Network


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

Hotspot


Dive into the research topics where Yajie Liang is active.

Publication


Featured researches published by Yajie Liang.


Journal of Neuroinflammation | 2012

Microglial TIR-domain-containing adapter-inducing interferon-β (TRIF) deficiency promotes retinal ganglion cell survival and axon regeneration via nuclear factor-κB.

Sen Lin; Yajie Liang; Jiqiang Zhang; Chen Bian; Hongli Zhou; Qiang Guo; Ying Xiong; Shurong Li; Bingyin Su

BackgroundTIR-domain-containing adapter-inducing interferon-β (TRIF) is the sole downstream adaptor of Toll-like receptor (TLR)3, which is one of the major signaling pathways in immune cells leading to neuroinflammation in the central nervous system. Overexpression of TRIF may lead to activation of inflammatory responses, and contribute to pathophysiological progression in both acute and chronic neurodegenerative retinal diseases. In the present study, was aimed to elucidate the contributions of TRIF to optic nerve (ON) regeneration and retinal ganglion cell (RGC) survival following injury to the ON, a widely studied model of central nervous system injury and of degenerative diseases such as glaucoma.MethodsWe used retrograde labeling with a fluorochrome, hydroxystilbamidine (Fluorogold) to evaluate RGC survival, and immunostaining with growth-associated protein-43 to evaluate axon regeneration in an ON crush model. Changes in microglial cytokines following RGC injury was examined with ELISA and real-time PCR. In vivo studies were carried out in wild-type and trif-/- mice. A Transwell co-culture system and migration test were used to mimic the crosstalk between microglia and RGCs. TRIF-associated downstream adaptors were determined by western blotting.ResultsCompared with wild-type (WT) mice, TRIF knockout (KO) mice displayed a robust ability to regenerate axons 3 or 7 days after nerve injury. In addition, RGC survival was considerably higher in trif-/- than in WT mice. ON lesion induced less microglial activation in trif-/- than in WT mice. and more WT microglia distorted and migrated toward the foramen opticum. In the transwell system, few trif-/- microglia migrated through the membrane when stimulated by the performed lesion on RGC axons in a transwell system. Inactivation of microglial cells in trif-/- mice was associated with reduced production of inflammatory cytokines, as detected with real-time RT-PCR and ELISA. Furthermore western blot analysis showed that activation of known downstream effectors of TRIF, including TBK1, IKKε and NF-κB, were significantly inhibited by TRIF deficiency.ConclusionOur results indicate that TRIF deficiency promotes ON axon regeneration by attenuating microglial activation and consequently reducing the release of harmful cytokines via NF-κB inactivation.


Neuroscience Letters | 2010

Effects of C3 deficiency on inflammation and regeneration following spinal cord injury in mice

Qiang Guo; Shurong Li; Yajie Liang; Yanling Zhang; Jiqiang Zhang; Can Wen; Sen Lin; Hanzhi Wang; Bingyin Su

Inflammation can activate the complement system, which in turn enhances inflammation and aggravates secondary injury after spinal cord injury (SCI). As the three complement activation pathways converge at the cleavage of C3, we investigated whether inhibiting complement activation in C3-deficient mice would reduce secondary injury after SCI and improve axon regeneration. Weight-drop contusion injury (5g, 6cm) was created in wild-type or C3-deficient mice. Astrocytes (ASTs) activation, TNF-α expression, and axon regeneration were investigated in vivo. In other studies, dorsal root ganglia (DRGs) were co-cultured with mechanically injured ASTs in vitro to evaluate effects on neurite outgrowth. Our results show that, after injury, C3-deficient mice exhibit higher BBB scores than wild-type mice. In addition, ASTs activation was inhibited, TNF-α expression process was delayed in vivo and inhibited in vitro, and nerve fiber regeneration was improved in C3-deficient mice. DRGs co-cultured with mechanically injured ASTs from C3-deficient mice also showed improved neurite outgrowth. We conclude that C3 deficiency can inhibit inflammation through suppressing ASTs activation and TNF-α expression, thereby reducing secondary injury and improving neural regeneration and functional recovery after SCI. The above results suggest that complement inhibition may be a potential therapy to promote central nervous system regeneration by targeting C3.


Journal of Neurochemistry | 2008

Intrastriatal injection of colchicine induces striatonigral degeneration in mice

Yajie Liang; Shurong Li; Can Wen; Yanling Zhang; Qiang Guo; Hanzhi Wang; Bingyin Su

Recent studies from environmental toxicology and molecular genetics demonstrate that midbrain dopamine (DA) neurons are particularly vulnerable to microtubule depolymerizing agents, indicating the involvement of microtubule dysfunction in the pathogenesis of Parkinson’s disease. Here we show that intrastriatal injection of colchicine (COL), a well‐known microtubule disruptor, induced degeneration of striatonigral pathway. Microtubule disruption caused by unilateral injection of COL blocked the retrograde axonal transport of fluorogold previously injected into striatum and induced substantial death of striatal and DA neurons in substantia nigra pars compacta. Furthermore, COL‐induced pathologic changes were associated with robust glial reaction, which may be conducive to the degeneration of striatonigral pathway. We also found that intrastriatal injection of COL resulted in side bias in spontaneous turning activities and apomorphine‐induced rotational behavior. Together, our results provide in vivo data lending support to the concept that microtubule dysfunction may play a significant role in the death of DA neurons, though glial reaction may be involved and contribute to the degenerative process. Moreover, intrastriatal COL may serve as another experimental model of striatonigral degeneration (Parkinson’s variant of multiple system atrophy), given the concurrent loss of both striatal and DA neurons.


Neuroscience Letters | 2012

Expression profiling of Rab GTPases reveals the involvement of Rab20 and Rab32 in acute brain inflammation in mice

Yajie Liang; Sen Lin; Liyun Zou; Hongli Zhou; Jiqiang Zhang; Bingyin Su; Ying Wan

Rab GTPases have emerged as central regulators of vesicle trafficking and are essential for cytokine production during the pathogenesis of neuroinflammation. To characterize the roles of different Rab proteins in brain inflammation, we used quantitative PCR (qPCR) to examine the expression profiles of all members of the Rab family in an experimental model of brain inflammation in mice. We found that Rab20 and Rab32 were substantially up-regulated during the acute phase of inflammation. The increased expression of Rab20 was also confirmed by immunostaining of inflamed brains at different timepoints. The concomitant overexpression of Rabs (Rab20 and Rab32) and early response proinflammatory cytokines (TNF-α and IL-1β) suggested that these Rabs may be important for subsequent inflammatory responses in brain. Furthermore, we found that the expression of certain Rabs was dramatically reduced in cultured primary microglia, which was not observed in the in vivo profiling. In N9, a microglial cell line, however, there was no increase in the expression of Rab20 or Rab32, but Rab3c was significantly overexpressed. These results collectively indicate that Rabs may participate in inflammatory response in microglia during brain inflammation. The differential regulation of individual Rabs in different experimental systems is a caveat for the analysis of Rab functions.


Brain Research | 2007

Complement 3-deficient mice are not protected against MPTP-induced dopaminergic neurotoxicity.

Yajie Liang; Shurong Li; Qiang Guo; Yanliang Zhang; Can Wen; Qiang Zou; Bingyin Su

Recent studies have invoked inflammation as a major contributor to the pathogenesis of Parkinsons disease (PD). Emerging evidence indicated that components of complement system may be involved in such disorder and contribute to its development. We thus observed the influence of deficiency of complement 3 (C3), the key component of complement system, on the death of dopaminergic neurons in substantia nigra pars compacta (SNpc) and the loss of dopaminergic fibers in striatum induced by acute or chronic administration of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Immunohistochemical staining of dopaminergic neurons in SNpc and neurochemical analysis of dopamine and its metabolites in striata revealed that there was no significant difference between the two genotypes. Longer survival time also indicated that C3 might not mediate the spontaneous recovery of dopaminergic fibers in mouse striatum acutely challenged by MPTP. We conclude that, despite growing evidence indicating the involvement of complement system in the pathogenesis of PD, our data do not support a role for C3 in this established model of PD, as indicated by results from HPLC analysis and immunohistochemical staining.


Neuroscience Letters | 2006

Potential neuroprotective effect of low dose whole-body γ-irradiation against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced dopaminergic toxicity in C57 mice

Yajie Liang; Shurong Li; Qiang Zou; Bingyin Su

Low dose whole-body gamma-irradiation is recently reported to confer neuroprotection against optic nerve crush and contusive spinal cord injury. Here, we extended the study and investigated whether the pretreatment of a single low dose whole-body gamma-irradiation may have a preventive effect in MPTP-induced model of PD. One week after the last MPTP treatment, HPLC determination of striatal dopamine and immunostaining for tyrosine hydroxylase (TH), CD11b and GFAP to detect dopamine neurons and associated glial reaction in the substantia nigra pars compacta (SNpc) were performed. MPTP treatment reduced striatal DA levels significantly; nigral TH immunoreactivity was reduced to a lower extent; robust gliosis was also observed in SNpc. We found that 3.5 Gy irradiation but not 5.5 Gy restores the level of DA and its metabolites decreased by MPTP. However, there was no difference in the number of TH positive neurons between 3.5 Gy irradiated and saline treated mice after MPTP treatment. Irradiation also did not have obvious influence on microgliosis and astroglial reaction induced by MPTP treatment. In conclusion, the results presented here demonstrated that low dose whole-body gamma-irradiation renders neuroprotection against MPTP-mediated damage of striatal dopaminergic nerve fibers, though it does not seem to influence the MPTP-induced reduction of SNpc dopaminergic neurons and associated glial responses.


Neuroscience Letters | 2009

Potential mechanisms of neuroprotection induced by low dose total-body γ-irradiation in C57 mice administered with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)

Yanling Zhang; Shurong Li; Yajie Liang; Can Wen; Qiang Guo; Bingyin Su

Low dose total-body gamma-irradiation (TBI) was reported to confer neuroprotection against MPTP-induced dopaminergic neurotoxicity. After being pretreated with a single low dose (0.5Gy, 2.0Gy or 3.5Gy) TBI, C57BL/6 mice were administered with MPTP (15mg/kg, four times, 2h apart) intraperitoneally (i.p.). In the group pretreated with 2.0Gy TBI, with lower lymphocytes number, neuroprotection was found by High Performance Liquid Chromatography (HPLC) determination of the striatal dopamine. Contrarily, in the group pretreated with 0.5Gy TBI, with higher lymphocytes number, dopaminergic neuron toxicity was enhanced. So it was probably the decrease of lymphocytes, not the radiation hormesis that rendered the potential neuroprotection. And it was the balance between radiation injury and lymphocytopenia neuroprotection that decided the effect of low dose gamma-irradiation on MPTP-induced dopaminergic neurotoxicity.


International Journal of Developmental Neuroscience | 2008

Microtubule dysfunction play a significant role in the demise of DA neurons

Bingyin Su; Yajie Liang; S.R. Li

Netrin-1 functions as an attractant or repellent during axon guidance. In the visual system, netrin has been implicated in retinal ganglion cell (RGC) axonpathfinding. InXenopus,netrin attracts RGC axons out of the eye but once they reach the optic tectum they are repelled by netrin suggesting that netrin may function as a target recognition signal in the brain. Herewe examinedwhether netrin-1 is involved in wiring events that follow successful axon pathfinding byexaminingnetrin’s contribution toaxonarborizationandsynapse formation at the target. Expression patterns of netrin and its receptors in stage 44/45 tadpoles, when RGC axons actively branch and form synapses, supported this possibility. Time-lapse confocal microscopy of individual RGCs co-expressing GFP-synaptobrevin and DsRed2 demonstrated a role for DCC-mediated netrin signaling in synaptogenesis and axon arbor growth. In controls, RGC axon arbors become morphologically more complex by the dynamic addition, elimination, and stabilizationofpresynaptic sites and axon branches.Microinjection of netrin-1 into the tectumproducedmore dynamic branchingbehaviorwhich resulted in a significant increase in total axon branch number by 24 h relative to controls. This increase in branch addition was paralleled by an increase in presynaptic site number that was maintained through the 24-h period. Microinjection of DCC function blocking antibodies prevented the increase in branch number and axon arbor growth normallyobserved incontrol axonsaswell as theassociated increase in presynaptic site number. Dynamic analysis of axon arbors demonstrated that the effects of anti-DCC on axon morphology and presynaptic connectivity were due to a specific decrease in new branch addition within the entire 24-h imaging period. Together, these results indicate that in the absence ofDCC signalingRGCaxons fail to branch and differentiate, and support a novel role for netrin in later phases of retinotectal development.


Autonomic Neuroscience: Basic and Clinical | 2008

Hybrid synapse formation between spinal motoneurons and superior cervical ganglion neurons in vitro: A study of the functional reconstruction of visceral organs

Can Wen; Shurong Li; Hanzhi Wang; Jiqiang Zhang; Yajie Liang; Qiang Guo; Yanling Zhang; Chuan-Guo Xiao; Bingyin Su


Brain Research | 2008

Corrigendum to “Complement 3-deficient mice are not protected against MPTP-induced dopaminergic neurotoxicity” [Brain Res. 1178 (2007) 132─140]

Yajie Liang; Shurong Li; Qiang Guo; Yanling Zhang; Can Wen; Qiang Zou; Bingyin Su

Collaboration


Dive into the Yajie Liang's collaboration.

Top Co-Authors

Avatar

Bingyin Su

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Shurong Li

Chengdu Medical College

View shared research outputs
Top Co-Authors

Avatar

Qiang Guo

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Can Wen

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Yanling Zhang

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Jiqiang Zhang

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Hanzhi Wang

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Qiang Zou

Chengdu Medical College

View shared research outputs
Top Co-Authors

Avatar

Sen Lin

Chengdu Medical College

View shared research outputs
Top Co-Authors

Avatar

Hongli Zhou

Chengdu Medical College

View shared research outputs
Researchain Logo
Decentralizing Knowledge