Network


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

Hotspot


Dive into the research topics where Mei Hu is active.

Publication


Featured researches published by Mei Hu.


CNS Neuroscience & Therapeutics | 2012

PPARγ Agonist Pioglitazone Reverses Memory Impairment and Biochemical Changes in a Mouse Model of Type 2 Diabetes Mellitus

Li-Ying Jiang; Xiao-Yun Wang; Li-Ping Liu; Yan Long; Mei Hu; Ming-Xing Liao; Qi-Long Ding; Wei Hu; Jia-chang Li; Hao Hong

Aims: Pioglitazone, known as a peroxisome proliferator‐activated receptor γ (PPARγ) agonist, is used to treat type 2 diabetes mellitus (T2DM). T2DM has been associated with reduced performance on numerous domains of cognitive function. Here, we investigated the effects of pioglitazone on memory impairment in a mouse model with defects in insulin sensitivity and secretion, namely high‐fat diet (HFD) streptozotocin (STZ)‐induced diabetic mice. Methods: ICR mice were fed with HFD for 4 weeks and then injected with a single low dose of STZ followed by continued HFD feeding for an additional 4 weeks. Pioglitazone (18 mg/kg, 9 mg/kg body weight) was orally administered for 6 weeks once daily. Y‐maze test and Morris water maze test (MWM) were employed for testing learning and memory. Serum glucose, serum insulin, serum triglyceride, brain β‐amyloid peptide (Aβ), brain β‐site amyloid precursor protein cleaving enzyme (BACE1), brain nuclear factor κB (NF‐κB), and brain receptor for advanced glycation end products (RAGE) were also tested. Results: The STZ/HFD diabetic mice, characterized by hyperglycemia, hyperlipemia and hypoinsulinemia, performed poorly on Y‐maze and MWM hence reflecting impairment of learning and memory behavior with increases of Aβ40/Aβ42, BACE1, NF‐κB, and RAGE in brain. Treatment of PPARγ agonist, pioglitazone (18 or 9 mg/kg body weight), significantly reversed diabetes‐induced impairment of learning and memory behavior, which is involved in decreases of Aβ40/Aβ42 via inhibition of NF‐κB, BACE1 and RAGE in brain as well as attenuation of hyperglycemia, hyperlipemia, and hypoinsulinemia. Conclusions: It is concluded that PPARγ agonist pioglitazone may be considered as potential pharmacological agents for the management of cognitive dysfunction in T2DM.


Neuropharmacology | 2013

Leukotriene D4 induces cognitive impairment through enhancement of CysLT1R-mediated amyloid-β generation in mice

Xiao-Yun Wang; Hao Hong; Yan Long; Yong-qi Li; Guo-qing Xiang; Li-Ying Jiang; Han-Ting Zhang; Li-Ping Liu; Ming-Xing Miao; Mei Hu; Ting-Ting Zhang; Wei Hu; Hui Ji; Feng-Ying Ye

Amyloid plaques in the extracellular parenchyma mainly consist of amyloid-β peptides (Aβ), one of the pathological hallmarks in Alzheimers disease (AD). In the present study, we examined neuroinflammation, amyloidogenesis, and memory performance following intracerebral infusions of leukotriene D4 (LTD4) in mice. The results demonstrated that intracerebral infusions of LTD4 (1 ng/mouse) produced memory impairment as determined by Morris water maze test and Y-maze test in mice, and caused the accumulation of Aβ1-40 and Aβ1-42 in the hippocampus and cortex through increased activity of β- and γ-secretases accompanied with increased expression of amyloid precursor protein (APP). LTD4 also induced expression of cysteinyl leukotriene receptor 1 (CysLT(1)R) and NF-κB p65 in the hippocampus and cortex. Pretreatment with pranlukast (1.5 ng/mouse, intracerebroventricularly), a CysLT(1)R antagonist, blocked LTD4-induced amyloidogenesis, memory deficits. Pranlukast (0.6 μM) also prevented LTD4 (20 nM)-induced amyloidogenesis in the cultured neurons in vitro. Moreover, LTD4-induced increases in CysLT(1)R and NF-κB p65 in the brain were also attenuated by pranlukast. These results suggest that LTD4 increases Aβ peptide burden via activation of CysLT(1)R, which further affects APP levels and activity of β- and γ-secretases via the NF-κB pathway. Our findings identify CysLT(1)R signaling as a novel proinflammatory and proamyloidogenic pathway, and suggest a rationale for development of therapeutics targeting the CysLT(1)R in neuroinflammatory diseases such as AD.


Neuropharmacology | 2016

Antidiabetic drugs restore abnormal transport of amyloid-β across the blood–brain barrier and memory impairment in db/db mice

Fang Chen; Rong Rong Dong; Kai Long Zhong; Arijit Ghosh; Su Su Tang; Yan Long; Mei Hu; Ming Xing Miao; Jian Min Liao; Hong Bing Sun; Ling Yi Kong; Hao Hong

Previous studies have shown significant changes in amyloid-β (Aβ) transport across the blood-brain barrier (BBB) under diabetic conditions with hypoinsulinemia, which is involved in diabetes-associated cognitive impairment. Present study employed db/db mice with hyperinsulinemia to investigate changes in Aβ transport across the BBB, hippocampal synaptic plasticity, and restorative effects of antidiabetic drugs. Our results showed that db/db mice exhibited similar changes in Aβ transport across the BBB to that of insulin-deficient mice. Chronic treatment of db/db mice with antidiabetic drugs such as metformin, glibenclamide and insulin glargine significantly decreased Aβ influx across the BBB determined by intra-arterial infusion of (125)I-Aβ(1-40), and expression of the receptor for advanced glycation end products (RAGE) participating in Aβ influx. Insulin glargine, but not, metformin or glibenclamide increased Aβ efflux across the BBB determined by stereotaxic intra-cerebral infusion of (125)I-Aβ(1-40), and expression of the low-density lipoprotein receptor related protein 1 (LRP1) participating in Aβ efflux. Moreover, treatment with these drugs significantly decreased hippocampal Aβ(1-40) or Aβ(1-42) and inhibited neuronal apoptosis. The drugs also ameliorated memory impairment confirmed by improved performance on behavioral tasks. However, insulin glargine or glibenclamide, but not metformin, restored hippocampal synaptic plasticity characterized by enhancing in vivo long-term potentiation (LTP). Further study found that these three drugs significantly restrained NF-κB, but only insulin glargine enhanced peroxisome proliferator-activated receptor γ (PPARγ) activity at the BBB in db/db mice. Our data indicate that the antidiabetic drugs can partially restore abnormal Aβ transport across the BBB and memory impairment under diabetic context.


Acta Pharmacologica Sinica | 2013

Pioglitazone ameliorates memory deficits in streptozotocin-induced diabetic mice by reducing brain β-amyloid through PPARγ activation.

Li-Ping Liu; Tianhua Yan; Li-Ying Jiang; Wei Hu; Meng Hu; Chao Wang; Qian Zhang; Yan Long; Jiang-qing Wang; Yong-qi Li; Mei Hu; Hao Hong

Aim:To examine the effects of pioglitazone, a PPARγ agonist, on memory performance and brain amyloidogenesis in streptozotocin (STZ)-induced diabetic mice.Methods:ICR male mice were injected with STZ (150 mg/kg, iv) to induce experimental diabetes. Pioglitazone (9 and 18 mg·kg-1·d-1, po) was administered for 6 weeks. Passive avoidance and Morris water maze (MWM) tests were used to evaluate cognitive function. The blood glucose and serum insulin levels were detected using the glucose oxidase method and an ELISA assay, respectively. β-amyloid (Aβ), β-amyloid precursor protein (APP), β-amyloid precursor protein cleaving enzyme 1 (BACE1), NF-κB p65, the receptor for advanced glycation end products (RAGE) and PPARγ in the brains were analyzed using Western blotting assays.Results:The STZ-induced diabetic mice characterized by hyperglycemia and hypoinsulinemia performed poorly in both the passive avoidance and MWM tests, accompanied by increased Aβ1–40/Aβ1–42, APP, BACE1, NF-κB p65 and RAGE levels and decreased PPARγ level in the hippocampus and cortex. Chronic pioglitazone treatment significantly ameliorated the memory deficits and amyloidogenesis of STZ-induced diabetic mice, and suppressed expression of APP, BACE1, RAGE and NF-κB p65, and activated PPARγ in the hippocampus and cortex. However, pioglitazone did not significantly affect blood glucose and insulin levels.Conclusion:Pioglitazone ameliorates memory deficits in STZ-induced diabetic mice by reducing brain Aβ level via activation of PPARγ, which is independent of its effects on blood glucose and insulin levels. The results suggest that pioglitazone may be used for treating the cognitive dysfunction in type 1 diabetes mellitus.


The International Journal of Neuropsychopharmacology | 2014

Protective effect of pranlukast on Aβ1–42-induced cognitive deficits associated with downregulation of cysteinyl leukotriene receptor 1

Miao-jin Ji; Lan Chen; Mei Hu; Yan Long; Yong-qi Li; Ming-Xing Miao; Jia-chang Li; Ning Li; Hui Ji; Xijing Chen; Hao Hong

Deposition of extracellular amyloid-β (Aβ) peptide is one of the pathological hallmarks of Alzheimers disease (AD). Accumulation of Aβ is thought to associate with cognition deficits, neuroinflammation and apoptosis observed in AD. However, effective neuroprotective approaches against Aβ neurotoxicity are unavailable. In the present study, we analysed the effects of pranlukast, a selective cysteinyl leukotriene receptor 1 (CysLT₁R) antagonist, on the impairment of learning and memory formation induced by Aβ and the probable underlying electrophysiological and molecular mechanisms. We found that bilateral intrahippocampal injection of Aβ₁₋₄₂ resulted in a significant decline of spatial learning and memory of mice in the Morris water maze (MWM) and Y-maze tests, together with a serious depression of in vivo hippocampal long-term potentiation (LTP) in the CA1 region of the mice. Importantly, this treatment caused significant increases in CysLT₁R expression and subsequent NF-κB signaling, caspase-3 activation and Bcl-2 downregulation in the hippocampus or prefrontal cortex. Oral administration of pranlukast at 0.4 or 0.8 mg/kg for 4 wk significantly reversed Aβ₁₋₄₂-induced impairments of cognitive function and hippocampal LTP in mice. Furthermore, pranlukast reversed Aβ₁₋₄₂-induced CysLT₁R upregulation, and markedly suppressed the Aβ₁₋₄₂-triggered NF-κB pathway, caspase-3 activation and Bcl-2 downregulation in the hippocampus and prefrontal cortex in mice. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) assay confirmed its presence in the brain after oral administration of pranlukast in mice. These data disclose novel findings about the therapeutic potential of pranlukast, revealing a previously unknown therapeutic possibility to treat memory deficits associated with AD.


Neurochemistry International | 2013

Leukotriene D4 induces amyloid-β generation via CysLT1R-mediated NF-κB pathways in primary neurons

Xiao Yun Wang; Su Su Tang; Mei Hu; Yan Long; Yong Qi Li; Ming Xing Liao; Hui Ji; Hao Hong

Although the pathogenesis of sporadic Alzheimers disease (AD) is not clearly understood, neuroinflammation has been known to play a role in the pathogenesis of AD. To investigate a functional link between the neuroinflammation and AD, the effect of leukotriene D4 (LTD4), an inflammatory lipid mediator, was studied on amyloid-β generation in vitro. Application of LTD4 to cell monolayers at concentrations up to 40 nM LTD4 caused increases in the Aβ releases. Concentrations ≥ 40 nM LTD4 decreased neuronal viability. Application of 20 nM LTD4 caused a significant increase in Aβ generation, as assessed by ELISA or Western blotting, without significant cytotoxicity. At this concentration, exposure of neurons to LTD4 for 24h produced maximal effect in the Aβ generation, and significant increases in the expressions of cysteinyl leukotriene 1 receptor (CysLT(1)R) and activity of β- or γ-secretase with complete abrogation by the selective CysLT(1)R antagonist pranlukast. Exposure of neurons to LTD4 for 1h showed activation of NF-κB pathway, by assessing the levels of p65 or phospho-p65 in the nucleus, and either CysLT(1)R antagonist pranlukast or NF-κB inhibitor PDTC prevented the nuclear translocation of p65 and the consequent phosphorylation. PDTC also inhibited LTD4-induced elevations of β- or γ-secretase activity and Aβ generation in vitro. Overall, our data show for the first time that LTD4 causes Aβ production by enhancement of β- or γ-secretase resulting from activation of CysLT(1)R-mediated NF-κB signaling pathway. These findings provide a novel pathologic link between neuroinflammation and AD.


Neurochemistry International | 2014

Montelukast rescues primary neurons against Aβ1-42-induced toxicity through inhibiting CysLT1R-mediated NF-κB signaling.

Jin’e Lai; Zhen Lin Mei; Hao Wang; Mei Hu; Yan Long; Ming Xing Miao; Ning Li; Hao Hong

Amyloid-β peptide (Aβ), which can invoke a cascade of inflammatory responses, is considered to play a causal role in the development and progress of Alzheimers disease (AD). Montelukast, known as a cysteinyl leukotriene receptor 1 (CysLT1R) antagonist, is currently used for treatment of inflammatory diseases such as asthma. We have previously reported that CysLT1R activation is involved in Aβ generation. In this study, we investigated rescuing effect of CysLT1R antagonist montelukast on Aβ1-42-induced neurotoxicity in primary neurons. Our data showed that Aβ1-42 elicited a marked increase of CysLT1R expression in primary mouse neurons. This increment of CysLT1R expression was accompanied by increases of inflammatory factors such as NF-κB p65, tumor necrosis factor-α (TNFα) and interleukin-1β (IL-1β) as well as pro-apoptotic protein Caspase-3 activation and anti-apoptosis protein Bcl-2 reduction. Aβ1-42-mediated increase of CysLT1R expression was associated with Aβ1-42-induced cytotoxicity as measured by MTT reduction assay and lactate dehydrogenase (LDH) release assay. This observation was confirmed with treatment of montelukast, a selective CysLT1R antagonist, which had significant effect on Aβ1-42-induced cytotoxicity. Moreover, blockade of CysLT1R with montelukast reversed Aβ1-42-mediated increase of CysLT1R expression, and concomitant changes of the pro-inflammatory factors and the apoptosis-related proteins. The results demonstrate that montelukast rescued neurons against Aβ1-42-induced neurotoxicity, neuroinflammation and apoptosis by down-regulating CysLT1R-mediated NF-κB signaling, suggesting that CysLT1R may be a potential target for AD, and its antagonist may have beneficial effects for treatment of AD.


Pharmacology, Biochemistry and Behavior | 2014

Pretreatment with antiasthmatic drug ibudilast ameliorates Aβ1–42-induced memory impairment and neurotoxicity in mice

Hao Wang; Zhen Lin Mei; Kai Long Zhong; Mei Hu; Yan Long; Ming Xing Miao; Ning Li; Tianhua Yan; Hao Hong

Amyloid-β peptide (Aβ) is thought to be associated with the progressive neuronal death observed in Alzheimers disease (AD). However, effective neuroprotective approaches against Aβ neurotoxicity are unavailable. Here, we investigated possible preventive effects of ibudilast, as a pharmacologic phosphodiesterase inhibitor, currently used for treatment of inflammatory diseases such as asthma, on Aβ 1-42-induced neuroinflammatory, apoptotic responses and memory impairment. We found that pretreatment with ibudilast (4 or 12 mg/kg, i.p.) significantly ameliorated impaired spatial learning and memory in intracerebroventricularly (ICV) Aβ 1-42-injected mice, as evidenced by decrease in escape latency during acquisition trials and increase in exploratory activities in the probe trial in Morris water maze (MWM) task, and by increase in the number of correct choices and decrease in latency to enter the shock-free compartment in Y-maze test. Further study showed that ibudilast prevented generation of pro-inflammatory cytokines such as NF-κB p65 and TNF-α as well as pro-apoptotic molecule caspase-3 activation and anti-apoptotic protein Bcl-2 downregulation in both hippocampus and cortex of ICV Aβ 1-42-injected mice. Taken together, our findings suggest that ibudilast has preventive effects on Aβ-induced cognitive impairment via inhibiting neuroinflammatory and apoptotic responses.


Brain Behavior and Immunity | 2017

Preventive effect of genetic knockdown and pharmacological blockade of CysLT1R on lipopolysaccharide (LPS)-induced memory deficit and neurotoxicity in vivo

Fang Chen; Arijit Ghosh; Feng Wu; Mei Hu; Hong-Bin Sun; Ling-Yi Kong; Hao Hong

Previously we reported that cysteinyl leukotrienes (Cys-LTs) and the type 1 receptor for Cys-LTs (CysLT1R) are related to amyloid β (Aβ)-induced neurotoxicity. The aim of the current study was to find out the role of CysLT1R on lipopolysaccharide (LPS)-induced cognitive deficit and neurotoxicity. shRNA-mediated knockdown or pharmacological blockade (by pranlukast) of CysLT1R were performed in ICR mice for 21days prior to systemic infusion of LPS. From day 22, LPS was administered for 7days and then a set of behavioral, histopathological and biochemical tests were employed to test memory, neuroinflammation and apoptotic responses in the mouse hippocampus. LPS (only)-treated mice showed poor performance in both Morris water maze (MWM) and Y-maze tests. However, shRNA-mediated knockdown or pranlukast-treated blockade of CysLT1R improved performance of the mice in these tests. To find out the possible underlying mechanisms, we assessed several parameters such as microglial activation (by immunohistochemistry), level of CysLT1R (by WB and qRT-PCR) and the inflammatory/apoptotic pathways (by ELISA or TUNEL or WB) in the mouse hippocampus. LPS-induced memory impairment was accompanied by activation of microglia, higher level of CysLT1R, IL-1β, TNF-α and nuclear NF-κB p65. LPS also caused apoptosis in the hippocampus as detected by TUNEL staining, further supplemented by detection of increased Caspase-3 and a reduced Bcl-2/Bax ratio. All of these adverse changes in the mouse hippocampus were inhibited by pretreatment with CysLT1R-shRNA and pranlukast. Through this study we suggest that CysLT1R shares a strong correlation with LPS-associated memory deficit, neuroinflammation and apoptosis and CysLT1R could be a novel target for preventive measures to intervene the progression of Alzheimers disease (AD)-like phenotypes.


British Journal of Pharmacology | 2016

PPARγ agonists regulate bidirectional transport of amyloid‐β across the blood–brain barrier and hippocampus plasticity in db/db mice

Hao Wang; Fang Chen; Kai Long Zhong; Su Su Tang; Mei Hu; Yan Long; Ming Xing Miao; Jian Min Liao; Hong Bing Sun; Hao Hong

There is emerging evidence suggesting that abnormal transport of amyloid‐β (Aβ) across the blood–brain barrier (BBB) is involved in diabetes‐associated cognitive decline. We investigated whether PPARγ agonists restore Aβ transport across the BBB and hippocampal plasticity in db/db mice.

Collaboration


Dive into the Mei Hu's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Feng Wu

Xi'an Jiaotong University

View shared research outputs
Top Co-Authors

Avatar

Li-Ping Liu

Anhui Medical University

View shared research outputs
Top Co-Authors

Avatar

Wei Hu

Anhui Medical University

View shared research outputs
Top Co-Authors

Avatar

Han-Ting Zhang

West Virginia University

View shared research outputs
Top Co-Authors

Avatar

Biao Wang

Xi'an Jiaotong University

View shared research outputs
Top Co-Authors

Avatar

Guan Tao Du

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Guang Jun Liu

Nanjing Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge