Network


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

Hotspot


Dive into the research topics where Li-Na Hou is active.

Publication


Featured researches published by Li-Na Hou.


BMC Pulmonary Medicine | 2014

mAChRs activation induces epithelial-mesenchymal transition on lung epithelial cells

Kai Yang; Yun Song; Ya-Bing Tang; Zu-Peng Xu; Wei Zhou; Li-Na Hou; Liang Zhu; Zhihua Yu; Hongzhuan Chen; Yong-Yao Cui

BackgroundEpithelial-mesenchymal transition (EMT) has been proposed as a mechanism in the progression of airway diseases and cancer. Here, we explored the role of acetylcholine (ACh) and the pathway involved in the process of EMT, as well as the effects of mAChRs antagonist.MethodsHuman lung epithelial cells were stimulated with carbachol, an analogue of ACh, and epithelial and mesenchymal marker proteins were evaluated using western blot and immunofluorescence analyses.ResultsDecreased E-cadherin expression and increased vimentin and α-SMA expression induced by TGF-β1 in alveolar epithelial cell (A549) were significantly abrogated by the non-selective mAChR antagonist atropine and enhanced by the acetylcholinesterase inhibitor physostigmine. An EMT event also occurred in response to physostigmine alone. Furthermore, ChAT express and ACh release by A549 cells were enhanced by TGF-β1. Interestingly, ACh analogue carbachol also induced EMT in A549 cells as well as in bronchial epithelial cells (16HBE) in a time- and concentration-dependent manner, the induction of carbachol was abrogated by selective antagonist of M1 (pirenzepine) and M3 (4-DAMP) mAChRs, but not by M2 (methoctramine) antagonist. Moreover, carbachol induced TGF-β1 production from A549 cells concomitantly with the EMT process. Carbachol-induced EMT occurred through phosphorylation of Smad2/3 and ERK, which was inhibited by pirenzepine and 4-DAMP.ConclusionsOur findings for the first time indicated that mAChR activation, perhaps via M1 and M3 mAChR, induced lung epithelial cells to undergo EMT and provided insights into novel therapeutic strategies for airway diseases in which lung remodeling occurs.


Toxicology and Applied Pharmacology | 2012

Protective effects of anisodamine on cigarette smoke extract-induced airway smooth muscle cell proliferation and tracheal contractility

Guang-Ni Xu; Kai Yang; Zu-Peng Xu; Liang Zhu; Li-Na Hou; Hong Qi; Hongzhuan Chen; Yong-Yao Cui

Anisodamine, an antagonist of muscarinic acetylcholine receptors (mAChRs), has been used therapeutically to improve smooth muscle function, including microvascular, intestinal and airway spasms. Our previous studies have revealed that airway hyper-reactivity could be prevented by anisodamine. However, whether anisodamine prevents smoking-induced airway smooth muscle (ASM) cell proliferation remained unclear. In this study, a primary culture of rat ASM cells was used to evaluate an ASM phenotype through the ability of the cells to proliferate and express contractile proteins in response to cigarette smoke extract (CSE) and intervention of anisodamine. Our results showed that CSE resulted in an increase in cyclin D1 expression concomitant with the G0/G1-to-S phase transition, and high expression of M2 and M3. Functional studies showed that tracheal hyper-contractility accompanied contractile marker α-SMA high-expression. These changes, which occur only after CSE stimulation, were prevented and reversed by anisodamine, and CSE-induced cyclin D1 expression was significantly inhibited by anisodamine and the specific inhibitor U0126, BAY11-7082 and LY294002. Thus, we concluded that the protective and reversal effects and mechanism of anisodamine on CSE-induced events might involve, at least partially, the ERK, Akt and NF-κB signaling pathways associated with cyclin D1 via mAChRs. Our study validated that anisodamine intervention on ASM cells may contribute to anti-remodeling properties other than bronchodilation.


Pharmacological Research | 2013

TNF-α-induced CXCL8 production by A549 cells: Involvement of the non-neuronal cholinergic system

Zu-Peng Xu; P. Devillier; Guang-Ni Xu; Hong Qi; Liang Zhu; Wei Zhou; Li-Na Hou; Ya-Bin Tang; Kai Yang; Zhihua Yu; Huimin Chen; Yun Cui

It was recently suggested that the non-neuronal cholinergic system has a regulatory role in pulmonary inflammation. We investigated this systems involvement in the control of cytokine production by the A549 human alveolar epithelial cell line. CXCL8 and acetylcholine (ACh) concentrations were measured using ELISA and LC-MS/MS, respectively. The mRNA expression of muscarinic receptor (MR) subtypes was determined using RT-PCR. In A549 cells, TNF-α increased the release of CXCL8 and ACh and the expression of the subtype 3 MR (M3R). Furthermore, TNF-α-induced CXCL8 secretion was (i) inhibited by the MR antagonist tiotropium and the M3R antagonist 4-DAMP and (ii) enhanced by the M1/M3R agonist pilocarpine and the cholinesterase inhibitor physostigmine. Taken as a whole, these results suggest that ACh release by A549 cells enhances TNF-α-induced CXCL8 secretion through activation of the M3R. Western blot analysis revealed that pilocarpine and physostigmine enhanced the TNF-α-induced phosphorylation of ERK1/2 and p38 MAPK and the degradation of IκBα. Inhibition of these pathways with specific inhibitors abrogated the pilocarpine-induced CXCL8 release. Our results suggest that the TNF-α-induced secretion of CXCL8 in A549 cells is regulated by the release of ACh, the latters binding to the M3R and the downstream activation of NF-κB and the ERK1/2 and p38 MAPK signaling pathways. Our findings suggest that MR antagonists may have anti-inflammatory effects by preventing pro-inflammatory events driven by endogenous, non-neuronal ACh.


Pharmacological Research | 2013

Up-regulation of KCa3.1 promotes human airway smooth muscle cell phenotypic modulation

Zhihua Yu; Yanxia Wang; Yun Song; Hao-Zhong Lu; Li-Na Hou; Yun Cui; Huimin Chen

Airway smooth muscle (ASM) cell phenotype modulation, characterized by reversible switching between contractile and proliferative phenotypes, is considered to contribute to proliferative diseases such as allergic asthma and chronic obstructive pulmonary disease (COPD). KCa3.1 has been suggested to be involved in regulating ASM cell activation, proliferation, and migration. However, little is known regarding the exact role of KCa3.1 in ASM cell phenotypic modulation. To elucidate the role of KCa3.1 in regulating ASM cell phenotypic modulation, we investigated the effects of KCa3.1 channels on ASM contractile marker protein expression, proliferation and migration of primary human bronchial smooth muscle (BSM) cells. We found that PDGF increased KCa3.1 channel expression in BSM cells with a concomitant marked decrease in the expression of contractile phenotypic marker proteins including smooth muscle myosin heavy chain (SMMHC), smooth muscle α-actin (α-SMA), myocardin and KCa1.1. These changes were significantly attenuated by the KCa3.1 blocker, TRAM-34, or gene silencing of KCa3.1. Pharmacological blockade or gene silencing of KCa3.1 also suppressed PDGF-induced human BSM cell migration and proliferation accompanied by a decrease in intracellular free Ca(2+) levels as a consequence of membrane depolarization, resulting in a reduction in cyclin D1 level and cell cycle arrest at G0-G1 phase. Additionally, PDGF-induced up-regulation of KCa3.1 and down-regulation of BSM contractile marker proteins were regulated by the ERK inhibitor U0126 and the AKT inhibitor LY294002. These findings highlight a novel role for the KCa3.1 channel in human BSM cell phenotypic modulation and provide a potential target for therapeutic intervention for proliferative airway diseases.


Journal of Controlled Release | 2016

Nanoformulated alpha-mangostin ameliorates Alzheimer's disease neuropathology by elevating LDLR expression and accelerating amyloid-beta clearance.

Lei Yao; Xiao Gu; Qingxiang Song; Xiao-Lin Wang; Meng Huang; Meng Hu; Li-Na Hou; Ting Kang; Jun Chen; Hongzhuan Chen; Xiaoling Gao

Alzheimers disease (AD), the most common form of dementia, is now representing one of the largest global healthcare challenges. However, an effective therapy is still lacking. Accumulation of amyloid-beta (Aβ) in the brain is supposed to trigger pathogenic cascades that eventually lead to AD. Therefore, Aβ clearance strategy is being actively pursued as a promising disease modifying therapy. Here, we found that α-mangostin (α-M), a polyphenolic xanthone derivative from mangosteen, up-regulated low density lipoprotein receptor (LDLR) expression in microglia and liver cells, and efficiently facilitated Aβ clearance. However, the in vivo application of α-M is limited due to its hydrophobic nature, poor aqueous solubility and stability, and thus low bioavailability and accumulation in the target organs. To overcome this limitation, α-M was encapsulated into the core of poly(ethylene glycol)-poly(l-lactide) (PEG-PLA) nanoparticles [NP(α-M)]. Such nanoencapsulation improved the biodistribution of α-M in both the brain and liver, enhanced the brain clearance of (125)I-radiolabeled Aβ1-42 in an LDLR-dependent manner, reduced Aβ deposition, attenuated neuroinflammatory responses, ameliorated neurologic changes and reversed behavioral deficits in AD model mice. These findings justified the concept that polyphenol-mediated modulation of LDLR expression might serve as a safe and efficient disease-modifying therapy for AD by accelerating Aβ clearance. It also demonstrated the powerful capacity of nanotechnology in modulating the biodistribution behavior of drug to improve its therapeutic efficacy in AD.


International Immunopharmacology | 2012

Role of M3 mAChR in in vivo and in vitro models of LPS-induced inflammatory response

Zu-Peng Xu; Kai Yang; Guang-Ni Xu; Liang Zhu; Li-Na Hou; Wen-Hui Zhang; Hongzhuan Chen; Yong-Yao Cui

OBJECTIVE We tested the potential role of the mAChR in lipopolysaccharide (LPS)-induced inflammatory response in in vivo and in vitro models and a possible signaling pathway involved in the inflammatory process. METHODS Anesthetized mice were challenged with intratracheal LPS to induce acute lung injury. The cytology and histopathology changes, expression of cytokines and pulmonary vascular permeability were used to evaluate the effects of the cholinergic agent. Alveolar macrophage cell line NR8383 was also used to confirm the role of mAChRs and the molecular mechanisms underlying the LPS-induced events. RESULTS LPS-induced acute lung injury (ALI) was significantly improved by atropine (a non-selective mAChR antagonist) and 4-DAMP (a M3 mAChR antagonist), as indicated by the diminution of neutrophil infiltration, pulmonary vascular permeability and IL-6 and TNF-α production. LPS-induced TNF-α production from the alveolar macrophage was significantly inhibited by atropine and 4-DAMP, but not pirenzepine (a M1 mAChR antagonist) and methoctramine (a M2 mAChR antagonist). Interestingly, LPS-induced TNF-α production was enhanced by the muscarinic receptor agonist pilocarpine, and treatment with pilocarpine alone was able to trigger TNF-α production from the alveolar macrophage, which was effectively attenuated by 4-DAMP. Western blot analysis showed that LPS-induced degradation of IκBα was strongly blocked by atropine/4-DAMP both in vivo and in vitro, indicating that M3 mAChR was involved in LPS-induced lung inflammation by mediating the NF-κB signaling pathway. CONCLUSION Our findings bring the evidence that the blockage of mAChR exerts anti-inflammatory properties, in which the M3 mAChR plays an important role in the LPS-induced lung inflammation.


International Immunopharmacology | 2011

Modulatory effect of anisodamine on airway hyper-reactivity and eosinophilic inflammation in a murine model of allergic asthma.

Zu-Peng Xu; Hao Wang; Li-Na Hou; Zheng Xia; Liang Zhu; Hongzhuan Chen; Yong-Yao Cui

Anisodamine, a peripheral muscarinic receptor antagonist, is a naturally occurring atropine derivative that has been isolated, synthesized and characterized by scientists in China. In the present investigation, we evaluated the modulatory effects of anisodamine on airway hyper-reactivity and inflammation in a murine model of allergic asthma. Asthma model was induced successfully by ovalbumin. The activation of cells, airway eosinopilia, cytokine production, and airway function were examined. Our results collectively show that anisomanine could significantly suppress the accumulation of eosinophils into the airways and dramatically inhibited the histological changes in OVA-induced mice. Additionally, anisodamine could restore the Th1/Th2 balance in BALF by downregulating the level of Th2 cell-associated cytokine IL-4 (p<0.01) and upregulating the level of Th1 cell-associated cytokine IFN-γ (p<0.01). In addition, pretreatment with anisodamine also showed strong suppression of allergen-induced bronchial hyper-reactivity with maximum contraction decreasing from 0.45 ± 0.02 g to 0.28 ± 0.03 g (p<0.01). These results suggested the modulatory effects of anisodamine on Th1/Th2 balance by enhancing Th1-related and suppressing Th2-related parameters, as well as its potential application in airway hyper-reactivity and eosinophilic inflammation.


CNS Neuroscience & Therapeutics | 2014

A New Motif in the N‐Terminal of Acetylcholinesterase Triggers Amyloid‐β Aggregation and Deposition

Li-Na Hou; Jianrong Xu; Qing-Nan Zhao; Xiao-Ling Gao; Yong-Yao Cui; Jun Xu; Hao Wang; Hongzhuan Chen

As a molecular chaperone, acetylcholinesterase (AChE; EC 3.1.1.7) plays a critical role in the pathogenesis of Alzheimers disease (AD). The peripheral anionic site (PAS) of AChE has been indicated as the amyloid‐β (Aβ) binding domain. The goal of this study was to determine other motifs in AChE involved in Aβ aggregation and deposition.


Frontiers in Pharmacology | 2017

The Potassium Channel KCa3.1 Represents a Valid Pharmacological Target for Astrogliosis-Induced Neuronal Impairment in a Mouse Model of Alzheimer’s Disease

Tianjiao Wei; Mengni Yi; Wen Gu; Li-Na Hou; Qin Lu; Zhihua Yu; Hongzhuan Chen

Alzheimer’s disease (AD) is a neurodegenerative disorder characterized by progressive decline of cognitive function. Astrogliosis plays a critical role in AD by instigating neuroinflammation, which leads ultimately to cognition decline. We previously showed that the intermediate-conductance Ca2+-activated potassium channel (KCa3.1) is involved in astrogliosis-induced by TGF-β in vitro. In the present study, we investigated the contribution of KCa3.1 channels to astrogliosis-mediated neuroinflammation, using TgAPP/PS1 mice as a model for AD. We found that KCa3.1 expression was increased in reactive astrocytes as well as in neurons in the brains of both TgAPP/PS1 mice and AD patients. Pharmacological blockade of KCa3.1 significantly reduced astrogliosis, microglial activation, neuronal loss, and memory deficits. KCa3.1 blockade inhibited astrocyte activation and reduced brain levels of IL-1β, TNF-α, iNOS, and COX-2. Furthermore, we used primary co-cultures of cortical neurons and astrocytes to demonstrate an important role for KCa3.1 in the process of astrogliosis-induced neuroinflammatory responses during amyloid-β (Aβ)-induced neuronal loss. KCa3.1 was found to be involved in the Aβ-induced activated biochemical profile of reactive astrocytes, which included activation of JNK MAPK and production of reactive oxygen species. Pharmacological blockade of KCa3.1 attenuated Aβ-induced reactive astrocytes and indirect, astrogliosis-mediated damage to neurons. Our data clearly indicate a role for astrogliosis in AD pathogenesis and suggest that KCa3.1 inhibition might represent a good therapeutic target for the treatment of AD. Highlights: (1) Blockade of KCa3.1 in APP/PS1 transgenic mice attenuated astrogliosis and neuron loss, and an attenuation of memory deficits. (2) Blockade of KCa3.1 attenuated Aβ-induced indirect, astrogliosis-mediated damage to neurons in vitro via activation of JNK and ROS.


Pharmacological Research | 2015

Carbocysteine restores steroid sensitivity by targeting histone deacetylase 2 in a thiol/GSH-dependent manner.

Yun Song; Hao-Zhong Lu; Jianrong Xu; Xiao-Lin Wang; Wei Zhou; Li-Na Hou; Liang Zhu; Zhihua Yu; Hongzhuan Chen; Yong-Yao Cui

Steroid insensitivity is commonly observed in patients with chronic obstructive pulmonary disease. Here, we report the effects and mechanisms of carbocysteine (S-CMC), a mucolytic agent, in cellular and animal models of oxidative stress-mediated steroid insensitivity. The following results were obtained: oxidative stress induced higher levels of interleukin-8 (IL-8) and tumor necrosis factor alpha (TNF-α), which are insensitive to dexamethasone (DEX). The failure of DEX was improved by the addition of S-CMC by increasing histone deacetylase 2 (HDAC2) expression/activity. S-CMC also counteracted the oxidative stress-induced increase in reactive oxygen species (ROS) levels and decreases in glutathione (GSH) levels and superoxide dismutase (SOD) activity. Moreover, oxidative stress-induced events were decreased by the thiol-reducing agent dithiothreitol (DTT), enhanced by the thiol-oxidizing agent diamide, and the ability of DEX was strengthened by DTT. In addition, the oxidative stress-induced decrease in HDAC2 activity was counteracted by S-CMC by increasing thiol/GSH levels, which exhibited a direct interaction with HDAC2. S-CMC treatment increased HDAC2 recruitment and suppressed H4 acetylation of the IL-8 promoter, and this effect was further ablated by addition of buthionine sulfoximine, a specific inhibitor of GSH synthesis. Our results indicate that S-CMC restored steroid sensitivity by increasing HDAC2 expression/activity in a thiol/GSH-dependent manner and suggest that S-CMC may be useful in a combination therapy with glucocorticoids for treatment of steroid-insensitive pulmonary diseases.

Collaboration


Dive into the Li-Na Hou's collaboration.

Top Co-Authors

Avatar

Hongzhuan Chen

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Liang Zhu

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Yong-Yao Cui

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Wei Zhou

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Zu-Peng Xu

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Jianrong Xu

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Kai Yang

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Yun Song

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Zhihua Yu

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Guang-Ni Xu

Shanghai Jiao Tong University

View shared research outputs
Researchain Logo
Decentralizing Knowledge