Network


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

Hotspot


Dive into the research topics where Ching-Yi Lai is active.

Publication


Featured researches published by Ching-Yi Lai.


Journal of General Virology | 2010

Glial activation involvement in neuronal death by Japanese encephalitis virus infection.

Chun-Jung Chen; Yen-Chuan Ou; Shih-Yi Lin; Shue-Ling Raung; Su-Lan Liao; Ching-Yi Lai; Shih-Yun Chen; Jian-Hong Chen

Japanese encephalitis is characterized by profound neuronal destruction/dysfunction and concomitant microgliosis/astrogliosis. Although substantial activation of glia is observed in Japanese encephalitis virus (JEV)-induced Japanese encephalitis, the inflammatory responses and consequences of astrocytes and microglial activation after JEV infection are not fully understood. In this study, infection of cultured neurons/glia with JEV caused neuronal death and glial activation, as evidenced by morphological transformation, increased cell proliferation and elevated tumour necrosis factor (TNF)-alpha, interleukin (IL)-1beta, IL-6 and RANTES (regulated upon activation, normal T-cell expressed and secreted) production. Replication-competent JEV caused all glial responses and neurotoxicity. However, replication-incompetent JEV lost these abilities, except for the ability to change microglial morphology. The bystander damage caused by activated glia also contributed to JEV-associated neurotoxicity. Microglia underwent morphological changes, increased cell proliferation and elevated TNF-alpha, IL-1beta, IL-6 and RANTES expression in response to JEV infection. In contrast, IL-6 and RANTES expression, but no apparent morphological changes, proliferation or TNF-alpha/IL-1beta expression, was demonstrated in JEV-infected astrocytes. Supernatants of JEV-infected microglia, but not JEV-infected astrocytes, induced glial activation and triggered neuronal death. Antibody neutralization studies revealed that TNF-alpha and IL-1beta, but not RANTES or IL-6, released by activated microglia appeared to play roles in JEV-associated neurotoxicity. In conclusion, following JEV infection, neuronal death was accompanied by concomitant microgliosis and astrogliosis, and neurotoxic mediators released by JEV-activated microglia, rather than by JEV-activated astrocytes, had the ability to amplify the microglial response and cause neuronal death.


Life Sciences | 2010

Inhibition of nitric oxide production by quercetin in endotoxin/cytokine-stimulated microglia

Tsung-Kuei Kao; Yen-Chuan Ou; Shue-Ling Raung; Ching-Yi Lai; Su-Lan Liao; Chun-Jung Chen

AIMS Flavonoids possess several biological and pharmacological activities. Quercetin, a naturally occurring flavonoid, has been shown to down-regulate inflammatory responses and provide neuroprotection. However, the mechanisms underlying the anti-inflammatory properties of quercetin are poorly understood. In the present study, we investigated the modulatory effect of quercetin against neuroinflammation. MAIN METHODS We herein describe a potential regulatory mechanism by which quercetin suppresses nitric oxide (NO) production by lipopolysaccharide (LPS)/interferon-gamma (IFN-gamma)-stimulated BV-2 microglial cells. The underlying regulatory cascades were approached by biochemical and pharmacological strategies. KEY FINDINGS Quercetin produced an inhibitory effect on inducible nitric oxide synthase (iNOS) expression and NO production. Biochemical studies revealed that the anti-inflammatory effect of quercetin was accompanied by the down-regulation of extracellular signal-regulated kinase, c-Jun N-terminal kinase, p38, Akt, Src, Janus kinase-1, Tyk2, signal transducer and activator of transcription-1, and NF-kappaB. In addition, quercetin scavenged free radicals and produced inhibitory effects on serine/threonine and tyrosine phosphatase activities. Intriguingly, the accumulation of lipid rafts, which is the critical step for signaling, was disrupted by quercetin. SIGNIFICANCE The data indicate that the anti-inflammatory action of quercetin may be attributable to its raft disrupting and anti-oxidant effects. These distinct mechanisms work in synergy to down-regulate iNOS expression and NO production.


Journal of Nutritional Biochemistry | 2011

Luteolin inhibits cytokine expression in endotoxin/cytokine-stimulated microglia

Tsung-Kuei Kao; Yen-Chuan Ou; Shih-Yi Lin; Hung-Chuan Pan; Pei-Jyuan Song; Shue-Ling Raung; Ching-Yi Lai; Su-Lan Liao; Hsi-Chi Lu; Chun-Jung Chen

Microglial activation plays a pivotal role in the pathogenesis of neurodegenerative disease by producing excessive proinflammatory cytokines and nitric oxide (NO). Luteolin, a naturally occurring polyphenolic flavonoid antioxidant, has potent anti-inflammatory and neuroprotective properties both in vitro and in vivo. However, the molecular mechanism of luteolin-mediated immune modulation in microglia is not fully understood. In the present study, we report the inhibitory effect of luteolin on lipopolysaccharide (LPS)/interferon γ (IFN-γ)-induced NO and proinflammatory cytokine production in rat primary microglia and BV-2 microglial cells. Luteolin concentration-dependently abolished LPS/IFN-γ-induced NO, tumor necrosis factor α (TNF-α) and interleukin 1β (IL-1β) production as well as inducible nitric oxide synthase (iNOS) protein and mRNA expression. Luteolin exerted an inhibitory effect on transcription factor activity including nuclear factor κB (NF-κB), signal transducer and activator of transcription 1 (STAT1) and interferon regulatory factor 1 (IRF-1) in LPS/IFN-γ-activated BV-2 microglial cells. Biochemical and pharmacological studies revealed that the anti-inflammatory effect of luteolin was accompanied by down-regulation of extracellular signal-regulated kinase (ERK), p38, c-Jun N-terminal kinase (JNK), Akt and Src. Further studies have demonstrated that the inhibitory effect of luteolin on intracellular signaling execution and proinflammatory cytokine expression is associated with resolution of oxidative stress and promotion of protein phosphatase activity. Together, these results suggest that luteolin suppresses NF-κB, STAT1 and IRF-1 signaling, thus attenuating inflammatory response of brain microglial cells.


Glia | 2012

Glutamate released by Japanese encephalitis virus-infected microglia involves TNF-α signaling and contributes to neuronal death

Chun-Jung Chen; Yen-Chuan Ou; Cheng-Yi Chang; Hung-Chuan Pan; Su-Lan Liao; Shih-Yun Chen; Shue-Ling Raung; Ching-Yi Lai

The substantial activation of microglia in Japanese encephalitis virus (JEV)‐induced Japanese encephalitis found in numerous studies demonstrates that the disease pathogenesis involves bystander damage caused by microglia‐released mediators. Previously, we reported that microglia synthesized and secreted bioactive mediators with neurotoxic potential into the cultured supernatants in response to JEV infection. In this study, we found that the supernatants of JEV‐infected microglia caused MK801‐inhibitable neuronal damage in cultured neurons, indicating a potential excitotoxic mechanism. Infection with JEV was found to elicit the extracellular glutamate accumulation from microglia but not from neuron and astrocyte cultures. The glutaminase inhibitor 6‐diazo‐5‐oxo‐L‐norleucine, cystine/glutamate antiporter inhibitor α‐aminoadipic acid, and the gap junction inhibitor carbenoxolone reduced JEV infection‐induced microglial glutamate release and neurotoxicity. We further demonstrated that tumor necrosis factor‐alpha (TNF‐α) was a key cytokine which stimulated extensive microglial glutamate release by up‐regulating glutaminase expression via signals involving protein kinase C, cAMP responsive element‐binding protein, and CAAT‐enhancer‐binding protein‐beta. Although the elevated expression of excitatory amino acid transporter 1 and 2 was observed in JEV‐infected cells, the glutamate uptake activity was significantly inhibited by TNF‐α. The JEV infection‐induced alterations, such as the extracellular glutamate release and glutamate‐mediated excitoneurotoxicity, also occurred in neuron/glia cultures. Our findings support a potential link between neuroinflammation and the development of excitotoxic neuronal injury in Japanese encephalitis. The link between neuroinflammation and excitotoxic death may involve a mechanism in which TNF‐α released by microglia plays a facilitory role in glutamate excitoneurotoxicity via up‐regulation of glutamate synthesis and down‐regulation of glutamate uptake.


Journal of Virology | 2014

Infection of Pericytes In Vitro by Japanese Encephalitis Virus Disrupts the Integrity of the Endothelial Barrier

Chun-Jung Chen; Yen-Chuan Ou; Jian-Ri Li; Cheng-Yi Chang; Hung-Chuan Pan; Ching-Yi Lai; Su-Lan Liao; Shue-Ling Raung; Chen-Jung Chang

ABSTRACT Though the compromised blood-brain barrier (BBB) is a pathological hallmark of Japanese encephalitis-associated neurological sequelae, the underlying mechanisms and the specific cell types involved are not understood. BBB characteristics are induced and maintained by cross talk between brain microvascular endothelial cells and neighboring elements of the neurovascular unit. In this study, we show a potential mechanism of disruption of endothelial barrier integrity during the course of Japanese encephalitis virus (JEV) infection through the activation of neighboring pericytes. We found that cultured brain pericytes were susceptible to JEV infection but were without signs of remarkable cytotoxicity. JEV-infected pericytes were found to release biologically active molecules which activated ubiquitin proteasome, degraded zonula occludens-1 (ZO-1), and disrupted endothelial barrier integrity in cultured brain microvascular endothelial cells. Infection of pericytes with JEV was found to elicit elevated production of interleukin-6 (IL-6), which contributed to the aforementioned endothelial changes. We further demonstrated that ubiquitin-protein ligase E3 component n-recognin-1 (Ubr 1) was a key upstream regulator which caused proteasomal degradation of ZO-1 downstream of IL-6 signaling. During JEV central nervous system trafficking, endothelial cells rather than pericytes are directly exposed to cell-free viruses in the peripheral bloodstream. Therefore, the results of this study suggest that subsequent to primary infection of endothelial cells, JEV infection of pericytes might contribute to the initiation and/or augmentation of Japanese encephalitis-associated BBB breakdown in concerted action with other unidentified barrier disrupting factors.


Journal of Neurochemistry | 2012

Endothelial Japanese encephalitis virus infection enhances migration and adhesion of leukocytes to brain microvascular endothelia via MEK-dependent expression of ICAM1 and the CINC and RANTES chemokines

Ching-Yi Lai; Yen-Chuan Ou; Cheng-Yi Chang; Hung-Chuan Pan; Chen-Jung Chang; Su-Lan Liao; Hong-Lin Su; Chun-Jung Chen

Currently, the underlying mechanisms and the specific cell types associated with Japanese encephalitis‐associated leukocyte trafficking are not understood. Brain microvascular endothelial cells represent a functional barrier and could play key roles in leukocyte central nervous system trafficking. We found that cultured brain microvascular endothelial cells were susceptible to Japanese encephalitis virus (JEV) infection with limited amplification. This type of JEV infection had negligible effects on cell viability and barrier integrity. Instead, JEV‐infected endothelial cells attracted more leukocytes adhesion onto surfaces and the supernatants promoted chemotaxis of leukocytes. Infection with JEV was found to elicit the elevated production of intercellular adhesion molecule‐1, cytokine‐induced neutrophil chemoattractant‐1, and regulated‐upon‐activation normal T‐cell expressed and secreted, contributing to the aforementioned leukocyte adhesion and chemotaxis. We further demonstrated that extracellular signal‐regulated kinase was a key upstream regulator which stimulated extensive endothelial gene induction by up‐regulating cytosolic phospholipase A2, NF‐κB, and cAMP response element‐binding protein via signals involving phosphorylation. These data suggest that JEV infection could activate brain microvascular endothelial cells and modify their characteristics without compromising the barrier integrity, making them favorable for the recruitment and adhesion of circulating leukocytes, thereby together with other unidentified barrier‐disrupting mechanisms contributing to Japanese encephalitis and associated neuroinflammation.


The American Journal of Chinese Medicine | 2010

Graptopetalum paraguayense E. Walther Leaf Extracts Protect Against Brain Injury in Ischemic Rats

Tsung-Kuei Kao; Yen-Chuan Ou; Shue-Ling Raung; Wen-Ying Chen; Yu-Ju Yen; Ching-Yi Lai; Su-Tze Chou; Chun-Jung Chen

As practice in folk medicine, Graptopetalum paraguayense E. Walther possesses several biological/pharmacological activities including hepatoprotective, anti-oxidant, and anti-inflammatory. We investigated the neuroprotective potential of Graptopetalum paraguayense E. Walther leaf extracts on inflammation-mediated ischemic brain injury. Water (GWE), 50% alcohol (GE50) extracts of Graptopetalum paraguayense E. Walther, and extracts obtained from further extraction of GE50 with ethyl acetate (GEE) were used. Oral administration of GEE, but not GWE or GE50, for 2 weeks protected animals against cerebral ischemia/reperfusion brain injury. The neuroprotective effect of GEE was accompanied by reductions in brain infarction, neurological deficits, caspase-3 activity, malondialdehyde content, microglia activation, and inducible nitric oxide synthase (iNOS) expression. Since microglia-mediated inflammation plays critical roles in ischemic brain injury, anti-inflammatory potential of Graptopetalum paraguayense E. Walther leaf extracts was further investigated on lipopolysaccharide (LPS)/interferon-gamma (IFN-gamma-activated BV-2 microglial cells. GEE decreased H(2)O(2)- and LPS/IFN-gamma-induced free radical generation and LPS/IFN-gamma-induced iNOS expression. Mechanistic study revealed that the neuroactive effects of GEE were markedly associated with anti-oxidative potential, activation of serine/threonine and tyrosine phosphatases, and down-regulation of extracellular signal-regulated kinase, c-Jun N-terminal kinase, p38, Akt, Src, Janus kinase-1, Tyk2, signal transducer and activator of transcription-1, and NF-kappaB and might be attributed to the presence of polyphenolic compounds such as gallic acid, genistin, daidzin, and quercetin. Together, our findings point out its potential therapeutic strategies that target microglia activation, oxidative stress, and iNOS expression to reduce ischemic brain injury and suggest that Graptopetalum paraguayense E. Walther leaf extracts represent a valuable source for the development of neuroprotective agents.


Glia | 2015

Disruption of in vitro endothelial barrier integrity by Japanese encephalitis virus‐Infected astrocytes

Cheng-Yi Chang; Jian-Ri Li; Wen-Ying Chen; Yen-Chuan Ou; Ching-Yi Lai; Yu-Hui Hu; Chih-Cheng Wu; Chen-Jung Chang; Chun-Jung Chen

Blood–brain barrier (BBB) characteristics are induced and maintained by crosstalk between brain microvascular endothelial cells and neighboring cells. Using in vitro cell models, we previously found that a bystander effect was a cause for Japanese encephalitis‐associated endothelial barrier disruption. Brain astrocytes, which neighbor BBB endothelial cells, play roles in the maintenance of BBB integrity. By extending the scope of relevant studies, a potential mechanism has been shown that the activation of neighboring astrocytes could be a cause of disruption of endothelial barrier integrity during the course of Japanese encephalitis viral (JEV) infection. JEV‐infected astrocytes were found to release biologically active molecules that activated ubiquitin proteasome, degraded zonula occludens‐1 (ZO‐1) and claudin‐5, and disrupted endothelial barrier integrity in cultured brain microvascular endothelial cells. JEV infection caused astrocytes to release vascular endothelial growth factor (VEGF), interleukin‐6 (IL‐6), and matrix metalloproteinases (MMP‐2/MMP‐9). Our data demonstrated that VEGF and IL‐6 released by JEV‐infected astrocytes were critical for the proteasomal degradation of ZO‐1 and the accompanying disruption of endothelial barrier integrity through the activation of Janus kinase‐2 (Jak2)/signal transducer and activator of transcription‐3 (STAT3) signaling as well as the induction of ubiquitin–protein ligase E3 component, n‐recognin‐1 (Ubr 1) in endothelial cells. MMP‐induced endothelial barrier disruption was accompanied by MMP‐mediated proteolytic degradation of claudin‐5 and ubiquitin proteasome‐mediated degradation of ZO‐1 via extracellular VEGF release. Collectively, these data suggest that JEV infection could activate astrocytes and cause release of VEGF, IL‐6, and MMP‐2/MMP‐9, thereby contributing, in a concerted action, to the induction of Japanese encephalitis‐associated BBB breakdown. GLIA 2015;63:1915–1932


Toxicology Letters | 2011

Signaling cascades mediate astrocyte death induced by zinc.

Su-Lan Liao; Yen-Chuan Ou; Shih-Yi Lin; Tsung-Kuei Kao; Hung-Chuan Pan; Cheng-Yi Chang; Ching-Yi Lai; Hsi-Chi Lu; Wen-Yi Wang; Chun-Jung Chen

Zinc overload is known to cause the death of neural cells. Although the activation of extracellular signal-regulated kinase (ERK) and cytosolic phospholipase A(2) (cPLA(2)) have been implicated in zinc-induced astrocyte death, the detailed mechanisms of their activation and upstream regulatory cascades are incompletely understood. Here, we report that protein kinase C (PKC)- and Src-related Ras/Raf/ERK cascades and ERK-associated cPLA(2) participate in astrocyte death caused by ZnCl(2). Sustained exposure to ZnCl(2) caused damage to astrocytes in a time- and concentration-dependent manner. The cell death caused by ZnCl(2) was accompanied by increased reactive oxygen species (ROS) generation, PKC-α membrane association, Src phosphorylation, Ras membrane association, Raf phosphorylation, ERK phosphorylation, and cPLA(2) activation, and decreased protein phosphatase activity. Pharmacological studies revealed that these activations/inactivations all contributed to ZnCl(2)-induced astrocyte death. ROS, such as superoxide, appear to be a key trigger in response to ZnCl(2) treatment in astrocytes because of the attenuations in protein phosphatase inhibition, signaling activation, and cell death by antioxidant treatments. Mechanistic studies had suggested that ROS/PKC-α/Ras/Raf/ERK and ROS/Src/Ras/Raf/ERK were potential signals linking zinc and cPLA(2). These observations indicated that ROS/PKC-α/Ras/Raf/ERK and ROS/Src/Ras/Raf/ERK signaling and cPLA(2) were actively involved in zinc-induced astrocyte damage.


Brain Behavior and Immunity | 2017

Interplay of inflammatory gene expression in pericytes following Japanese encephalitis virus infection

Cheng-Yi Chang; Jian-Ri Li; Yen-Chuan Ou; Shih-Yi Lin; Ya-Yu Wang; Wen-Ying Chen; Yu-Hui Hu; Ching-Yi Lai; Chen-Jung Chang; Chun-Jung Chen

Neuroinflammation is a pathological hallmark and has been implicated in the pathogenesis of Japanese encephalitis. Although brain pericytes show regulatory effects on neuroinflammation, their involvement in Japanese encephalitis-associated neuroinflammation is not understood. Here, we demonstrated that brain microvascular pericytes could be an alternative cellular source for the induction and/or amplification of neuroinflammation caused by Japanese encephalitis virus (JEV) infection. Infection of cultured pericytes with JEV caused profound production of IL-6, RANTES, and prostaglandin E2 (PGE2). Mechanistic studies revealed that JEV infection elicited an elevation of the toll-like receptor 7 (TLR7)/MyD88 signaling axis, leading to the activation of NF-κB through IKK signaling and p65 phosphorylation as well as cAMP response element-binding protein (CREB) via phosphorylation. We further demonstrated that extracellular signal-regulated kinase (ERK) could be an alternative regulator in transducing signals to NF-κB, CREB, and cytosolic phospholipase A2 (cPLA2) through the phosphorylation mechanism. Released IL-6 and RANTES played an active role in the disruption of endothelial barrier integrity and leukocyte chemotaxis, respectively. cPLA2/PGE2 had a role in activating NF-κB and CREB DNA-binding activities and inflammatory cytokine transcription via the EP2/cAMP/PKA mechanism in an autocrine loop. These inflammatory responses and biochemical events were also detected in the brain of JEV-infected mice. The current findings suggest that pericytes might have pathological relevance in Japanese encephalitis-associated neuroinflammation through a TLR7-related mechanism. The consequences of pericyte activation are their ability to initiate and/or amplify inflammatory cytokine expression by which cellular function of endothelial cells and leukocytes are regulated in favor of CNS infiltration by leukocytes.

Collaboration


Dive into the Ching-Yi Lai's collaboration.

Top Co-Authors

Avatar

Chun-Jung Chen

National Chung Hsing University

View shared research outputs
Top Co-Authors

Avatar

Su-Lan Liao

National Chung Hsing University

View shared research outputs
Top Co-Authors

Avatar

Cheng-Yi Chang

Central Taiwan University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Hung-Chuan Pan

National Yang-Ming University

View shared research outputs
Top Co-Authors

Avatar

Shih-Yi Lin

National Yang-Ming University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jian-Ri Li

Chung Shan Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Wen-Ying Chen

National Chung Hsing University

View shared research outputs
Top Co-Authors

Avatar

Chen-Jung Chang

University of the Sciences

View shared research outputs
Researchain Logo
Decentralizing Knowledge