Network


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

Hotspot


Dive into the research topics where Jichao Yuan is active.

Publication


Featured researches published by Jichao Yuan.


Journal of Neuroinflammation | 2014

Curcumin attenuates acute inflammatory injury by inhibiting the TLR4/MyD88/NF-κB signaling pathway in experimental traumatic brain injury

Haitao Zhu; Chen Bian; Jichao Yuan; Weihua Chu; Xin Xiang; Fei Chen; Cheng-shi Wang; Hua Feng; Jiangkai Lin

BackgroundTraumatic brain injury (TBI) initiates a neuroinflammatory cascade that contributes to substantial neuronal damage and behavioral impairment, and Toll-like receptor 4 (TLR4) is an important mediator of thiscascade. In the current study, we tested the hypothesis that curcumin, a phytochemical compound with potent anti-inflammatory properties that is extracted from the rhizome Curcuma longa, alleviates acute inflammatory injury mediated by TLR4 following TBI.MethodsNeurological function, brain water content and cytokine levels were tested in TLR4-/- mice subjected to weight-drop contusion injury. Wild-type (WT) mice were injected intraperitoneally with different concentrations of curcumin or vehicle 15 minutes after TBI. At 24 hours post-injury, the activation of microglia/macrophages and TLR4 was detected by immunohistochemistry; neuronal apoptosis was measured by FJB and TUNEL staining; cytokines were assayed by ELISA; and TLR4, MyD88 and NF-κB levels were measured by Western blotting. In vitro, a co-culture system comprised of microglia and neurons was treated with curcumin following lipopolysaccharide (LPS) stimulation. TLR4 expression and morphological activation in microglia and morphological damage to neurons were detected by immunohistochemistry 24 hours post-stimulation.ResultsThe protein expression of TLR4 in pericontusional tissue reached a maximum at 24 hours post-TBI. Compared with WT mice, TLR4-/- mice showed attenuated functional impairment, brain edema and cytokine release post-TBI. In addition to improvement in the above aspects, 100 mg/kg curcumin treatment post-TBI significantly reduced the number of TLR4-positive microglia/macrophages as well as inflammatory mediator release and neuronal apoptosis in WT mice. Furthermore, Western blot analysis indicated that the levels of TLR4 and its known downstream effectors (MyD88, and NF-κB) were also decreased after curcumin treatment. Similar outcomes were observed in the microglia and neuron co-culture following treatment with curcumin after LPS stimulation. LPS increased TLR4 immunoreactivity and morphological activation in microglia and increased neuronal apoptosis, whereas curcumin normalized this upregulation. The increased protein levels of TLR4, MyD88 and NF-κB in microglia were attenuated by curcumin treatment.ConclusionsOur results suggest that post-injury, curcumin administration may improve patient outcome by reducing acute activation of microglia/macrophages and neuronal apoptosis through a mechanism involving the TLR4/MyD88/NF-κB signaling pathway in microglia/macrophages in TBI.


Journal of Surgical Research | 2015

Curcumin improves neural function after spinal cord injury by the joint inhibition of the intracellular and extracellular components of glial scar

Jichao Yuan; Mingming Zou; Xin Xiang; Haitao Zhu; Weihua Chu; Wei Liu; Fei Chen; Jiangkai Lin

BACKGROUND Spinal cord injury (SCI) is characterized by a high rate of disability and imposes a heavy burden on society and patients. SCI can activate glial cells and lead to swelling, hyperplasty, and reactive gliosis, which can severely reduce the space for nerve growth. Glial cells can secrete a large amount of extracellular inhibitory components, thus altering the microenvironment of axon growth. Both these factors seriously impede nerve regeneration. In the present study, we investigate whether curcumin (cur), a phytochemical compound with potent anti-inflammatory effect, plays a role in the repair of SCI. MATERIALS AND METHODS We established a rat model of SCI and treated the animals with different concentrations of cur. Using behavioral assessment, immunohistochemistry, real-time polymerase chain reaction, Western blotting, and enzyme-linked immunosorbent assay, we detected the intracellular and extracellular components of glial scar and related cytokines such as tumor necrosis factor (TNF)-α, interleukin (IL)-1β, nuclear factor (NF)-κb, transforming growth factor (TGF)-β1, TGF-β2, and sex determining region Y-box (SOX)-9. RESULTS We found that cur inhibited the expression of proinflammatory cytokines, such as TNF-α, IL-1β, and NF-κb; reduced the expression of the intracellular components glial fibrillary acidic protein through anti-inflammation; and suppressed the reactive gliosis. Also, cur inhibited the generation of TGF-β1, TGF-β2, and SOX-9; decreased the deposition of chondroitin sulfate proteoglycan by inhibiting the transforming growth factors and transcription factor; and improved the microenvironment for nerve growth. Through the joint inhibition of the intracellular and extracellular components of glial scar, cur significantly reduced glial scar volume and improved the Basso, Beattie, and Bresnahan locomotor rating and axon growth. CONCLUSIONS Our data support a role for curcumin in promoting neural function recovery after SCI by the joint inhibition of the intracellular and extracellular components of glial scar, providing an important strategy for treating SCI.


Oncotarget | 2017

M2 microglia promotes neurogenesis and oligodendrogenesis from neural stem/progenitor cells via the PPARγ signaling pathway

Jichao Yuan; Hongfei Ge; Wei Liu; Haitao Zhu; Yaxing Chen; Xuan Zhang; Yang Yang; Yi Yin; Weixiang Chen; Wanjiang Wu; Yunfeng Yang; Jiangkai Lin

Neural stem/progenitor cells (NSPCs) are an important source of cells for cell replacement therapy after nerve injury. How to induce NSPCs differentiation towards neurons and oligodendrocytes is a challenging issue in neuroscience research. In the present study, we polarized microglia into M1 and M2 phenotype, used their supernatants to induce NSPCs differentiation, and investigated the effects of different microglia phenotypes on NSPCs differentiation and their mechanisms. We discovered that, after exposure to M1 phenotype supernatant, NSPCs differentiated into fewer Tuj-1+ and Olig2+ cells, but more GFAP+ cells. Meanwhile, a significantly increased number of Tuj-1+ and Olig2+ cells and smaller number of GFAP+ cells were generated by M2 microglia supernatant-induced NSPCs differentiation. We also observed that 15d-PGJ2, an endogenous ligand of PPARγ, was elevated in M2 phenotype supernatant and could activate PPARγ expression in NSPCs, whereas use of the PPARγ inhibitor GW9662, could reduce the percentage of differentiated neurons and oligodendrocytes. Our study results confirm that M2 microglia supernatant can activate the PPARγ signaling pathway and promote neurogenesis and oligodendrogenesis from NSPCs differentiation. The present study provides a further theoretical basis for induction of NSPCs oriented differentiation.


Neuroreport | 2017

L-borneol induces transient opening of the blood–brain barrier and enhances the therapeutic effect of cisplatin

Yi Yin; Liu Cao; Hongfei Ge; Wangsheng Duanmu; Liang Tan; Jichao Yuan; Chen Tunan; Fei Li; Rong Hu; Fabao Gao; Hua Feng

The blood–brain barrier (BBB) protects the central nervous system from external insults by limiting substance diffusion through the endothelial interface. The presence of the BBB makes drug delivery in neurological disorders very challenging. Cisplatin has been shown to be cytotoxic to glioma cells, but substantial limitations exist in its clinical applications due to difficulties in penetration across the BBB. Here, we show that L-borneol, a messenger drug widely used in traditional Chinese medicine, can induce transient disruption of the BBB after 20 min of oral administration. The permeability of the BBB began to recover within 1 h of the administration of L-borneol. Different dosages of L-borneol (100, 150, 300, 600, and 900 mg/kg) could induce significant Evans blue leakage (P<0.05). Oral administration of L-borneol elevated cisplatin concentrations in peritumoral tissue (1.24±0.12 &mgr;g/g) and tumor loci (1.41±0.13 &mgr;g/g), compared with those in the paraffin control (0.88±0.10 and 0.92±0.15 &mgr;g/g, respectively) (P<0.05). Furthermore, we found that the median survival period of tumor-bearing mice was significantly higher in the cisplatin plus L-borneol group (24.0±4.9 days) than in the cisplatin plus vehicle group (19.3±3.9 days) (P<0.05). The neurological deficits were more severe in the vehicle and cisplatin plus vehicle groups at 14 and 21 days after implantation of intracranial glioma cells than in the cisplatin plus L-borneol group. In conclusion, our results indicate that the transient opening of the BBB induced by L-borneol could enhance cisplatin accumulation within the glioma tissue and improve the survival of tumor-bearing mice.


Neuroscience Letters | 2016

Curcumin reduces brain-infiltrating T lymphocytes after intracerebral hemorrhage in mice

Wei Liu; Jichao Yuan; Haitao Zhu; Xuan Zhang; Lan Li; Xiaojun Liao; Zexian Wen; Yaxing Chen; Hua Feng; Jiangkai Lin

T lymphocytes contribute to inflammation, thereby exacerbating neuronal injury after cerebral ischemia. An increasing amount of evidence indicates that inflammation is a key contributor to intracerebral hemorrhage (ICH)-induced secondary brain injury. Curcumin, a low-molecular-weight curry spice that is derived from the Curcuma longa plant, suppresses T lymphocyte proliferation and migration. Based on these findings, we investigated whether treatment with curcumin would reduce the number of cerebral T lymphocytes in mice with experimentally induced ICH. We found that a large number of T lymphocytes infiltrated the brain at 3days post-ICH. Curcumin significantly improved neurological scores and reduced brain edema in mice with ICH, consistent with a role in reducing neuroinflammation and neurovascular injury. Using flow cytometry, we observed significantly fewer T lymphocytes in brain samples obtained from the curcumin-treated group than in samples obtained from the vehicle-treated group. Moreover, Western blot analysis and immunostaining indicated that treatment with curcumin significantly reduced the expression of a vascular cell adhesion molecule-1 (VCAM-1), interferon-γ (INF-γ) and interleukin-17 (IL-17) in the mouse brain at 72h post-ICH. Our results suggest that administering curcumin may alleviate cerebral inflammation resulting from ICH, at least in part by reducing the infiltration of T lymphocytes into the brain. Therefore, preventing T lymphocytes from infiltrating the brain may become a new strategy for treating clinical ICH.


Brain Research | 2017

Curcumin inhibits glial scar formation by suppressing astrocyte-induced inflammation and fibrosis in vitro and in vivo

Jichao Yuan; Wei Liu; Haitao Zhu; Yaxing Chen; Xuan Zhang; Lan Li; Weihua Chu; Zexian Wen; Hua Feng; Jiangkai Lin

Spinal cord injury (SCI) leads to glial scar formation by astrocytes, which severely hinders neural regeneration. Curcumin (cur) can inhibit glial scar formation, but the underlying mechanism is not fully understood. Using both in vivo and in vitro experiments, the current study investigated the phenotypic transformation of astrocytes following cur and siRNA intervention during the processes of inflammation and fibrosis and determined details of the relationship between cur treatment and the glial scar components GFAP and CSPG. We found that cur and NF-κb p65 siRNA could inhibit astrocyte activation through suppressing NF-κb signaling pathway, which led to down-regulate the expression of chemokines MCP-1, RANTES and CXCL10 released by astrocytes and decreased macrophage and T-cell infiltration, thus reducing the inflammation in the glial scar. In addition, silencing SOX-9 may reduce the deposition of extracellular matrix CSPG; whereas its over-expression could increase the CSPG expression. Cur suppressedSOX-9-inducedCSPG deposition, reduced α-SMA (an important symbol of fibrosis) expression in astrocytes, altered astrocyte phenotype, and inhibited glial scar formation by regulating fibrosis. This study confirmed that cur could regulate both the NF-κb and SOX9 signaling pathways and reduce the expression of intracellular and extracellular glial scar components through dual-target regulating both inflammation and fibrosis after SCI in the rat. This study provides an important hypothesis centered on the dual inhibition of intracellular and extracellular glial scar components as a treatment strategy for SCI.


Scientific Reports | 2016

Poly-L-ornithine enhances migration of neural stem/progenitor cells via promoting α-Actinin 4 binding to actin filaments

Hongfei Ge; Anyong Yu; Jingyu Chen; Jichao Yuan; Yi Yin; Wangsheng Duanmu; Liang Tan; Yang Yang; Chuan Lan; Weixiang Chen; Hua Feng; Rong Hu

The recruitment of neural stem/progenitor cells (NSPCs) for brain restoration after injury is a promising regenerative therapeutic strategy. This strategy involves enhancing proliferation, migration and neuronal differentation of NSPCs. To date, the lack of biomaterials, which facilitate these processes to enhance neural regeneration, is an obstacle for the cell replacement therapies. Our previous study has shown that NSPCs grown on poly-L-ornithine (PO) could proliferate more vigorously and differentiate into more neurons than that on Poly-L-Lysine (PLL) and Fibronectin (FN). Here, we demonstrate that PO could promote migration of NSPCs in vitro, and the underlying mechanism is PO activates α-Actinins 4 (ACTN4), which is firstly certified to be expessed in NSPCs, to promote filopodia formation and therefore enhances NSPCs migration. Taken together, PO might serve as a better candidate for transplanted biomaterials in the regenerative therapeutic strategy, compared with PLL and FN.


Brain Research | 2017

T lymphocytes infiltration promotes blood-brain barrier injury after experimental intracerebral hemorrhage

Xuan Zhang; Wei Liu; Jichao Yuan; Haitao Zhu; Yang Yang; Zexian Wen; Yaxing Chen; Lan Li; Jiangkai Lin; Hua Feng

T lymphocytes migrate into the brain after intracerebral hemorrhage (ICH) and promote cerebral inflammation, thus exacerbating neuronal injury. However, the relationship between of T lymphocytes infiltration and blood-brain barrier (BBB) injury after ICH has not been clarified. In this study, we investigated the spatial-temporal distribution of infiltrating T lymphocytes after ICH in C57BL/6 mice by immunofluorescence and flow cytometry, and the accompanying change rules of BBB permeability were detected by Evans blue dye leakage and tight junction protein expression. Furthermore, T lymphocyte-deficient nude mice and T lymphocyte-decreased C57BL/6 mice treated with fingolimod were used to verify the relationship between T lymphocytes infiltration and BBB leakage after ICH. Here, we reported that brain-infiltrating T lymphocytes in the hemorrhagic hemisphere began to accumulate on the first day and peaked on the fifth day after ICH; BBB leakage also at peaked on the fifth day. Moreover, T lymphocyte-deficient nude mice showed minor BBB leakage after ICH compared with C57BL/6 control mice. Similarly, fingolimod treatment can significantly decrease T lymphocyte infiltration and promote BBB integrity compared with a vehicle control. Overall, our results suggested that suppression of T lymphocyte infiltration may be a novel way to improve BBB integrity after ICH.


Journal of the American Heart Association | 2018

Epothilone B Benefits Nigrostriatal Pathway Recovery by Promoting Microtubule Stabilization After Intracerebral Hemorrhage

Yang Yang; Xuan Zhang; Hongfei Ge; Wei Liu; Eryi Sun; Yuanyuan Ma; Hengli Zhao; Rongwei Li; Weixiang Chen; Jichao Yuan; Qianwei Chen; Yujie Chen; Xin Liu; John H. Zhang; Rong Hu; Xiaotang Fan; Hua Feng

Background Many previous clinical studies have demonstrated that the nigrostriatal pathway, which plays a vital role in movement adjustment, is significantly impaired after stroke, according to medical imaging and autopsies. However, the basic pathomorphological changes have been poorly investigated to date. This study was designed to explore the pathomorphological changes, mechanism, and therapeutic method of nigrostriatal impairment after intracerebral hemorrhage (ICH). Methods and Results Intrastriatal injection of autologous blood or microtubule depolymerization reagent nocodazole was performed to mimic the pathology of ICH in C57/BL6 mice. Immunofluorescence, Western blotting, electron microscopy, functional behavioral tests, and anterograde and retrograde neural circuit tracking techniques were used in these mice. The data showed that the number of dopamine neurons and the dopamine concentration were severely decreased and that fine motor function was impaired after ICH. Microtubule depolymerization was the main contributor to the loss of dopamine neurons and to motor function deficits after ICH, as was also proven by intrastriatal injection of nocodazole. Moreover, administration of the microtubule stabilizer epothilone B (1.5 mg/kg) improved the integrity of the nigrostriatal pathway neural circuit, increased the number of dopamine neurons (4598±896 versus 3125±355; P=0.034) and the dopamine concentration (4.28±0.99 versus 3.08±0.75 ng/mg; P=0.041), and enhanced fine motor functional recovery associated with increased acetylated α‐tubulin expression to maintain microtubule stabilization after ICH. Conclusions Our results clarified the pathomorphological changes of the nigrostriatal pathway after ICH and found that epothilone B helped alleviate nigrostriatal pathway injury after ICH, associated with promoting α‐tubulin acetylation to maintain microtubule stabilization, thus facilitating motor recovery.


Journal of Surgical Research | 2014

Curcumin increased the differentiation rate of neurons in neural stem cells via wnt signaling in vitro study

Fei Chen; Haoxiang Wang; Xin Xiang; Jichao Yuan; Weihua Chu; Xingsen Xue; Haitao Zhu; Hongfei Ge; Mingming Zou; Hua Feng; Jiangkai Lin

Collaboration


Dive into the Jichao Yuan's collaboration.

Top Co-Authors

Avatar

Hua Feng

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Haitao Zhu

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Jiangkai Lin

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Wei Liu

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Xuan Zhang

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Hongfei Ge

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Weihua Chu

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Yaxing Chen

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Fei Chen

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Xin Xiang

Third Military Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge