect, in addition to the mechanisms already associated with HMGB1 function, such as nuclear DNA damage repair and nuclear transcription. The dysfunction and the improvement of mitochondrial DNA damage repair functions are tightly associated with the exacerbation and rescue, respectively, of symptoms, supporting the involvement of mitochondrial DNA quality control by HMGB1 in SCA1 pathology. Moreover, we show that the rescue of Purkinje cell dendrites and dendritic spines by HMGB1 could be downstream effects. Although extracellular HMGB1 triggers inflammation mediated by Toll‐like receptor and receptor for advanced glycation end products, upregulation of intracellular HMGB1 does not induce such side effects. Thus, viral delivery of HMGB1 is a candidate approach by which to modify the disease progression of SCA1 even after the onset.


Scientific Reports | 2015

Fasting activates macroautophagy in neurons of Alzheimer’s disease mouse model but is insufficient to degrade amyloid-beta

Xigui Chen; Kanoh Kondo; Kazumi Motoki; Hidenori Homma; Hitoshi Okazawa

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


Scientific Reports | 2016

HMGB1, a pathogenic molecule that induces neurite degeneration via TLR4-MARCKS, is a potential therapeutic target for Alzheimer's disease.

Kyota Fujita; Kazumi Motoki; Kazuhiko Tagawa; Xigui Chen; Hiroshi Hama; Kazuyuki Nakajima; Hidenori Homma; Takuya Tamura; Hirohisa Watanabe; Masahisa Katsuno; Chiemi Matsumi; Masunori Kajikawa; Takashi Saito; Takaomi Saido; Gen Sobue; Atsushi Miyawaki; Hitoshi Okazawa

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


Human Molecular Genetics | 2016

Targeting TEAD/YAP-transcription-dependent necrosis, TRIAD, ameliorates Huntington's disease pathology.

Ying Mao; Xigui Chen; Min Xu; Kyota Fujita; Kazumi Motoki; Toshikazu Sasabe; Hidenori Homma; Miho Murata; Kazuhiko Tagawa; Takuya Tamura; Julia A. Kaye; Steven Finkbeiner; Giovanni Blandino; Marius Sudol; Hitoshi Okazawa

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


Molecular Psychiatry | 2015

In utero gene therapy rescues microcephaly caused by Pqbp1-hypofunction in neural stem progenitor cells

H. Ito; Hiroki Shiwaku; C. Yoshida; Hidenori Homma; H. Luo; Xigui Chen; Kyota Fujita; Luciana Musante; Utz Fischer; Suzanne Frints; Corrado Romano; Yoshiho Ikeuchi; T. Shimamura; S. Imoto; S. Miyano; S-i Muramatsu; Takeshi Kawauchi; Mikio Hoshino; Marius Sudol; A. Arumughan; E. E. Wanker; T. Rich; Charles E. Schwartz; Fumio Matsuzaki; Azad Bonni; Vera M. Kalscheuer; Hitoshi Okazawa

Human mutations in PQBP1, a molecule involved in transcription and splicing, result in a reduced but architecturally normal brain. Examination of a conditional Pqbp1-knockout (cKO) mouse with microcephaly failed to reveal either abnormal centrosomes or mitotic spindles, increased neurogenesis from the neural stem progenitor cell (NSPC) pool or increased cell death in vivo. Instead, we observed an increase in the length of the cell cycle, particularly for the M phase in NSPCs. Corresponding to the developmental expression of Pqbp1, the stem cell pool in vivo was decreased at E10 and remained at a low level during neurogenesis (E15) in Pqbp1-cKO mice. The expression profiles of NSPCs derived from the cKO mouse revealed significant changes in gene groups that control the M phase, including anaphase-promoting complex genes, via aberrant transcription and RNA splicing. Exogenous Apc4, a hub protein in the network of affected genes, recovered the cell cycle, proliferation, and cell phenotypes of NSPCs caused by Pqbp1-cKO. These data reveal a mechanism of brain size control based on the simple reduction of the NSPC pool by cell cycle time elongation. Finally, we demonstrated that in utero gene therapy for Pqbp1-cKO mice by intraperitoneal injection of the PQBP1-AAV vector at E10 successfully rescued microcephaly with preserved cortical structures and improved behavioral abnormalities in Pqbp1-cKO mice, opening a new strategy for treating this intractable developmental disorder.


Human Molecular Genetics | 2016

RpA1 ameliorates symptoms of mutant ataxin-1 knock-in mice and enhances DNA damage repair

Juliana Bosso Taniguchi; Kanoh Kondo; Kyota Fujita; Xigui Chen; Hidenori Homma; Takeaki Sudo; Ying Mao; Kei Watase; Toshihiro Tanaka; Kazuhiko Tagawa; Takuya Tamura; Shin-ichi Muramatsu; Hitoshi Okazawa

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


Nature Communications | 2017

Developmental YAPdeltaC determines adult pathology in a model of spinocerebellar ataxia type 1

Kyota Fujita; Ying Mao; Shigenori Uchida; Xigui Chen; Hiroki Shiwaku; Takuya Tamura; Hikaru Ito; Kei Watase; Hidenori Homma; Kazuhiko Tagawa; Marius Sudol; Hitoshi Okazawa

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


Scientific Reports | 2016

Identification of hepta-histidine as a candidate drug for Huntington's disease by in silico-in vitro- in vivo-integrated screens of chemical libraries

Tomomi Imamura; Kyota Fujita; Kazuhiko Tagawa; Teikichi Ikura; Xigui Chen; Hidenori Homma; Takuya Tamura; Ying Mao; Juliana Bosso Taniguchi; Kazumi Motoki; Makoto Nakabayashi; Nobutoshi Ito; Kazunori D. Yamada; Kentaro Tomii; Hideyuki Okano; Julia A. Kaye; Steven Finkbeiner; Hitoshi Okazawa

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


Nature Communications | 2018

Targeting Tyro3 ameliorates a model of PGRN-mutant FTLD-TDP via tau-mediated synaptic pathology

Kyota Fujita; Xigui Chen; Hidenori Homma; Kazuhiko Tagawa; Mutsuki Amano; Ayumu Saito; Seiya Imoto; Hiroyasu Akatsu; Yoshio Hashizume; Kozo Kaibuchi; Satoru Miyano; Hitoshi Okazawa

Mutations in the progranulin (PGRN) gene cause a tau pathology-negative and TDP43 pathology-positive form of frontotemporal lobar degeneration (FTLD-TDP). We generated a knock-in mouse harboring the R504X mutation (PGRN-KI). Phosphoproteomic analysis of this model revealed activation of signaling pathways connecting PKC and MAPK to tau prior to TDP43 aggregation and cognitive impairments, and identified PKCα as the kinase responsible for the early-stage tau phosphorylation at Ser203. Disinhibition of Gas6 binding to Tyro3 due to PGRN reduction results in activation of PKCα via PLCγ, inducing tau phosphorylation at Ser203, mislocalization of tau to dendritic spines, and spine loss. Administration of a PKC inhibitor, B-Raf inhibitor, or knockdown of molecules in the Gas6-Tyro3-tau axis rescues spine loss and cognitive impairment of PGRN-KI mice. Collectively, these results suggest that targeting of early-stage and aggregation-independent tau signaling represents a promising therapeutic strategy for this disease.Progranulin (PGRN) mutations cause frontotemporal lobe dementia with TDP-43 pathology. Here the authors develop a mutant PGRN knock-in mouse model of the disease, and show that Tyro3, a tyrosine kinase membrane receptor that acts upstream of PKC and MAPK, is inhibited by PGRN which contributes to pathology in this model.

Collaboration


Dive into the Xigui Chen's collaboration.

Top Co-Authors

Avatar

Hidenori Homma

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Hitoshi Okazawa

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Kyota Fujita

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Kazuhiko Tagawa

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Takuya Tamura

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Kazumi Motoki

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Ying Mao

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Kanoh Kondo

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Hikaru Ito

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge