Network


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

Hotspot


Dive into the research topics where Peihui Lin is active.

Publication


Featured researches published by Peihui Lin.


Nature | 2009

NAADP mobilizes calcium from acidic organelles through two-pore channels

Peter J. Calcraft; Margarida Ruas; Zui Pan; Xiaotong Cheng; Abdelilah Arredouani; Xuemei Hao; Jisen Tang; Katja Rietdorf; Lydia Teboul; Kai Ting Chuang; Peihui Lin; Rui Xiao; Chunbo Wang; Yingmin Zhu; Yakang Lin; Christopher N. Wyatt; John Parrington; Jianjie Ma; A. Mark Evans; Antony Galione; Michael X. Zhu

Ca2+ mobilization from intracellular stores represents an important cell signalling process that is regulated, in mammalian cells, by inositol-1,4,5-trisphosphate (InsP3), cyclic ADP ribose and nicotinic acid adenine dinucleotide phosphate (NAADP). InsP3 and cyclic ADP ribose cause the release of Ca2+ from sarcoplasmic/endoplasmic reticulum stores by the activation of InsP3 and ryanodine receptors (InsP3Rs and RyRs). In contrast, the nature of the intracellular stores targeted by NAADP and the molecular identity of the NAADP receptors remain controversial, although evidence indicates that NAADP mobilizes Ca2+ from lysosome-related acidic compartments. Here we show that two-pore channels (TPCs) comprise a family of NAADP receptors, with human TPC1 (also known as TPCN1) and chicken TPC3 (TPCN3) being expressed on endosomal membranes, and human TPC2 (TPCN2) on lysosomal membranes when expressed in HEK293 cells. Membranes enriched with TPC2 show high affinity NAADP binding, and TPC2 underpins NAADP-induced Ca2+ release from lysosome-related stores that is subsequently amplified by Ca2+-induced Ca2+ release by InsP3Rs. Responses to NAADP were abolished by disrupting the lysosomal proton gradient and by ablating TPC2 expression, but were only attenuated by depleting endoplasmic reticulum Ca2+ stores or by blocking InsP3Rs. Thus, TPCs form NAADP receptors that release Ca2+ from acidic organelles, which can trigger further Ca2+ signals via sarcoplasmic/endoplasmic reticulum. TPCs therefore provide new insights into the regulation and organization of Ca2+ signals in animal cells, and will advance our understanding of the physiological role of NAADP.


Nature Cell Biology | 2009

MG53 nucleates assembly of cell membrane repair machinery

Chuanxi Cai; Haruko Masumiya; Noah Weisleder; Noriyuki Matsuda; Miyuki Nishi; Moonsun Hwang; Jae-Kyun Ko; Peihui Lin; Angela Thornton; Xiaoli Zhao; Zui Pan; Shinji Komazaki; Marco Brotto; Hiroshi Takeshima; Jianjie Ma

Dynamic membrane repair and remodelling is an elemental process that maintains cell integrity and mediates efficient cellular function. Here we report that MG53, a muscle-specific tripartite motif family protein (TRIM72), is a component of the sarcolemmal membrane-repair machinery. MG53 interacts with phosphatidylserine to associate with intracellular vesicles that traffic to and fuse with sarcolemmal membranes. Mice null for MG53 show progressive myopathy and reduced exercise capability, associated with defective membrane-repair capacity. Injury of the sarcolemmal membrane leads to entry of the extracellular oxidative environment and MG53 oligomerization, resulting in recruitment of MG53-containing vesicles to the injury site. After vesicle translocation, entry of extracellular Ca2+ facilitates vesicle fusion to reseal the membrane. Our data indicate that intracellular vesicle translocation and Ca2+-dependent membrane fusion are distinct steps involved in the repair of membrane damage and that MG53 may initiate the assembly of the membrane repair machinery in an oxidation-dependent manner.


Nature | 2007

TRIC channels are essential for Ca2+ handling in intracellular stores.

Masayuki Yazawa; Christopher Ferrante; Jue Feng; Kazuhiro Mio; Toshihiko Ogura; Miao Zhang; Peihui Lin; Zui Pan; Shinji Komazaki; Kazuhiro Kato; Miyuki Nishi; Xiaoli Zhao; Noah Weisleder; Chikara Sato; Jianjie Ma; Hiroshi Takeshima

Cell signalling requires efficient Ca2+ mobilization from intracellular stores through Ca2+ release channels, as well as predicted counter-movement of ions across the sarcoplasmic/endoplasmic reticulum membrane to balance the transient negative potential generated by Ca2+ release. Ca2+ release channels were cloned more than 15 years ago, whereas the molecular identity of putative counter-ion channels remains unknown. Here we report two TRIC (trimeric intracellular cation) channel subtypes that are differentially expressed on intracellular stores in animal cell types. TRIC subtypes contain three proposed transmembrane segments, and form homo-trimers with a bullet-like structure. Electrophysiological measurements with purified TRIC preparations identify a monovalent cation-selective channel. In TRIC-knockout mice suffering embryonic cardiac failure, mutant cardiac myocytes show severe dysfunction in intracellular Ca2+ handling. The TRIC-deficient skeletal muscle sarcoplasmic reticulum shows reduced K+ permeability, as well as altered Ca2+ ‘spark’ signalling and voltage-induced Ca2+ release. Therefore, TRIC channels are likely to act as counter-ion channels that function in synchronization with Ca2+ release from intracellular stores.


Circulation Research | 2010

Cardioprotection of Ischemia/Reperfusion Injury by Cholesterol-Dependent MG53-Mediated Membrane Repair

Xianhua Wang; Wenjun Xie; Yi Zhang; Peihui Lin; Liang Han; Peidong Han; Yanru Wang; Zheng Chen; Guangju Ji; Ming Zheng; Noah Weisleder; Rui-Ping Xiao; Hiroshi Takeshima; Jianjie Ma; Heping Cheng

Rationale: Unrepaired cardiomyocyte membrane injury causes irreplaceable cell loss, leading to myocardial fibrosis and eventually heart failure. However, the cellular and molecular mechanisms of cardiac membrane repair are largely unknown. MG53, a newly identified striated muscle-specific protein, is involved in skeletal muscle membrane repair. But the role of MG53 in the heart has not been determined. Objective: We sought to investigate whether MG53 mediates membrane repair in cardiomyocytes and, if so, the cellular and molecular mechanism underlying MG53-mediated membrane repair in cardiomyocytes. Moreover, we determined possible cardioprotective effect of MG53-mediated membrane repair. Methods and Results: We demonstrated that MG53 is crucial to the emergency membrane repair response in cardiomyocytes and protects the heart from stress-induced loss of cardiomyocytes. Disruption of the sarcolemmal membrane by mechanical, electric, chemical, or metabolic insults caused rapid and robust translocation of MG53 toward the injury sites. Ablation of MG53 prevented sarcolemmal resealing after infrared laser–induced membrane damage in intact heart, and exacerbated mitochondrial dysfunction and loss of cardiomyocytes during ischemia/reperfusion injury. Unexpectedly, the MG53-mediated cardiac membrane repair was mediated by a cholesterol-dependent mechanism: depletion of membrane cholesterol abolished, and its recovery restored injury-induced membrane translocation of MG53. The redox status of MG53 did not affect initiation of MG53 translocation, whereas MG53 oxidation conferred stability to the membrane repair patch. Conclusions: Thus, cholesterol-dependent MG53-mediated membrane repair is a vital, heretofore unappreciated cardioprotective mechanism against a multitude of insults and may bear important therapeutic implications.


Science Translational Medicine | 2012

Recombinant MG53 protein modulates therapeutic cell membrane repair in treatment of muscular dystrophy

Noah Weisleder; Norio Takizawa; Peihui Lin; Xianhua Wang; Chunmei Cao; Yan Zhang; Tao Tan; Christopher Ferrante; H. Zhu; Pin-Jung Chen; Rosalie Yan; Matthew Sterling; Xiaoli Zhao; Moonsun Hwang; Miyuki Takeshima; Chuanxi Cai; Heping Cheng; Hiroshi Takeshima; Rui-Ping Xiao; Jianjie Ma

Recombinant human MG53 protein can increase membrane repair after injury in cells and can reduce pathology in animal models of muscle injury and muscular dystrophy. Mending Muscle To repair a torn muscle, one might require a little bit of ice and a lot of rest. For those with Duchenne muscular dystrophy (DMD), however, muscle degeneration is not as easily repaired, and patients ultimately experience difficulty standing, walking, and breathing. DMD results from a lack of the protein dystrophin, which is located at the cell membrane to help muscle fibers repair themselves. There is no cure for DMD, but Weisleder and colleagues have now shown that exogenous delivery of a different repair protein, Mitsugumin 53 (MG53), to cells can prevent muscle damage in cell culture and in mice. The authors first showed that muscle and nonmuscle cells treated with recombinant human MG53 (rhMG53) in vitro were resistant to mechanical, chemical, and photo damage because MG53 localized to the injury site and provided protection. In vivo, Weisleder and colleagues showed that dystrophin-deficient mdx mice treated intramuscularly or intravenously with rhMG53 displayed reduced muscle damage and decreased muscle pathology compared to saline-treated controls, even in the presence of a membrane-damaging toxin. This repair process also worked in muscle fibers isolated from mdx mice that were deficient in either of two natural repair proteins, MG53 or dysferlin, suggesting that exogenous delivery of rhMG53 works by a new mechanism—other than the intracellular machinery—to patch up damaged cell membrane. Soluble MG53 protein therapy could be a viable treatment for DMD that avoids the well-known limitations of dystrophin gene replacement therapy. Toward translation, Weisleder et al. have further demonstrated that exogenous MG53 is nontoxic and safe in animals. The ability of the protein to preserve muscle function and to enhance repair capacity in humans has yet to be shown, but additional studies in larger animals and human muscle fibers will give a clearer indication of its therapeutic potential. Mitsugumin 53 (MG53), a muscle-specific TRIM family protein, is an essential component of the cell membrane repair machinery. Here, we examined the translational value of targeting MG53 function in tissue repair and regenerative medicine. Although native MG53 protein is principally restricted to skeletal and cardiac muscle tissues, beneficial effects that protect against cellular injuries are present in nonmuscle cells with overexpression of MG53. In addition to the intracellular action of MG53, injury to the cell membrane exposes a signal that can be detected by MG53, allowing recombinant MG53 protein to repair membrane damage when provided in the extracellular space. Recombinant human MG53 (rhMG53) protein purified from Escherichia coli fermentation provided dose-dependent protection against chemical, mechanical, or ultraviolet-induced damage to both muscle and nonmuscle cells. Injection of rhMG53 through multiple routes decreased muscle pathology in the mdx dystrophic mouse model. Our data support the concept of targeted cell membrane repair in regenerative medicine, and present MG53 protein as an attractive biological reagent for restoration of membrane repair defects in human diseases.


Nature Communications | 2013

MG53-induced IRS-1 ubiquitination negatively regulates skeletal myogenesis and insulin signalling

Jae Sung Yi; Jun Sub Park; Young Mi Ham; Nga Nguyen; Na Rae Lee; Jin Hong; Bong Woo Kim; Hyun Jung Lee; Chang Seok Lee; Byung Cheon Jeong; Hyun Kyu Song; Hana Cho; Yoon Ki Kim; Jae Seon Lee; Kyong Soo Park; Haksub Shin; Inho Choi; Seung Hee Lee; Woo Jin Park; Shi Young Park; Cheol Soo Choi; Peihui Lin; Malith Karunasiri; Tao Tan; Pu Duann; H. Zhu; Jianjie Ma; Young Gyu Ko

Mitsugumin 53 (MG53) negatively regulates skeletal myogenesis by targeting insulin receptor substrate 1 (IRS-1). Here, we show that MG53 is an ubiquitin E3 ligase that induces IRS-1 ubiquitination with the help of an E2-conjugating enzyme, UBE2H. Molecular manipulations that disrupt the E3-ligase function of MG53 abolish IRS-1 ubiquitination and enhance skeletal myogenesis. Skeletal muscles derived from the MG53-/- mice show an elevated IRS-1 level with enhanced insulin signalling, which protects the MG53-/- mice from developing insulin resistance when challenged with a high-fat/high-sucrose diet. Muscle samples derived from human diabetic patients and mice with insulin resistance show normal expression of MG53, indicating that altered MG53 expression does not serve as a causative factor for the development of metabolic disorders. Thus, therapeutic interventions that target the interaction between MG53 and IRS-1 may be a novel approach for the treatment of metabolic diseases that are associated with insulin resistance.


Journal of Biological Chemistry | 2011

Polymerase Transcriptase Release Factor (PTRF) Anchors MG53 Protein to Cell Injury Site for Initiation of Membrane Repair

H. Zhu; Peihui Lin; Gejing De; Kyoung-Han Choi; Hiroshi Takeshima; Noah Weisleder; Jianjie Ma

Plasma membrane repair is an essential process for maintenance of homeostasis at the cellular and tissue levels, whereas compromised repair capacity contributes to degenerative human diseases. Our recent studies show that MG53 is essential for muscle membrane repair, and defects in MG53 function are linked to muscular dystrophy and cardiac dysfunction. Here we report that polymerase I and transcript release factor (PTRF), a gene known to regulate caveolae membrane structure, is an indispensable component of the membrane repair machinery. PTRF acts as a docking protein for MG53 during membrane repair potentially by binding exposed membrane cholesterol at the injury site. Cells lacking expression of endogenous PTRF show defective trafficking of MG53 to membrane injury sites. A mutation in PTRF associated with human disease results in aberrant nuclear localization of PTRF and disrupts MG53 function in membrane resealing. Although RNAi silencing of PTRF leads to defective muscle membrane repair, overexpression of PTRF can rescue membrane repair defects in dystrophic muscle. Our data suggest that membrane-delimited interaction between MG53 and PTRF contributes to initiation of cell membrane repair, which can be an attractive target for treatment or prevention of tissue injury in human diseases.


Journal of Neuropathology and Experimental Neurology | 2011

Dysferlin, annexin A1, and mitsugumin 53 are upregulated in muscular dystrophy and localize to longitudinal tubules of the T-system with stretch.

Leigh B. Waddell; Frances A. Lemckert; Xi F. Zheng; Jenny Tran; Frances J. Evesson; J. Hawkes; Angela Lek; Neil Street; Peihui Lin; Nigel F. Clarke; Andrew P. Landstrom; Michael J. Ackerman; Noah Weisleder; Jianjie Ma; Kathryn N. North; Sandra T. Cooper

Mutations in dysferlin cause an inherited muscular dystrophy because of defective membrane repair. Three interacting partners of dysferlin are also implicated in membrane resealing: caveolin-3 (in limb girdle muscular dystrophy type 1C), annexin A1, and the newly identified protein mitsugumin 53 (MG53). Mitsugumin 53 accumulates at sites of membrane damage, and MG53-knockout mice display a progressive muscular dystrophy. This study explored the expression and localization of MG53 in human skeletal muscle, how membrane repair proteins are modulated in various forms of muscular dystrophy, and whether MG53 is a primary cause of human muscle disease. Mitsugumin 53 showed variable sarcolemmal and/or cytoplasmic immunolabeling in control human muscle and elevated levels in dystrophic patients. No pathogenic MG53 mutations were identified in 50 muscular dystrophy patients, suggesting that MG53 is unlikely to be a common cause of muscular dystrophy in Australia. Western blot analysis confirmed upregulation of MG53, as well as of dysferlin, annexin A1, and caveolin-3 to different degrees, in different muscular dystrophies. Importantly, MG53, annexin A1, and dysferlin localize to the t-tubule network and show enriched labeling at longitudinal tubules of the t-system in overstretch. Our results suggest that longitudinal tubules of the t-system may represent sites of physiological membrane damage targeted by this membrane repair complex.


Molecular Therapy | 2012

Enhancing Muscle Membrane Repair by Gene Delivery of MG53 Ameliorates Muscular Dystrophy and Heart Failure in δ-Sarcoglycan-deficient Hamsters

Bo He; Ru Hang Tang; Noah Weisleder; Bin Xiao; Zhenhua Yuan; Chuanxi Cai; H. Zhu; Peihui Lin; Chunping Qiao; Jianbin Li; Christina Mayer; Juan Li; Jianjie Ma; Xiao Xiao

Muscular dystrophies (MDs) are caused by genetic mutations in over 30 different genes, many of which encode for proteins essential for the integrity of muscle cell structure and membrane. Their deficiencies cause the muscle vulnerable to mechanical and biochemical damages, leading to membrane leakage, dystrophic pathology, and eventual loss of muscle cells. Recent studies report that MG53, a muscle-specific TRIM-family protein, plays an essential role in sarcolemmal membrane repair. Here, we show that systemic delivery and muscle-specific overexpression of human MG53 gene by recombinant adeno-associated virus (AAV) vectors enhanced membrane repair, ameliorated pathology, and improved muscle and heart functions in δ-sarcoglycan (δ-SG)-deficient TO-2 hamsters, an animal model of MD and congestive heart failure. In addition, MG53 overexpression increased dysferlin level and facilitated its trafficking to muscle membrane through participation of caveolin-3. MG53 also protected muscle cells by activating cell survival kinases, such as Akt, extracellular signal-regulated kinases (ERK1/2), and glycogen synthase kinase-3β (GSK-3β) and inhibiting proapoptotic protein Bax. Our results suggest that enhancing the muscle membrane repair machinery could be a novel therapeutic approach for MD and cardiomyopathy, as demonstrated here in the limb girdle MD (LGMD) 2F model.


Journal of Biological Chemistry | 2006

The presenilin-2 loop peptide perturbs intracellular Ca2+ homeostasis and accelerates apoptosis

Chuanxi Cai; Peihui Lin; King Ho Cheung; Na Li; Christina Levchook; Zui Pan; Christopher Ferrante; Gabrielle L. Boulianne; J. Kevin Foskett; David Danielpour; Jianjie Ma

In cells undergoing apoptosis, a 22-amino-acid presenilin-2-loop peptide (PS2-LP, amino acids 308–329 in presenilin-2) is generated through cleavage of the carboxyl-terminal fragment of presenilin-2 by caspase-3. The impact of PS2-LP on the progression of apoptosis, however, is not known. Here we show that PS2-LP is a potent inducer of the mitochondrial-dependent cell death pathway when transduced as a fusion protein with HIV-TAT. Biochemical and functional studies demonstrate that TAT-PS2-LP can interact with the inositol 1,4,5-trisphosphate receptor and activate Ca2+ release from the endoplasmic reticulum. These results indicate that PS2-LP-mediated alteration of intracellular Ca2+ homeostasis may be linked to the acceleration of apoptosis. Therefore, targeting the function of PS2-LP could provide a useful therapeutic tool for the treatment of cancer and degenerative diseases.

Collaboration


Dive into the Peihui Lin's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Zui Pan

Ohio State University

View shared research outputs
Top Co-Authors

Avatar

H. Zhu

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Chuanxi Cai

Albany Medical College

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tao Tan

Ohio State University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge