Network


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

Hotspot


Dive into the research topics where Jingquan Gao is active.

Publication


Featured researches published by Jingquan Gao.


Brain Research Bulletin | 2011

Baicalin attenuates global cerebral ischemia/reperfusion injury in gerbils via anti-oxidative and anti-apoptotic pathways.

Yonggang Cao; Xiaoyuan Mao; Chunyan Sun; Ping Zheng; Jingquan Gao; Xiaorui Wang; Dongyu Min; Hongli Sun; Ni Xie; Jiqun Cai

Baicalin is an important medicinal herb purified from the dry roots of Scutellaria baicalensis Georgi. The present study was undertaken to evaluate the neuroprotective effects of baicalin in gerbils subjected to transient global cerebral ischemic-reperfusion injury. Baicalin at doses of 50, 100 and 200mg/kg was intraperitoneally injected into the gerbils immediately after cerebral ischemia. Seven days after reperfusion, hematoxylin and eosin (HE) staining was performed to analyze hippocampal CA1 pyramidal damage histopathologically. In addition, in order to understand the potential protective mechanism of baicalin, we examined anti-oxidative enzymes, such superoxide dismutase (SOD), glutathione peroxidase (GSH-PX), non-enzymatic scavenger glutathione (GSH) and measured the content of malondialdehyde (MDA) in hippocampus. The mRNA and protein expressions of BDNF were determined in ischemic hippocampus by real-time RT-PCR and Western blot, respectively. Evidence for neuronal apoptosis was detected by real-time RT-PCR, Western blot and caspase-3 activity measurement. Histopathological examination showed that the administration of baicalin by the dose of 100 and 200mg/kg significantly attenuated ischemia-induced neuronal cell damage. Reduced level of MDA, obviously elevated activities of SOD and GSH as well as GSH-PX were also found in baicalin-treated groups. Further investigation demonstrated that treatment with baicalin remarkably promoted the expression of BDNF and inhibited the expression of caspase-3 at mRNA and protein levels by real-time RT-PCR and Western blot, respectively. Besides, caspase-3 activity assay also elucidated that the administration of baicalin could significantly suppress caspase-3 in ischemic gerbils hippocampus. Theses findings suggest that baicalins neuroprotection appears to be associated with its anti-oxidative and anti-apoptotic properties in global cerebral ischemia in the gerbils.


Life Sciences | 2017

Fingolimod (FTY720) attenuates social deficits, learning and memory impairments, neuronal loss and neuroinflammation in the rat model of autism

Hongmei Wu; Xuelai Wang; Jingquan Gao; Shuang Liang; Yanqiu Hao; Caihong Sun; Wei Xia; Yonggang Cao; Lijie Wu

Aims: To investigate the effect of FTY720 on the valproic acid (VPA) rat model of autism. Main methods: As an animal model of autism, we used intraperitoneal injection of VPA on embryonic day 12.5 in Wistar rats. The pups were given FTY720 orally at doses of 0.25, 0.5 and 1 mg/kg daily from postnatal day 15 to 35. Social behavior, spatial learning and memory were assessed at the end of FTY720 treatment. The histological change, oxidative stress, neuroinflammatory responses, and apoptosis‐related proteins in the hippocampus were evaluated. Key findings: FTY720 (1 mg/kg) administration to VPA‐exposed rats (1) improved social behavior, spatial learning and memory impairment; (2) resulted in a reduction in neuronal loss and apoptosis of pyramidal cells in hippocampal CA1 regions; (3) inhibited activation of microglial cells, in turn lowering the level of pro‐inflammatory cytokines interleukin‐1&bgr; (IL‐1&bgr;) and IL‐6 in the hippocampus; (4) changed Malondialdehyde (MDA) levels, Glutathione (GSH) levels, superoxide dismutase (SOD) activity and Glutathione Peroxidase (GSH‐Px) activity in the hippocampus; (6) inhibited the elevated Bax and caspase‐3 protein levels and enhanced the relative expression level of Bcl‐2 in the hippocampus; and (7) increased phospho‐Ca2 +/calmodulin‐dependent protein kinase II (p‐CaMKII), phospho‐cAMP‐response element binding protein (p‐CREB) and Brain Derived Neurotrophic Factor (BDNF) protein expression in the hippocampus. Significance: FTY720 rescues social deficit, spatial learning and memory impairment in VPA‐exposed rats. FTY720 exerts both a direct protection for neurons and an indirect modulation of inflammation‐mediated neuron loss as a possible mechanism of neuroprotection.


International Journal of Developmental Neuroscience | 2016

Neuroprotective effects of docosahexaenoic acid on hippocampal cell death and learning and memory impairments in a valproic acid-induced rat autism model

Jingquan Gao; Xuelai Wang; Hongli Sun; Yonggang Cao; Shuang Liang; Han Wang; Yanming Wang; Feng Yang; Fengyu Zhang; Lijie Wu

Prenatal exposure to valproic acid (VPA) in rat offspring is capable of inducing experimental autism with neurobehavioral aberrations. This study investigated the effect of docosahexaenoic acid (DHA) on hippocampal cell death, learning and memory alteration in an experimental rat autism model. We found that DHA supplementation (75, 150 or 300 mg/kg/day, 21 days) rescued the VPA (600 mg/kg) induced DHA reduction in plasma and hippocampus in a dose‐dependent manner, increased the levels of hippocampal p‐CaMKII and p‐CREB without affecting total protein level, and altered BDNF‐AKT‐Bcl‐2 signaling pathway, as well as inhibited the activity of caspase‐3. DHA also influenced the content of malondialdehyde (MDA) and the activities of antioxidant enzymes in the VPA‐treated offspring. Consistent with the previous results, we also observed that 300 mg/kg DHA supplementation markedly increased the cell survival, decreased the cell apoptosis, and increased mature neuronal cell in the hippocampus in VPA‐treated offspring. Utilizing the Morris water maze test, we found that DHA prevented cognitive impairment in offspring of VPA‐treated rats. The data suggested that DHA may play a neuroprotective role in hippocampal neuronal cell and ameliorates dysfunctions in learning and memory in this rat autism model. Thus, DHA could be used as treatment intervention for mitigating behavioral dysfunctions in autism spectrum disorder (ASD).


Journal of Nutritional Biochemistry | 2016

Maternal DHA supplementation protects rat offspring against impairment of learning and memory following prenatal exposure to valproic acid

Jingquan Gao; Hongmei Wu; Yonggang Cao; Shuang Liang; Caihong Sun; Peng Wang; Ji Wang; Hongli Sun; Lijie Wu

Docosahexaenoic acid (22:6n-3; DHA) is known to play a critical role in postnatal brain development. However, there have been no studies investigating the preventive effect of DHA on prenatal valproic acid (VPA)-induced behavioral and molecular alterations in offspring. The present study was to evaluate the neuroprotective effects in offspring using maternal feeding of DHA to rats exposed to VPA in pregnancy. In the present study, rats were exposed to VPA on day 12.5 of pregnancy; DHA was administered at the dosages of 100, 300 and 500 mg/kg/day for 3 weeks from day 1 to 21 of pregnancy. The results showed that maternal feeding of DHA to the prenatal exposed to VPA (1) prevented VPA-induced learning and memory impairment but did not change social-related behavior, (2) increased total DHA content in offspring plasma and hippocampus, (3) rescued VPA-induced neuronal loss and apoptosis of pyramidal cells in hippocampal CA1, (4) influenced the content of malondialdehyde and glutathione and the activities of superoxide dismutase and glutathione in the hippocampus, (5) altered levels of apoptosis-related proteins (Bcl-2, Bax and caspase-3) and inhibited the activity of caspase-3 in offspring hippocampus and (6) enhanced relative levels of p-CaMKII and p-CREB proteins in the hippocampus. These findings suggest that maternal feeding with DHA may prevent prenatal VPA-induced impairment of learning and memory, normalize several different molecules associated with oxidative stress and apoptosis in the hippocampus of offspring, and exert preventive effects on prenatal VPA-induced brain dysfunction.


Psychopharmacology | 2018

Modulation of sphingosine 1-phosphate (S1P) attenuates spatial learning and memory impairments in the valproic acid rat model of autism

Hongmei Wu; Quanzhi Zhang; Jingquan Gao; Caihong Sun; Jia Wang; Wei Xia; Yonggang Cao; Yanqiu Hao; Lijie Wu

RationaleAutism spectrum disorders (ASD) are a set of pervasive neurodevelopmental disorders that manifest in early childhood, and it is growing up to be a major cause of disability in children. However, the etiology and treatment of ASD are not well understood. In our previous study, we found that serum levels of sphingosine 1-phosphate (S1P) were increased significantly in children with autism, indicating that S1P levels may be involved in ASD.ObjectiveThe objective of this study was to identify a link between increased levels of S1P and neurobehavioral changes in autism.MethodsWe utilized a valproic acid (VPA) -induced rat model of autism to evaluate the levels of S1P and the expression of sphingosine kinase (SphK), a key enzyme for S1P production, in serum and hippocampal tissue. Furthermore, we assessed cognitive functional changes and histopathological and neurochemical alterations in VPA-exposed rats after SphK blockade to explore the possible link between increased levels of S1P and neurobehavioral changes in autism.ResultsWe found that SphK2 and S1P are upregulated in hippocampal tissue from VPA-exposed rats, while pharmacological inhibition of SphK reduced S1P levels, attenuated spatial learning and memory impairments, increased the expression of phosphorylated CaMKII and CREB and autophagy-related proteins, inhibited cytochrome c release, decreased the expression of apoptosis related proteins, and protected against neuronal loss in the hippocampus.ConclusionWe have demonstrated that an increased level of SphK2/S1P is involved in the spatial learning and memory impairments of autism, and this signaling pathway represents a novel therapeutic target and direction for future studies.


Phytomedicine | 2018

Allicin improves the function of cardiac microvascular endothelial cells by increasing PECAM-1 in rats with cardiac hypertrophy

Pilong Shi; Yonggang Cao; Jingquan Gao; Jing Ren; Lina Ba; Chao Song; Hanping Qia; Wei Huang; Bowen Fu; Xueying Guan; Hongli Sun

OBJECTIVE Cardiac microvascular damage is significantly associated with the development of cardiac hypertrophy (CH). Researchers found that allicin could inhibit CH, but the relationship between cardiac microvessel and the inhibition of allicin on CH has not been reported. We aimed to investigate the effect of allicin on the function of cardiac microvascular endothelial cells (CMECs) in CH rat. MATERIALS AND METHODS The hemodynamic parameters were measured by BL-420F biological function experimental system and the indicators of the ventricular structure and function were measured by echocardiographic system. MTT assay was performed to assess the cell viability. Nitrite detection was performed to detect nitric oxide content. The morphology and molecular characteristics were detected by electron micrographs, immunofluorescence, quantitative real-time polymerase chain reaction (qRT-PCR), western blot. Wound healing experiment, analysis of tube formation and shear adaptation were performed to assess CMECs migration ability, angiogenesis and shear-responsiveness respectively. RESULT Our findings have identified that microvascular density was decreased by observing the expression of platelet endothelial cell adhesion molecule-1 (PECAM-1) in CH rats. Interestingly, allicin improved the distribution and expression of PECAM-1. Meanwhile, allicin enhanced the migration and angiogenesis ability of CMECs, activated PECAM-1-PI3K-AKT-eNOS signaling pathway, however, the role of allicin was disappear after PECAM-1 was silenced. Allicin decreased the expression of caspase-3 and receptor interacting protein 3 (RIP3), inhibited necroptosis, and increased the levels of Angiopoietin-2 (Ang-2) and platelet-derived growth factor receptor-β (PDGFR-β). Under 10 dyn/cm2 condition, allicin advanced the modification ability of CMECss shear-adaptation by activating PECAM-1. CONCLUSION Allicin provided cardioprotection for CH rats by improving the function of CMECs through increasing the expression of PECAM-1.


Journal of Cellular and Molecular Medicine | 2018

Activation of transient receptor potential vanilloid 3 channel (TRPV3) aggravated pathological cardiac hypertrophy via calcineurin/NFATc3 pathway in rats

Qianhui Zhang; Hanping Qi; Yonggang Cao; Pilong Shi; Chao Song; Lina Ba; Yunping Chen; Jingquan Gao; Shuzhi Li; Baiyan Li; Hongli Sun

Cardiac hypertrophy is a compensatory response to mechanical stimuli and neurohormonal factors, ultimately progresses to heart failure. The proteins of some transient receptor potential (TRP) channels, Ca2+‐permeable nonselective cation channel, are highly expressed in cardiomyocytes, and associated with the occurrence of cardiac hypertrophy. Transient receptor potential vanilloid 3 (TRPV3) is a member of TRP, however, the functional role of TRPV3 in cardiac hypertrophy remains unclear. TRPV3 was elevated in pathological cardiac hypertrophy, but not in swimming exercise‐induced physiological cardiac hypertrophy in rats. TRPV3 expression was also increased in Ang II–induced cardiomyocyte hypertrophy in vitro, which was remarkably increased by carvacrol (a nonselective TRPV channel agonist), and reduced by ruthenium red (a nonselective TRPV channel antagonist). Interestingly, we found that activated TRPV3 in Ang II–induced cardiomyocyte hypertrophy was accompanied with increasing intracellular calcium concentration, promoting calcineurin, and phosphorylated CaMKII protein expression, and enhancing NFATc3 nuclear translocation. However, blocking or knockdown of TRPV3 could inhibit the expressions of calcineurin, phosphorylated CaMKII and NFATc3 protein by Western blot. In conclusion, the activation of TRPV3 aggravated pathological cardiac hypertrophy through calcineurin/NFATc3 signalling pathway and correlated with the protein expression levels of calcineurin, phosphorylated CaMKII and NFATc3, revealing that TRPV3 might be a potential therapeutic target for cardiac hypertrophy.


Journal of Psychiatry & Neuroscience | 2016

Potential serum biomarkers from a metabolomics study of autism

Han Wang; Shuang Liang; Maoqing Wang; Jingquan Gao; Caihong Sun; Jia Wang; Wei Xia; Shiying Wu; Susan Sumner; Fengyu Zhang; Changhao Sun; Lijie Wu


Journal of Physiological Sciences | 2014

The mechanism of neurogenic pulmonary edema in epilepsy.

Hong Zhao; Guijun Lin; Mumu Shi; Jingquan Gao; Yanming Wang; Hongzhi Wang; Hongli Sun; Yonggang Cao


Naunyn-schmiedebergs Archives of Pharmacology | 2018

Transient receptor potential vanilloid-3 (TRPV3) activation plays a central role in cardiac fibrosis induced by pressure overload in rats via TGF-β 1 pathway

Yan Liu; Hanping Qi; Mingyao E; Pilong Shi; Qianhui Zhang; Shuzhi Li; Ye Wang; Yonggang Cao; Yunping Chen; Lina Ba; Jingquan Gao; Wei Huang; Hongli Sun

Collaboration


Dive into the Jingquan Gao's collaboration.

Top Co-Authors

Avatar

Hongli Sun

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Yonggang Cao

China Medical University (PRC)

View shared research outputs
Top Co-Authors

Avatar

Caihong Sun

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Lijie Wu

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Shuang Liang

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Wei Xia

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Han Wang

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Hongmei Wu

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Jia Wang

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Lina Ba

Harbin Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge