钠氢交换体1在内毒素性急性肺损伤中作用的实验研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
目的建立大鼠内毒素性急性肺损伤实验模型,观察实验模型中大鼠肺组织上钠氢交换体l的表达变化情况及其与炎症的关系。
     方法40只雄性清洁级SD大鼠,体重在250-350g之间,随机分成空白对照组(C组)、脂多糖2h组(L-2h组)、脂多糖4h组(L-4h组)和脂多糖6h组(L-6h组),每组各10只。C组大鼠给予股静脉输注生理盐水;L-2h组、L-4h组和L-6h组大鼠均给予股静脉输注6mg/kg脂多糖。实验开始后2h颈动脉放血处死C组和L-2h组大鼠,4小时和6小时后分别处死L-4h组和L-6h组大鼠。监测各组大鼠基本生命体征;光学显微镜下观察大鼠肺组织病理改变;计算急性肺损伤评分和肺组织湿/干重比值;检测支气管肺泡灌洗液中总蛋白、肿瘤坏死因子-α和巨噬细胞炎性蛋白的浓度;测定肺组织髓过氧化物酶活性;EMSA检测肺组织细胞核转录因子-κB的活性;免疫组化、RT-PCR和Western Blot检测肺组织钠氢交换体1 mRNA和蛋白的表达水平。
     结果(1)和空白对照组比较,脂多糖各组大鼠平均动脉压明显下降(均P<0.05)、心率和呼吸频率明显上升(均P<0.05);脂多糖4h组和6h组大鼠动脉血氧分压显著下降(均P<0.05),(2)空白对照组大鼠肺组织肺泡结构正常,肺泡腔无渗出物;脂多糖各组大鼠肺组织病理改变明显,肺泡间隔增厚,肺泡腔内可见较多的炎性细胞,肺泡腔内有血性渗出液。和空白对照组比较,脂多糖各组大鼠肺组织急性肺损伤评分均明显增高(P<0.05)。(3)和空白对照组比较,脂多糖各组大鼠肺组织湿/干重比均明显上升(P<0.05);支气管肺泡灌洗液中总蛋白、肿瘤坏死因子-α和巨噬细胞炎性蛋白的浓度均明显增高(均P<0.05)。(4)和空白对照组比较,脂多糖各组大鼠肺组织髓过氧化物酶活性均明显增高(P<0.05)。(5)和空白对照组比较,脂多糖各组大鼠肺组织核转录因子-κB的活性均明显增高(P<0.05)。(6)和空白对照组比较,免疫组化检测脂多糖各组大鼠肺组织NHE1的表达水平均明显增高(P<0.05)。(7)和空白对照组比较,脂多糖各组大鼠肺组织钠氢交换体1mRNA的表达水平均明显增高(P<0.05)。(8)和空白对照组比较,脂多糖各组大鼠肺组织钠氢交换体1的蛋白表达水平均明显增高(P<0.05)。
     结论以6mg/kg的剂量给予大鼠静脉注射脂多糖后4h就产生了明显的急性肺损伤,表现为大鼠基本生命体征的内毒素休克症状出现;大鼠大体标本组织学炎症病理改变;肺组织含水量增加;肺组织及支气管肺泡灌洗液中相关炎性因子增加;与此同时,脂多糖各组大鼠肺组织钠氢交换体1mRNA和蛋白的表达水平也随着急性肺损伤炎症反应的加重而明显增加。
     目的探讨阿米洛利预先给药对脂多糖诱导的大鼠内毒素性急性肺损伤的保护作用及可能机制。
     方法40只雄性清洁级SD大鼠,体重在250-350g之间,随机分成空白对照组(C组)、脂多糖组(L组)、阿米洛利组(A组)和阿米洛利预给药组(AL组)。C组静脉输注生理盐水3ml;L组依次静脉输注1ml生理盐水及2ml脂多糖;A组依次静脉输注1ml阿米洛利及2ml生理盐水;AL组先输注1ml阿米洛利再输注2ml脂多糖。静脉注射脂多糖/阿米洛利6小时后颈动脉放血处死大鼠。光学显微镜下观察大鼠肺组织病理改变;计算急性肺损伤评分和肺组织湿/干重比值;测定支气管肺泡灌洗液中总蛋白的浓度;检测肺组织髓过氧化物酶活性、丙二醛浓度和超氧化物歧化酶活力;半定量免疫组化法检测肺组织过氧亚硝酸盐硝基酪氨酸残基的含量;RT-PCR检测肺组织钠氢交换体1mRNA的表达水平;Western blot法检测钠氢交换体1、细胞外信号调节激酶及p38丝裂原活化蛋白激酶的蛋白表达水平。
     结果(1)空白对照组和阿米洛利组大鼠肺组织肺泡结构正常,肺泡腔无渗出物;脂多糖组和阿米洛利预给药组大鼠肺组织病理改变明显加重,肺泡间隔增厚,肺泡腔内可见较多炎性细胞渗出。和空白对照组比较,脂多糖组和阿米洛利预给药组大鼠肺组织ALI评分明显上升(均P<0.05);与脂多糖组相比,阿米洛利预给药组大鼠肺组织ALI评分显著下降(P<0.05)。(2)与空白对照组相比,脂多糖组和阿米洛利预给药组大鼠肺组织湿/干重比值明显上升(均P<0.05);与脂多糖组相比,阿米洛利预给药组大鼠肺组织湿/干重比值显著下降(P<0.05)。(3)与空白对照组相比,脂多糖组和阿米洛利预给药组大鼠BALF中总蛋白浓度明显上升(均P<0.05);与脂多糖组相比,阿米洛利预给药组大鼠BALF中总蛋白浓度显著下降(P<0.05)。(4)与空白对照组相比,脂多糖组和阿米洛利预给药组大鼠肺组织MPO活性明显上升(均P<0.05);与脂多糖组相比,阿米洛利预给药组大鼠肺组织MPO活性显著下降(P<0.05)。(5)与空白对照组相比,脂多糖组和阿米洛利预给药组大鼠肺组织MDA浓度明显上升(均P<0.05);与脂多糖组相比,阿米洛利预给药组大鼠肺组织MDA浓度显著下降(P<0.05)。(6)与空白对照组相比,脂多糖组和阿米洛利预给药组大鼠肺组织湿/干重比明显上升(均P<0.05);与脂多糖组相比,阿米洛利预给药组大鼠肺组织湿/干重比显著下降(P<0.05)。(7)与空白对照组相比,脂多糖组和阿米洛利预给药组大鼠肺组织硝基酪氨酸的含量明显上升(均P<0.05);与脂多糖组相比,阿米洛利预给药组大鼠肺组织硝基酪氨酸的含量显著下降(P<0.05)。(8)与空白对照组相比,脂多糖组和阿米洛利预给药组大鼠肺组织NHE1 mRNA和蛋白的表达水平明显上升(均P<0.05);与脂多糖组相比,阿米洛利预给药组大鼠肺组织NHE1 mRNA和蛋白的表达水平显著下降(P<0.05)。(9)与空白对照组相比脂多糖组和阿米洛利预给药组大鼠肺组织ERK的蛋白表达水平明显上升(均P<0.05);与脂多糖组相比,阿米洛利预给药组大鼠肺组织ERK的蛋白表达水平显著下降(P<0.05)。(10)与空白对照组相比,脂多糖组和阿米洛利预给药组大鼠肺组织p38 MAPK的蛋白表达水平明显上升(均P<0.05);与脂多糖组相比,阿米洛利预给药组大鼠肺组织p38 MAPK的蛋白表达水平无显著差异(P>0.05)。
     结论大鼠静脉注射脂多糖产生急性肺损伤的同时伴随有肺组织钠氢交换体1、细胞外信号调节激酶及p38丝裂原活化蛋白激酶的显著激活;钠氢交换体1抑制剂阿米洛利预先给药对大鼠内毒素性急性肺损伤有保护作用,其机制可能与抑制细胞外信号调节激酶的激活有关。
     目的探讨丝裂原活化蛋白激酶家族在阿米洛利预先给药防治脂多糖诱导的大鼠内毒素性急性肺损伤中的作用及可能机制。
     方法70只雄性清洁级SD大鼠,体重在250-350g之间,随机均分成空白对照组(C组)、脂多糖组(L组)、阿米洛利预给药组(AL组)、PD098059预给药组(PL组)、SB203580预给药组(sL组)、阿米洛利联合PD098059预给药组(APL组)和阿米洛利联合SB203580预给药组(ASL组)。C组静脉输注生理盐水3ml;L组静脉输注脂多糖(6mg/kg)及生理盐水总容积共3ml; AL组先静脉输注阿米洛利(10mg/kg)再输注脂多糖(6mg/kg)及生理盐水总容积共3ml; PL组先静脉输注PD098059(0.3mg/kg)再输注脂多糖(6mg/kg)及生理盐水总容积共3ml; SL组先静脉输注SB203580 (10mg/kg)再输注脂多糖(6mg/kg)及生理盐水总容积共3ml; APL组先静脉输注阿米洛利(10mg/kg)再输注PD098059(0.3mg/kg)及脂多糖(6mg/kg),总容积共3ml; ASL组先静脉输注阿米洛利(10mg/kg)再输注SB203580(10mg/kg)及脂多糖(6mg/kg),总容积共3ml。实验开始6小时后颈动脉放血处死大鼠。光学显微镜下观察大鼠肺组织病理改变;计算急性肺损伤评分和肺组织湿/干重比值;测定支气管肺泡灌洗液中总蛋白、肿瘤坏死因子-α和巨噬细胞炎性蛋白的浓度;检测肺组织髓过氧化物酶活性;RT-PCR检测肺组织钠氢交换体1 mRNA的表达水平;Western blot法检测钠氢交换体1的蛋白表达水平。
     结果(1)空白对照组大鼠肺组织肺泡结构正常,肺泡腔无渗出物;其余各组各组大鼠肺组织病理改变明显,肺泡间隔增厚,肺泡腔内可见炎性细胞渗出。(2)与空白对照组相比,其余各组大鼠肺组织ALI评分和肺组织湿/干重比值;BALF中总蛋白、肿瘤坏死因子-α、巨噬细胞炎性蛋白浓度;肺组织髓过氧化物酶活性及钠氢交换体1 mRNA和蛋白的表达水平均明显上升(均P<0.05),其中以脂多糖组上升最显著(均P<0.01);与脂多糖组相比,阿米洛利预给药组上述各指标显著下降(均P<0.05);与阿米洛利预给药组相比,PD098059预给药组、阿米洛利联合PD098059预给药组及SB203580预给药组上述各指标均明显下降(均P<0.05);而PD098059预给药组、阿米洛利联合PD098059预给药组及SB203580预给药组上述各指标之间无明显差别;与这三组相比,阿米洛利联合SB203580预给药组上述各指标明显下降(均P<0.05)。
     结论钠氢交换体1抑制剂阿米洛利预先给药对大鼠内毒素性急性肺损伤有保护作用;细胞外信号调节激酶的阻滞剂可以完全取消阿米洛利的作用,说明阿米洛利防治大鼠内毒素性急性肺损伤的作用机制可能与抑制细胞外信号调节激酶的激活有关。
     目的观察钠氢交换体1 mRNA和蛋白在脂多糖活化鼠源性巨噬细胞中的表达变化及阿米洛利预先给药对它的影响。
     方法传代培养的鼠源性巨噬细胞株(RAW264.7细胞)以1×106个/ml的密度接种于6孔板,培养48 h后,随机分为4组:仅加RPMI 1640培养液的对照组;加300μM阿米洛利的阿米洛利组;加1μg/ml脂多糖的脂多糖组:加300μM阿米洛利干预30min后再加1μg/ml脂多糖的阿米洛利预给药组;实验开始24 h后,ELISA法和Griess法分别检测各组细胞培养上清液中肿瘤坏死因子-α浓度和一氧化氮的水平、应用细胞内活性氧敏感性荧光探针DCFH-DA检测各组细胞内活性氧的水平;实验开始3h后,采用逆转录-聚合酶链反应(RT-PCR)检测各组细胞内钠氢交换体1 mRNA的表达水平;实验开始6h后,应用免疫印迹(Western blotting)技术检测各组细胞内钠氢交换体1蛋白的表达水平。
     结果与空白对照组相比,阿米洛利组细胞培养上清液中肿瘤坏死因子-α的浓度和一氧化氮的水平、细胞内活性氧的水平及钠氢交换体1 mRNA和蛋白的表达水平均无显著性差异(均P>0.05);而脂多糖组和阿米洛利预给药组内上述指标均明显上升(均P<0.05);与脂多糖组相比,阿米洛利预给药组内上述指标均显著下降(均P<0.05)。
     结论脂多糖活化鼠源性巨噬细胞中钠氢交换体1被迅速激活、表达增加;钠氢交换体1抑制剂阿米洛利预先给药干预可以明显抑制这种变化。
PartⅠExpression of Na+/H+ exchanger 1 in endotoxin-induced acute lung injury in rats
     Objective To build the model of the acute lung injury(ALI) induced by lipopolysaccharide(LPS) in rats in vivo and to observe the changes of Na+/H+ exchanger 1(NHE1) mRNA and protein expressions in the injurious lungs.
     Methods Forty Specific pathogen-free male Sprague-Dawley rats weighing 250-350 g were randomly divided into the following experimental groups (10 rats in each group):control group (C group):rats received iv. injection of normal saline and sacrificed by carotid artery bleeding 2 h after the administration of saline; LPS-2 h group(L-2 h group):rats received iv. injection of 6 mg/kg LPS and sacrificed by carotid artery bleeding 2 h after the administration of LPS; LPS-4 h group(L-4 h group); rats received iv. injection of 6 mg/kg LPS and sacrificed by carotid artery bleeding 4 h after the administration of LPS; LPS-6 h group(L-6 h group):rats received iv. injection of 6 mg/kg LPS and sacrificed by carotid artery bleeding 6 h after the administration of LPS. The samples of pulmonary tissue and lung lavage fluid were collected after experiments. The basic vital signs of rats were monitored and recorded. The pathological changes in lung tissue were examined with optical microscopy. The acute lung injury score and lung tissue wet / dry weight ratio were calculated. The total protein, tumor necrosis factor-a and macrophage inflammatory protein concentration in bronchoalveolar lavage fluid were detected. The myeloperoxidase(MPO) activity of lung tissue were measured. The nuclear factor-KB (NF-κB) activity of lung tissue were assayed with electrophoretic mobility shift assay (EMSA). The NHE1 mRNA and protein expressions in lung tissue were detected by immunohistochemistry、reverse transcriptase-polymerase chain reaction (RT-PCR) and Western Blot.
     Results (1) Compared with the control group, the Mean Arterial Pressure(MAP) of rats in LPS-2 h group、LPS-4 h group and LPS-6 h group were decreased significantly (all P<0.05)% Heart Rate (HR) and Respiratory Frequency(RF) of rats in LPS-2 h group、LPS-4 h group and LPS-6 h group were significantly elevated(all P<0.05); Arterial partial pressure of oxygen(PaO2) of rats in LPS-4 h group and LPS-6 h group were decreased significantly(all P <0.05). (2) The lung tissue in control group rats showed normal alveolar septum and alveolar spaces without exudation; the lung tissue of rats in LPS-2h、LPS-4 h group and LPS-6 h group showed high levels of intra-alveolar exudates, hyaline membrane formation, inflammatory cell infiltration, intra-alveolar hemorrhage, and interstitial edema. Compared with the control group, the lung tissue ALI scores of rats in LPS-2 h、LPS-4 h group and LPS-6 h group were significantly elevated(all P<0.05), the overall differences between groups were statistically significant (P<0.05). (3) Compared with the control group, the lung tissue wet/ dry weight ratio of rats in LPS-2 h group、LPS-4 h group and LPS-6h group were significantly elevated(all P<0.05), the overall differences between groups were statistically significant (P<0.05); the total protein(TP), tumor necrosis factor-α(TNF-α) and macrophage inflammatory protein(MIP-2) concentration in bronchoalveolar lavage fluid of rats in LPS-2 h group、LPS-4 h group and LPS-6 h group were significantly elevated(all P<0.05), the overall differences between groups were statistically significant (all P<0.05). (4) Compared with the control group, the MPO activity of lung tissue in LPS-2 h group、LPS-4 h group and LPS-6 h group were significantly elevated(all P<0.05), the overall differences between groups were statistically significant(P<0.05). (5) Compared with the control group, the NF-κB activity of lung tissue in LPS-2 h group、LPS-4 h group and LPS-6 h group were significantly elevated(all P<0.05); the overall differences between groups were statistically significant(P<0.05). (6) Compared with the control group, the NHE1 protein immunohistochemistry expressions of lung tissue in LPS-2 h group、LPS-4 h group and LPS-6 h group were significantly elevated(all P<0.05); the overall differences between groups were statistically significant(P<0.05). (7) Compared with the control group, the NHE1 mRNA expressions of lung tissue in LPS-2 h group、LPS-4 h group and LPS-6 h group were significantly elevated(all P<0.05); the overall differences between groups were statistically significant(P<0.05). (8) Compared with the control group, the NHE1 protein expressions of lung tissue in LPS-2 h group、LPS-4 h group and LPS-6 h group were significantly elevated (all P<0.05); the overall differences between groups were statistically significant(P<0.05).
     Conclusion Rats produced significant acute lung injury after intravenous injection of LPS 4 h with 6mg/kg, which includes endotoxin shock symptoms、histological inflammation in rats gross specimen pathology、lung tissue water content increased、inflammatory cytokines in lung tissue and bronchoalveolar lavage fluid increased; at the same time, the NHE1 mRNA and protein expressions of lung tissue in each LPS group were significantly elevated and remarkably increased with the duration of acute lung inflammatory response.
     Objective To investigate the effects of amiloride pretreatment on the acute lung injury(ALI) induced by lipopolysaccharide(LPS) in rats and the underlying mechanism.
     Methods Forty Specific pathogen-free male Sprague-Dawley rats weighing 250-350g were randomly divided into the following experimental groups (10 rats in each group):control group (C group):rats received iv. injection of normal saline; amiloride group(A group):rats received iv. injection of 10 mg/kg amiloride; LPS group(L group):rats received iv. injection of 6mg/kg LPS; amiloride pretreatment group(AL group):rats received iv. injection of 10 mg/kg amiloride 30 min before the administration of LPS. Degree of ALI were assessed by wet/dry weight ratio(W/D) and lung histological examination. Concentrations of total protien in bronchoalveolar lavage fluid(BALF) were determined. The activity of myeloperoxidase(MPO)、the concentrations of malondialdehyde(MDA) and the Vitality of Superoxide dismutase(SOD) in lung tissue were also measured. The contents of nitrotyrosine(NT) in lung tissue were determined by semi-quantitative immunohistochemistry. The Na+/H+ exchanger 1 (NHE1) mRNA expressions in lung tissue were detected by reverse transcriptase-polymerase chain reaction (RT-PCR). The expression of NHE1、extracellular signal regulated kinase (ERK) and mitogen-activated protein kinase(MAPK) p38 in lung tissue were also determined by Western blot analysis.
     Results Lung tissue ALI scores、W/D ratio、MPO activity、MDA concentrations、SOD vitality and NT contents, concentrations of total protienin BALF, the expression of NHE1, MAPK p38 and ERK increased significantly in LPS group as compared with control group(all P<0.05). These increases were significantly reduced in amiloride pretreatment group(all P<0.05) except MAPK p38 expression.
     Conclusion NHE1 inhibitor amiloride pretreatment can attenuate ALI induced by LPS in rats. This effect may be related to inhibition of ERK activation.
     Part III
     Roles of mitogen-activated protein kinases in the protection effects of amiloride on endotoxin-induced acute lung injury in rats
     Objective To investigate the roles of mitogen-activated protein kinases(MAPKs) in the protection effects of amiloride pretreatment on the acute lung injury(ALI) induced by lipopolysaccharide(LPS) in rats.
     Methods Seventy specific pathogen-free male Sprague-Dawley rats weighing 250-350g were randomly divided into the following experimental groups (10 rats in each group):control group (C group):rats received iv. injection of normal saline; LPS group(L group):rats received iv. injection of 6mg/kg LPS; amiloride pretreatment group(AL group):rats received iv. injection of 10 mg/kg amiloride 30 mins before the administration of LPS; PD098059 pretreatment group(PL group):rats received iv. injection of 0.3mg/kg PD098059 30 mins before the administration of LPS; SB203580 pretreatment group(SL group):rats received iv. injection of 10mg/kg SB203580 30 mins before the administration of LPS; amiloride combined PD098059 pretreatment group(APL group):rats received iv. injection of 10 mg/kg amiloride and 0.3mg/kg PD098059 30 mins before the administration of LPS; amiloride combined SB203580 pretreatment group(ASL group):rats received iv. injection of 10 mg/kg amiloride and 10mg/kg SB203580 30 mins before the administration of LPS. All rats were sacrificed by carotid artery bleeding 6h after the experiment starting. The samples of pulmonary tissue and lung lavage fluid were collected after experiments. The pathological changes in lung tissue were examined with optical microscopy. The acute lung injury scores and lung tissue wet/dry weight ratio were calculated. The total protein, tumor necrosis factor-a and macrophage inflammatory protein concentration in bronchoalveolar lavage fluid were detected. The myeloperoxidase(MPO) activity of lung tissue were also measured; the NHE1 mRNA and protein expressions in lung tissue were detected by reverse transcriptase-polymerase chain reaction (RT-PCR) and Western Blot analysis.
     Results (1) The lung tissues in control group rats showed normal alveolar septum and alveolar spaces without exudation; the lung tissues of rats in the other groups showed high levels of intra-alveolar exudates, hyaline membrane formation, inflammatory cell infiltration, intra-alveolar hemorrhage, and interstitial edema. (2) Compared with the control group, concentrations of total protien、tumor necrosis factor-αand macrophage inflammatory protein in bronchoalveolar lavage fluid, lung tissue ALI scores、W/D ratio、MPO activity and NHE1 mRNA and protein expressions in the other groups were significantly increased (all P<0.05), the indicators above-mentioned in LPS group were the most significantly increased (all P<0.05); compared with the LPS group, the indicators above-mentioned in amiloride pretreatment group were decreased significantly (all P<0.05); compared with the amiloride pretreatment group, the indicators above-mentioned in PD098059 pretreatment group、SB203580 pretreatment group and amiloride combined PD098059 pretreatment group were decreased significantly (all P<0.05) however; compared with these three groups, the indicators above-mentioned in amiloride combined SB203580 pretreatment group were significantly decreased (all P<0.05) yet.
     Conclusion NHE1 inhibitor amiloride pretreatment can attenuate ALI induced by LPS in rats; extracellular signal-regulated kinase inhibitor can completely abolish this kind of protective effect of amiloride, which illustrated that this effect may be related to inhibition of extracellular signal-regulated kinase activation.
     Part IV Expressions of Na+/H+ exchanger 1 in lipopolysaccharide-activated mouse macrophage-like cells and the pretreatment effect of amiloride
     Objective To observe the changes of Na+/H+ exchanger 1(NHE1) mRNA and protein expressions in the lipopolysaccharide(LPS)-activated mouse macrophage-like cells and the pretreatment effect of amiloride.
     Methods The mouse macrophage-like cell line (RAW 264.7) cells were seeded in 6 well plate with the density of 1×106/ml and cultured in RPMI 1640 medium at 37℃in 5% CO2 incubator for 48 h in vitro, then all cells were randomly divided into 4 groups(n=12):group C:control; amiloride group:RAW 264.7 cells were co-incubated with amiloride 300μM (final concentration); LPS group:RAW 264.7 cells were co-incubated with LPS 1μg/ml (final concentration); amiloride pretreatment group:RAW 264.7 cells were co-incubated with amiloride 300μM (final concentration) for 30 mins before co-incubated with LPS 1μg/ml (final concentration). The concentrations of tumor necrosis factor-α(TNF-α) and the levels of nitric oxide(NO) in cell culture supernatants in each group were detected with enzyme linked immunosorbent assay(ELISA) and the Griess method 24 h after the start of the trial. The levels of intracellular reactive oxygen species(ROS) in each group were detected with the ROS sensitivity DCFH-DA fluorescent probe method 24 h after the start of the trial. The expressions of NHE1 mRNA in each group were measured by reverse transcription-polymerase chain reaction (RT-PCR) 3 h after the start of the trial. The levels of NHE1 protein expressions in each group were also detected with Western blot analysis 6 h after the start of the trial.
     Results Compared with the control group, the concentrations of TNF-a and the levels of NO in cell culture supernatants、the levels of intracellular ROS、the expressions of NHE1 mRNA and the levels of NHE1 protein expressions in amiloride group have no statistical difference(all P>0.05); but the indicators above-mentioned in LPS group and amiloride pretreatment group were significantly increased(all P<0.05). Compared with the LPS group, the indicators above-mentioned in amiloride pretreatment group were significantly decreased (all P<0.05).
     Conclusion NHE1 can be rapidly activated and the expressions of NHE1 were increased in the LPS-activated mouse macrophage-like cells; pretreatment intervention with NHE 1 inhibitor amiloride can restrain the changes.
引文
1. Bernard GR, Artigas A, Brigham KL, et al. Report of the American-European Consensus conference on acute respiratory distress syndrome:definitions, mechanisms, relevant outcomes, and clinical trial coordination:consensus Committee. J Crit Care.1994,9:72-81.
    2. Glassberg AE, Luce JM, Matthay MA, et al.. Reasons for nonenrollment in a clinical trial of acute lung injury. Chest,2008,134:719-23.
    3. Bernard GR, Artigas A, Brigham KL, et al. The American-European Consensus Conference on ARDS:definitions, mechanisms, relevant outcomes, and clinical trial coordination. Am J Respir Crit Care Med,1994,149:818-24.
    4. Ware LB, Matthay MA. The acute respiratory distress syndrome. N Engl J Med,2000,342: 1334-1349.
    5. Bhatia M, Moochhala S. Role of inflammatory mediators in the pathophysiology of acute respiratory distress syndrome. J Pathol 2004,202:145-156.
    6. Abraham E. Neutrophils and acute lung injury. Crit Care Med 2003,31:S195-199.
    7. Fan J, Ye RD, Malik AB. Transcriptional mechanisms in acute lung injury. Am J Physiol Lung Cell Mol Physiol,2001,281:L1037-L50.
    8. Christman JW, Sadikot RT, Blackwell T. The role of nuclear factor-κB in pulmonary diseases. Chest,2000,117:1482-1487.
    9. Bachofen M, Weibel ER. Alterations of the gas exchange apparatus in adult respiratory insufficiency associated with septicemia. Am Rev Respir Dis,1977,116:589-615.
    10. Bachofen M, Weibel ER. Structural alterations of lung parenchyma in the adult respiratory distress syndrome. Clin Chest Med,1982,3:35-56.
    11.Matthay MA, Eschenbacher WC, Goetzl EJ. Elevated concentrations of leukotriene D4 in pulmonary edema fluid of patients with adult respiratory distress syndrome. J Clin Immunol, 1984,4:479-483.
    12.Parsons PE, Fowler AA, Hyers T, et al. Chemotactic activity in bronchoalveolar lavage fluid from patients with the adult respiratory distress syndrome. Am Rev Respir Dis,1985,132: 490-493.
    13.Steinberg KP, Milberg JA, Martin TR, et al. Evolution of bronchoalveolar cell populations in the adult respiratory distress syndrome. Am J Respir Crit Care Med,1994,150:113-122.
    14. Warshawski FJ, Sibbald W, Driedger A,et al. Abnormal neutrophil-pulmonary interaction in the adult respiratory distress syndrome:qualitative and quantitative assessment of pulmonary-neutrophil kinetics in humans with in vivo indium-111 neutrophil scintigraphy. Am Rev Respir Dis,1986,133:792-804.
    15.Matthay MA. Conference summary:acute lung injury. Chest,1999,116:119S-126S.
    16.Prescott SM, McIntyre TM, Zimmerman G. Two of the usual suspects, platelet-activating factor and its receptor, implicated in acute lung injury. J Clin Invest,1999,104:1019-1020.
    17.Moraes TJ, Zurawska JH, Downey GP. Neutrophil granule contents in the pathogenesis of lung injury. Curr Opin Hematol,2006,13:21-27.
    18.Tsushima K, King L S, Aggarwal NR, et al. Acute Lung Injury Review. Internal Medicine, 2009,48:621-630.
    19.Suzuki T, Moraes T J, Vachon E, et al. Proteinase-activated receptor-1 mediates elastase-induced apoptosis of human lung epithelial cells. Am J Respir Cell Mol Biol, 2005,33:231-247.
    20.Gando S, Kameue T, Nanzaki S, et al. Increased neutrophil elastase, persistent intravascular coagulation, and decreased fibrinolytic activity in patients with posttraumatic acute respiratory distress syndrome. J Trauma,1997,42:1068-1072.
    21.Pugin J, Verghese G, Widmer M C, et al. The alveolar space is the site of intense inflammatory and profibrotic reactions in the early phase of ARDS. Crit Care Med,1999,27: 304-312.
    22.Jin SW, Zhang L, Lian QQ, et al. Posttreatment with Aspirin-Triggered Lipoxin A4 Analog Attenuates Lipopolysaccharide-Induced Acute Lung Injury in Mice:The Role of Heme Oxygenase-1. Anesthesia & Analgesia,2007,104:369-377.
    23.Geerts L, Jorens PG, Willems J, et al. Natural inhibitors of neutrophil function in acute respiratory distress syndrome. Crit Care Med,2001,29:1920-1924.
    24.Suter PM, Suter S, Girardin E, et al. High bronchoalveolar levels of tumor necrosis factor and its inhibitors, interleukin-1, interferon, and elastase in patients with adult respiratory distress syndrome after trauma, shock, or sepsis. Am Rev Respir Dis,1992,145:1016-1022.
    25.Sookhai S, Wang JJ, McCourt M, et al. A novel therapeutic strategy for attenuating neutrophil-mediated lung injury in vivo. Ann Surg,2002,235:285-291.
    26.Goodman R, Pugin J, Lee JS, et al. Cytokine mediated inflammation in acute lung injury. Cytokine Growth Factor Rev,2003,14:523-535.
    27.Jhumka Z, Pervaiz S, Clement MV. Resveratrol regulates the expression of NHE-1 by repressing its promoter activity:critical involvement of intracellular H2O2 and caspases 3 and 6 in the absence of cell death. Int J Biochem Cell Biol,2009,41:945-56.
    28.Lum H, Roebuck KA. Oxidant stress and endothelial cell dysfunction. Am J Physiol Cell Physiol,2001,280:C719-C741.
    29.Waters C M, Savla U, Panos R J. KGF prevents hydrogen peroxide-induced increases in airway epithelial cell permeability. Am J Physiol,1997,272:L681-L9.
    30.Chabot F, Mitchell JA, Gutteridge JMC, et al. Reactive oxygen species in acute lung injury. Eur Respir J,1998,11:745-757.
    31.Slepkov E R,Rainey J K,Sykes B D, et al. Structural and functional analysis of the Na+/H+exchanger. Biochem J,2007,401(Pt3):623-633.
    32.Orlowski J, Grinstein S. Diversity of the mammalian sodium/proton exchanger SLC9 gene family. Pflugers Arch,2004,447:549-565.
    33.Meima M E, Mackley J R,Barter D L. Beyond ion translocation:structural functions of the sodium-hydrogen exchanger isoform-1. Curr Opin Nephrol Hypertens,2007,16:365-72.
    34.Orlowski J, Kandasamy RA, Shull GE. Molecular cloning of putative members of the Na/H exchanger gene family. cDNA cloning, deduced amino acid sequence, and mRNA tissue expression of the rat Na/H exchanger NHE-1 and two structurally related proteins. J Biol Chem,1992,267:9331-9339.
    35.Akram S, Teong HF, Fliegel L, et al. Reactive oxygen species-mediated regulation of the Na+-H+ exchanger 1 gene expression connects intracellular redox status with cells' sensitivity to death triggers. Cell Death Differ,2006,13:628-641.
    36.Orlinska U, Newton RC. Modification of tumor necrosis factor-alpha (TNF-alpha) production by the Na(+)-dependent HCO3- cotransport in lipopolysaccharide-activated human monocytes. Immunopharmacology,1995,30:41-50.
    37.Kamachi F, Ban HS, Hirasawa N, et al. Inhibition of lipopolysaccharide-induced prostaglandin E2 production and inflammation by the Na+/H+ exchanger inhibitors.J Pharmacol Exp Ther,2007,321:345-352.
    38.Kaba NK, Schultz J, Law FY, et al. Inhibition of Na+/H+ exchanger enhances low pH-induced L-selectin shedding and β2-integrin surface expression in human neutrophils.Am J Physiol Cell Physiol,2008,295:C1454-1463.
    39.Patel S, Vemula J, Konikkat S, et al. Ion channel modulators mediated alterations in NO-induced free radical generation and neutrophil membrane potential. Free Radic Res,2009,43:514-521.
    1. Orlowski J, Grinstein S. Na+/H+ exchangers of mammalian cells. J Biol Chem 1997,272: 22373-22376.
    2. Orlowski J, Kandasamy RA, Shull GE. Molecular cloning of putative members of the Na/H exchanger gene family. cDNA cloning, deduced amino acid sequence, and mRNA tissue expression of the rat Na/H exchanger NHE-1 and two structurally related proteins. J Biol Chem 1992,267:9331-9339.
    3. Nemeth ZH, Deitch EA, Lu Q, et al. NHE blockade inhibits chemokine production and NF-kappaB activation in immunostimulated endothelial cells. Am J Physiol Cell Physiol, 2002,283:C396-403.
    4. Kamachi F, Ban HS, Hirasawa N, et al. Inhibition of lipopolysaccharide-induced prostaglandin E2 production and inflammation by the Na+/H+ exchanger inhibitors.J Pharmacol Exp Ther,2007,321:345-352.
    5. Pittet JF, Wiener-Kronish JP, McElroy MC, et al. Stimulation of lung epithelial liquid clearance by endogenous release of catecholamines in septic shock in anesthetized rats. J Clin Invest,1994,94:663-671.
    6. Modelska K, Matthay MA, McElroy MC, et al.Upregulation of alveolar liquid clearance after fluid resuscitation for hemorrhagic shock in rats. Am J Physiol,1997,273:L305-314.
    7. Lin D, Zeng BX, Zhang S H, et al. Rosiglitazone, a peroxisome proliferator-activated receptor-agonist, reduces acute lung injury in endotoxemic rats. Crit Care Med,2005, 33:2309-2316.
    8. Raghavendran K, Davidson BA, Hutson AD, et al. Predictive modeling and inflammatory biomarkers in rats with lung contusion and gastric aspiration. J Trauma,2009,67:1182-90.
    9. Nacio CM, Dietz UA, Malafaia O, et al. Action of tacrolimus on Wistar rat kidneys implanted with Walker 256 carcinosarcoma. Acta Cir Bras,2010,25:98-104.
    10. Murray JF, Matthay MA, Luce JM, et al. An expanded definition of the adult respiratory distress syndrome. Am Rev Respir Dis,1988,138:720-723.
    11.姚伟,钱桂生,杨晓静.NHE-1与大鼠肺动脉平滑肌细胞增殖和凋亡.中国病理生理杂志,2001,17:207-209.
    12. Bernard GR, Artigas A, Brigham KL, et al. The American-European Consensus Conference on ARDS:definitions, mechanisms, relevant outcomes, and clinical trial coordination. Am J Respir Crit Care Med,1994,149:818-24.
    13. Glassberg AE, Luce JM, Matthay MA, et al.. Reasons for nonenrollment in a clinical trial of acute lung injury. Chest,2008,134:719-23.
    14. Ware LB, Matthay MA. The acute respiratory distress syndrome. N Engl J Med,2000,342: 1334-1349.
    15. Hughes M, Mackirdy FN, Ross J, et al. Acute respiratory distress syndrome:an audit of incidence and outcome in Scottish intensive care units. Anaesthesia,2003,58:838-845.
    16. Andrade C F, Waddell T K, Keshavjee S, et al. Innate immunity and organ transplantation: the potential role of toll-like receptors. Am J Trans,2005,5:969-975.
    17. Tian J, Lin X, Guna R, et al. The effcts of hydroxyehtyl starch on lung capillary Permeability in endotoxic rats and Possible mechanisms.Anesth Analg,2004,98:768-74.
    18.侯一峰,周艳春,周永双.全身炎性反应综合征-急性肺损伤大鼠模型的研究.中国实验动物学报,2003,11:147-150.
    19.李琦,钱桂生,张青,等.不同剂量脂多糖对大鼠急性肺损伤效应的观察.第三军医大学学报,2004,26:871-873.
    20. Bhatia M, Moochhala S. Role of inflammatory mediators in the pathophysiology of acute respiratory distress syndrome. J Pathol,2004,202:145-156.
    21. Abraham E. Neutrophils and acute lung injury. Crit Care Med,2003,31:S195-199.
    22. Nathan CF. Secretory products of macrophages. J Clin Invest,1987,79:319-326.
    23. Goodman R, Pugin J, Lee JS, et al. Cytokine mediated inflammation in acute lung injury. Cytokine Growth Factor Rev,2003,14:523-535.
    24. Podrez E A,Poliakov E, Shen Z, et al. A novel family of atherogenic oxidized phospholipids promotes macrophage foam cell formation via the scavenger receptor CD36 and is enriched in atherosclerotic lesions. J Biol Chem,2002,277:38517-38523.
    25. Orlowski J, Kandasamy RA, Shull GE. Molecular cloning of putative members of the Na/H exchanger gene family. cDNA cloning, deduced amino acid sequence, and mRNA tissue expression of the rat Na/H exchanger NHE-1 and two structurally related proteins. J Biol Chem,1992,267:9331-9339.
    26. Akram S, Teong HF, Fliegel L, et al. Reactive oxygen species-mediated regulation of the Na+-H+ exchanger 1 gene expression connects intracellular redox status with cells' sensitivity to death triggers. Cell Death Differ,2006,13:628-641.
    27. Orlinska U, Newton RC. Modification of tumor necrosis factor-alpha (TNF-alpha) production by the Na(+)-dependent HCO3- cotransport in lipopolysaccharide-activated human monocytes. Immunopharmacology,1995,30:41-50.
    28. Lin D, Zeng BX, Shang Y. Decreased expression of peroxisome proliferator-activated receptor gamma in endotoxin-induced acute lung injury. Physiol Res,2006,55:291-299.
    29. Gao J, Zeng B X, Zhou J L, et al. Protective effects of early treatment with propofol on endotoxininduced acute lung injury in rats. British Journal of Anaesthesia,2004,92: 277-279.
    30. Hyers T M, Tricomi S M, Dettemneier A P.Tumor necrosis factor Levels in seurm and bronehoalveolar lvagae fluid of Patients with the adult respiratory distress syndrome. Am Rev Respir Dis,1991,144:268-271.
    1. Bernard GR, Artigas A, Brigham KL, et al. Report of the American-European Consensus conference on acute respiratory distress syndrome:definitions, mechanisms, relevant outcomes, and clinical trial coordination:consensus Committee. J Crit Care,1994,9:72-81.
    2. Glassberg AE, Luce JM, Matthay MA, et al.. Reasons for nonenrollment in a clinical trial of acute lung injury. Chest,2008,134:719-23.
    3. Bernard GR, Artigas A, Brigham KL, et al. The American-European Consensus Conference on ARDS:definitions, mechanisms, relevant outcomes, and clinical trial coordination. Am J Respir Crit Care Med,1994,149:818-24.
    4. Ware LB, Matthay MA. The acute respiratory distress syndrome. N Engl J Med,2000,342: 1334-1349.
    5. Bhatia M, Moochhala S. Role of inflammatory mediators in the pathophysiology of acute respiratory distress syndrome. J Pathol,2004,202:145-156.
    6. Abraham E. Neutrophils and acute lung injury. Crit Care Med 2003,31:S195-199.
    7. Slepkov ER,Rainey JK,Sykes BD, et al. Structural and functional analysis of the Na+/H+exchanger. Biochem J,2007,401(Pt3):623-633.
    8. Orlowski J, Grinstein S. Diversity of the mammalian sodium/proton exchanger SLC9 gene family. Pflugers Arch,2004,447:549-565.
    9. Meima ME, Mackley JR,Barter DL. Beyond ion translocation:structural functions of the sodium-hydrogen exchanger isoform-1. Curr Opin Nephrol Hypertens,2007,16:365-72.
    lO.Fujiyoshi N, Deitch EA, Feketeova E, et al. Amiloride combined with small-volume resuscitation with hypertonic saline is superior in ameliorating trauma-hemorrhagic shock-induced lung injury in rats to the administration of either agent alone. Crit Care Med, 2005,33:2592-2598.
    11. Lin D, Zeng BX, Shang Y. Decreased expression of peroxisome proliferator-activated receptor gamma in endotoxin-induced acute lung injury. Physiol Res,2006,55:291-299.
    12. Murray JF, Matthay MA, Luce JM, et al. An expanded definition of the adult respiratory distress syndrome. Am Rev Respir Dis,1988,138:720-723.
    13. Nemeth ZH, Deitch EA, Szabo C, et al. Na+/H+ exchanger blockade inhibits enterocyte inflammatory response and protects against colitis. Am J Physiol Gastrointest Liver Physiol,2002,283:G122-132.
    14. Kamachi F, Ban HS, Hirasawa N, et al. Inhibition of lipopolysaccharide-induced prostaglandin E2 production and inflammation by the Na+/H+ exchanger inhibitors.J Pharmacol Exp Ther,2007,321:345-352.
    15. Orlowski J, Kandasamy RA, Shull GE. Molecular cloning of putative members of the Na/H exchanger gene family. cDNA cloning, deduced amino acid sequence, and mRNA tissue expression of the rat Na/H exchanger NHE-1 and two structurally related proteins. J Biol Chem,1992,267:9331-9339.
    16. Pittet JF, Wiener-Kronish JP, McElroy MC, et al. Stimulation of lung epithelial liquid clearance by endogenous release of catecholamines in septic shock in anesthetized rats. J Clin Invest,1994,94:663-671.
    17. Modelska K, Matthay MA, McElroy MC, et al.Upregulation of alveolar liquid clearance after fluid resuscitation for hemorrhagic shock in rats. Am J Physiol,1997,273:L305-314.
    18. Abraham E. Neutrophils and acute lung injury. Crit Care Med,2003,31:S195-199.
    19. Podrez E A,Poliakov E, Shen Z, et al. A novel family of atherogenic oxidized phospholipids promotes macrophage foam cell formation via the scavenger receptor CD36 and is enriched in atherosclerotic lesions. J Biol Chem,2002,277:38517-38523.
    20. Akram S, Teong HF, Fliegel L, et al. Reactive oxygen species-mediated regulation of the Na+-H+ exchanger 1 gene expression connects intracellular redox status with cells' sensitivity to death triggers. Cell Death Differ,2006,13:628-641.
    21. Carter AB, Monick MM, Hunninghake GW. Both Erk and p38 kinases are necessary for cytokine gene transcription. Am J Respir Cell Mol Biol,1999,20:751-758.
    22. Bhatia M, Moochhala S. Role of inflammatory mediators in the pathophysiology of acute respiratory distress syndrome. J Pathol,2004,202:145-156.
    23. Patel S, Vemula J, Konikkat S, et al. Ion channel modulators mediated alterations in NO-induced free radical generation and neutrophil membrane potential. Free Radic Res, 2009,43:514-521.
    24. Kaba NK, Schultz J, Law FY, et al. Inhibition of Na+/H+ exchanger enhances low pH-induced L-selectin shedding and β2-integrin surface expression in human neutrophils. Am J Physiol Cell Physiol,2008,295:C1454-1463.
    25. Nakamura N, Tanaka S, Teko Y, et al. Four Na+/H+ exchanger isoforms are distributed to Golgi and post-Golgi compartments and are involved in organelle pH regulation. J Biol Chem,2005,280:1561-1572.
    1. Huang C, Jacobson K, Schaller M.D. MAP kinases and cell migration. J.Cell Sci,2004, 117:4619-4628.
    2. Murphy L.O, Blenis J. MAPK signal specificity:the right place at the right time.Trends Biochem. Sci,2006,31:268-275.
    3. Werry T.D, Christopoulos A, Sexton P.M. Mechanisms of ERK1/2 regulation by seven-transmembrane-domain receptors. Curr. Pharm. Des,2006,12:1683-1702.
    4. Hoffmann E.K, Pedersen S.F. Sensors and signal transduction pathways in vertebrate cell volume regulation. Contrib. Nephrol,2006,152:54-104.
    5. Pedersen S.F, O'Donnell M.E, Anderson S.E, et al. Physiology and pathophysiology of Na+/H+ exchange and Na+-K+-2Cl-cotransport in the heart, brain, and blood. Am. J. Physiol. Regul. Integr. Comp. Physiol,2006,291:R1-R25.
    6. Pedersen S.F.The Na+/H+ exchanger NHE1 in stress-induced signal transduction: Implications for cell proliferation and cell death. J. Euro Pflugers Archiv Physiology,2006, 452:249-259.
    7. Meima ME, Mackley JR,Barter DL. Beyond ion translocation:structural functions of the sodium-hydrogen exchanger isoform-1. Curr Opin Nephrol Hypertens,2007,16:365-72.
    8. Javadov S, Baetz D, Rajapurohitam V, et al. Antihypertrophic effect of Na+/H+ exchanger isoform 1 inhibition is mediated by reduced mitogen-activated protein kinase activation secondary to improved mitochondrial integrity and decreased generation of mitochondrial-derived reactive oxygen species. J. Pharmacol. Exp. Ther,2006, 317:1036-1043.
    9. Lin D, Zeng BX, Shang Y. Decreased expression of peroxisome proliferator-activated receptor gamma in endotoxin-induced acute lung injury. Physiol Res,2006,55:291-299.
    10. Fujiyoshi N, Deitch EA, Feketeova E, et al. Amiloride combined with small-volume resuscitation with hypertonic saline is superior in ameliorating trauma-hemorrhagic shock-induced lung injury in rats to the administration of either agent alone. Crit Care Med, 2005,33:2592-2598.
    11. Alessi DR, Cuenda A, Cohen P, et al. PD 098059 is a specific inhibitor of the activation of mitogen-activated protein kinase kinase in vitro and in vivo. J Biol Chem,1995, 270:27489-27494.
    12.Chen XL, Xia ZF, Yu YX, et al. p38 mitogen-activated protein kinase inhibition attenuates burn-induced liver injury in rats. Burns,2005,31:320-30.
    13.Bhatia M, Moochhala S. Role of inflammatory mediators in the pathophysiology of acute respiratory distress syndrome. J Pathol,2004,202:145-156.
    14.Goodman R, Pugin J, Lee JS, et al. Cytokine mediated inflammation in acute lung injury. Cytokine Growth Factor Rev,2003,14:523-535.
    15.Schnyder-Candrian S,Quesniaux V.F,Di Padova F, et al. Dual effects of p38 MAPK on TNF-dependent bronchoconstriction and TNF-independent neutrophil recruitment in lipopolysaccharide-induced acute respiratory distress syndrome. The Journal of Immunology,2005,175:262-269.
    16.Peifer C, Wagner G, Laufer S. New approaches to the treatment of inflammatory disorders small molecule inhibitors of p38 MAP kinase. Curr Top Med Chem,2006,6:113-49.
    17.Zhao D, Seth D M, Navar LG.Enhanced Distal Nephron Sodium Reabsorption in Chronic Angiotensin II-Infused Mice.Hypertension,2009,54:120-126.
    18.Deng P, Pang ZP, Lei Z, et al. Excitatory Roles of Protein Kinase C in Striatal Cholinergic Interneurons.J Neurophysiol,2009,102:2453-2461.
    19.Turner J H, Garnovskaya MN, Coaxum SD, et al. Ca2+-Calmodulin and Janus Kinase 2 Are Required for Activation of Sodium-Proton Exchange by the Gi-Coupled 5-Hydroxytryptamine1a Receptor J. Pharmacol. Exp. Ther,2007,320:314-322.
    20.Shrode LD, Rubie EA, Woodgett JR, Cytosolic alkalinization increases stress-activated protein kinase/c- Jun NH2-terminal kinase (SAPK/JNK) activity and p38 mitogen-activated protein kinase activity by a calcium-independent mechanism.J Biol Chem,1997,272:13653-13659.
    21.Jin I, Huang H, Smith B, et al. Protein Tyrosine Kinase Involvement in Learning-Produced Changes in Hermissenda Type B Photoreceptors. J Neurophysiol,2009,102:3573-3595.
    22.Liu FQ, Liu Y, Lui VC, et al. Hypoxia modulates lipopolysaccharide induced TNF-alpha expression in murine macrophages. Exp Cell Res,2008,314:6928-6937.
    1. Slepkov ER,Rainey JK,Sykes BD, et al. Structural and functional analysis of the Na+/H+exchanger. Biochem J,2007,401(Pt3):623-633.
    2. Meima ME, Mackley JR,Barter DL. Beyond ion translocation:structural functions of the sodium-hydrogen exchanger isoform-1. Curr Opin Nephrol Hypertens,2007,16:365-72.
    3. Jhumka Z, Pervaiz S, Clement MV. Resveratrol regulates the expression of NHE-1 by repressing its promoter activity:critical involvement of intracellular H2O2 and caspases 3 and 6 in the absence of cell death. Int J Biochem Cell Biol,2009,41:945-56.
    4. Orlowski J, Kandasamy RA, Shull GE. Molecular cloning of putative members of the Na/H exchanger gene family. cDNA cloning, deduced amino acid sequence, and mRNA tissue expression of the rat Na/H exchanger NHE-1 and two structurally related proteins. J Biol Chem,1992,267:9331-9339.
    5. Kaba NK, Schultz J, Law FY, et al. Inhibition of Na+/H+ exchanger enhances low pH-induced L-selectin shedding and β2-integrin surface expression in human neutrophils.Am J Physiol Cell Physiol,2008,295:C1454-1463.
    6. Kamachi F, Ban HS, Hirasawa N, et al. Inhibition of lipopolysaccharide-induced prostaglandin E2 production and inflammation by the Na+/H+ exchanger inhibitors. J Pharmacol Exp Ther,2007,321:345-352.
    7. Patel S, Vemula J, Konikkat S, et al. Ion channel modulators mediated alterations in NO-induced free radical generation and neutrophil membrane potential. Free Radic Res,2009,43:514-521.
    8. Nemeth ZH, Deitch EA, Lu Q, et al. NHE blockade inhibits chemokine production and NF-kappaB activation in immunostimulated endothelial cells. Am J Physiol Cell Physiol 2002,283:C396-403.
    9. Jin SW, Zhang L, Lian QQ, et al. Close Functional Coupling Between Ca2+ Release-Activated Ca2+ Channels and Reactive Oxygen Species Production in Murine Macrophages. Mediators of Inflammation,2006, Article ID 36192, Pages 1-8.
    10.Ulevitch RJ,Tobias PS. Receptor-dependent mechanisms of cell stimulation by bacterial endotoxin. Annu. Rev. Immunol,1995,13:437-457.
    11.Zou XJ, Yang L, Yao SL. Propofol depresses angiotensin Ⅱ-induced cardiomyocyte hypertrophy in vitro. Exp Biol Med (Maywood).2008 Feb;233:200-208.
    12.Feng YM, Chen Q, Zhou J, et al.Effects of Leptin on the expression of tumor necrosis factor-alpha in RAW264.7 cells. Chinese Clinical Rehabilitation,2006,10:181-184.
    13.吴国明,钱桂生,黄桂君,等.钠-氢蛋白1反义表达载体对人肺腺癌细胞钠-氢交换蛋白1基因表达的影响及其意义.中华结核和呼吸杂志,2004,27:191-194.
    14. Goodman R, Pugin J, Lee JS, et al. Cytokine mediated inflammation in acute lung injury. Cytokine Growth Factor Rev,2003,14:523-535.
    15.Akram S, Teong HF, Fliegel L, et al. Reactive oxygen species-mediated regulation of the Na+-H+ exchanger 1 gene expression connects intracellular redox status with cells' sensitivity to death triggers. Cell Death Differ,2006,13:628-641.
    16.Nakamura N, Tanaka S, Teko Y, et al. Four Na+/H+ exchanger isoforms are distributed to Golgi and post-Golgi compartments and are involved in organelle pH regulation. J Biol Chem,2005,280:1561-1572.
    17.Mastronarde JG, Monick MM, Gross TJ, et al. Amiloride inhibits cytokine production in epithelium infected with respiratory syncytial virus. Am J Physiol,1996,271:L201-207.
    1. Slepkov ER,Rainey JK,Sykes BD, et al. Structural and functional analysis of the Na+/H+exchanger. Biochem J,2007,401(Pt3):623-633.
    2. Orlowski J., Grinstein S. Diversity of the mammalian sodium/proton exchanger SLC9 gene family. Pfliigers Arch,2004,447:549-565.
    3. Meima ME, Mackley JR,Barter DL. Beyond ion translocation:structural functions of the sodium-hydrogen exchanger isoform-1. Curr Opin Nephrol Hypertens,2007,16:365-72.
    4. Harold FM, Papineau D. Cation transport and electrogenesis by Streptococcus faecalis. Ⅱ. Proton and sodium extrusion. J Membr Biol,1972,8:45-62.
    5. Sardet C, Franchi A, Pouyssegur J. Molecular cloning, primary structure, and expression of the human growth factor-activatable Na+/H+ antiporter. Cell,1989,56:271-280.
    6. Cardone RA, Casavola V, Reshkin SJ. The role of disturbed pH dynamics and the Na+/H+ exchanger in metastasis. Nat Rev Cancer,2005,5:786-795.
    7. Alexander RT, Grinstein S. Tethering, recycling and activation of the epithelial sodium-proton exchanger, NHE3. Exp. Biol,2009,212:1630-1637.
    8. Masereel B, Pochet L, Laeckmann D. An overview of inhibitors of Na(+)/H(+) exchanger. Eur J Med Chem,2003,38:547-554.
    9. Alexander RT, Malevanets A, Durkan AM, et al.Membrane Curvature Alters the Activation Kinetics of the Epithelial Na+/H+ Exchanger, NHE3. J. Biol. Chem,2007,282:7376-7384.
    10. Nakamura N, Tanaka S, Teko Y, et al. Kanazawa H. Four Na+/H+ exchanger isoforms are distributed to Golgi and post-Golgi compartments and are involved in organelle pH regulation. J. Biol. Chem,2005,280:1561-1572.
    11. Numata M, Orlowski J. Molecular cloning and characterization of a novel (Na+, K+)/H+ exchanger localized to the trans-Golgi network. J. Biol. Chem,2001,276:17387-17394.
    12. Wakabayashi S, Pang T, Su X, et al. A novel topology model of the human Na+/H+ exchanger isoform 1. J Biol Chem,2000,275:7942-7949.
    13. Slepkov ER, Rainey JK, Li X, et al. Structural and Functional Characterization of Transmembrane Segment IV of the NHE1 Isoform of the Na+/H+ Exchanger. J Biol Chem, 2005,280:17863-17872.
    14. Murtazina B, Booth BJ, Bullis BL, et al. Functional analysis of polar amino acid residues in membrane associated regions of the NHE1 isoform of the Na+/H+ exchanger. Eur. J. Biochem,2001,268:1-13.
    15. Noel J, Germain D, Vadnais J. Glutamate 346 of human Na+-H+ exchanger NHE1 is crucial for modulating both the affinity for Na+ and the interaction with amiloride derivatives. Biochemistry,2003,42:15361-15368.
    16. He B.Deng C.Zhang M, et al. Reduction of intracellular pH inhibits the expression of VEGF in K562 cells after targeted inhibition of the Na+/H+ exchanger. Leukemia Research, 2007,31:507-514.
    17. Lacroix J, Poet M, Maehrel C, et al. A mechanism for the activation of the Na/H exchanger NHE-1 by cytoplasmic acidification and mitogens. EMBO Rep,2004,5:91-96.
    18. Sole FD, Babich V, Moe OW. The Calcineurin Homologous Protein-1 Increases Na+/H+-Exchanger 3 Trafficking via Ezrin Phosphorylation J. Am. Soc. Nephrol,2009,20: 1776-1786.
    19.Nuesch J P F, Bar S, Lachmann S, et al. Ezrin-Radixin-Moesin Family Proteins Are Involved in Parvovirus Replication and Spreading. Virol,2009,83:5854-5863.
    20. Lee RJ, Harlow JM, Limberis MP, et al. HCO3- Secretion by Murine Nasal Submucosal Gland Serous Acinar Cells during Ca2+-stimulated Fluid Secretion. J G Phys,2008,132: 161-183.
    21. Schelling J R, Jawdeh BG. Regulation of cell survival by Na+/H+ exchanger-1.Am J Physiol Renal Physiol,2008,295:F625-F632.
    22. Nakamoto T, Brown DA, Catalan MA, et al. Purinergic P2X7 Receptors Mediate ATP-induced Saliva Secretion by the Mouse Submandibular Gland. J. Biol. Chem,2009, 284:4815-4822.
    23. Manucha W.Carrizo L.Ruete C, et al. Apoptosis induction is associated with decreased NHE1 expression in neonatal unilateral ureleric obstruction.BJ U Int,2007,100:191-198.
    24. Bourguignon LYW, Singleton PA, Diedrich F, et al. CD44 interaction with Na+-H+ exchanger (NHE1) creates acidic microenvironments leading to hyaluronidase-2 and cathepsin B activation and breast tumor cell invasion. J Biol Chem,2004,279: 26991-27007.
    25. Bullis BL, Li XJ, Rieder CV, et al. Properties of the Na+/H+ exchanger protein-detergent-resistant aggregation and membrane microdistribution. Eur J Biochem, 2002,269:4887-4895.
    26. Cavet ME, Akhter S, Murtazina R, et al. Half-lives of plasma membrane Na+/H+ exchangers NHE1-3:plasma membrane NHE2 has a rapid rate of degradation. Am J Physiol Cell Physiol,2001,281:C2039-C2048.
    27. Grinstein S, Rotin D, Mason MJ. Na+/H+ exchange and growth factor-induced cytosolic pH changes:Role in cellular proliferation. Biochim. Biophys. Acta,1989,988:73-97.
    28. Pedersen SF, O'Donnell M E, Anderson S, et al. Physiology and pathophysiology of Na+/H+ exchange and Na+-K+-2Cl-cotransport in the heart, brain, and blood. Am. J. Physiol. Regul. Integr. Comp. Physiol,2006,291:R1-R25.
    29. Masereel B., Pochet L., Laeckmann D. An overview of inhibitors of Na+/H+ exchanger. Eur. J. Med. Chem,2003,38:547-554.
    30. Ortiz-Acevedo A, Rigor R R, Maldonado H M, et al. Coordinated control of volume regulatory Na+/H+ and K+/H+ exchange pathways in Amphiuma red blood cells. Am. J. Physiol. Cell Physiol,2010,298:C510-C520.
    31. Grinstein S, Rotin D, Mason MJ. Na+/H+ exchange and growth factor-induced cytosolic pH changes. Role in cellular proliferation. Biochim. Biophys. Acta,1989,988:73-97.
    32. Wang J, Singh D, Fliegel L. The Na+/H+ exchanger potentiates growth and retinoic acid induced differentiation of P19 embryonal carcinoma cells. J. Biol. Chem, 1997,272:26545-26549.
    33. Wu KL., Khan S, Lakhe-Reddy S, et al. The NHE1 Na+/H+ exchanger recruits ezrin/radixin/moesin proteins to regulate Akt-dependent cell survival. J. Biol. Chem, 2004,279:26280-26286.
    34. Karmazyn M, Sawyer M, Fliegel L. The Na+/H+ exchanger:a target for cardiac therapeutic intervention. Curr. Drug Targets Cardiovasc. Haematol. Disord,2005,5:323-335.
    35. Bianchini L, Kapus A, Lukacs G, et al. Responsiveness of mutants of NHE1 isoform of Na+/H+ antiport to osmotic stress. Am J Physiol Cell Physiol,1995,269:C998-C1007.
    36. Snabaitis AK, Yokoyama H, Avkiran M. Roles of mitogen-activated protein kinases and protein kinase C in α1A-adrenoreceptor-mediated stimulation of the sarcolemmal Na+-H+ exchanger. Circ. Res,2000,86:214-220.
    37. Khaled AR, Moor AN, Li A, et al. Trophic factor withdrawal:p38 mitogen-activated protein kinase activates NHE1, which induces intracellular alkalinization. Mol. Cell. Biol, 2001,21:7545-7557.
    38. Takahashi E, Abe J, Gallis B, et al. p90RSK is a serum-stimulated Na+/H+ exchanger isoform-1 kinase. Regulatory phosphorylation of serine 703 of Na+/H+ exchanger isoform-1. J. Biol. Chem,1999,274:20206-20214.
    39. Tominaga T, Ishizaki T, Narumiya S, et al. p160ROCK mediates RhoA activation of Na-H exchange. EMBO J,1998,17:4712-4722.
    40. Yan W, Nehrke K, Choi J, et al. The Nck-interacting kinase (NIK) phosphorylates the Na+-H+ exchanger NHE1 and regulates NHE1 activation by platelet-derived growth factor. J. Biol. Chem,2001,276:31349-31356.
    41. Fliegel L, Walsh MP, Singh D, et al. Phosphorylation of the carboxyl-terminal domain of the Na+/H+ exchanger by Ca2+/calmodulin-dependent protein kinase Ⅱ. Biochem. J, 1992,282:139-145.
    42. Sauvage M, Maziere P, Fathallah H, et al. Insulin stimulates NHE1 activity by sequential activation of phosphatidylinositol 3-kinase and protein kinase Cζ in human erythrocytes. Eur. J. Biochem,2000,267:955-962.
    43. Misik AJ, Perreault K, Holmes CF, et al. Protein phosphatase regulation of Na+/H+ exchanger isoform I. Biochemistry,2005,44:5842-5852.
    44. Hoffmann EK, Pedersen SF. Sensors and signal transduction pathways in vertebrate cell volume regulation. Contributions to Nephrology,2006,152:54-104.
    45. Pedersen SF. The Na+/H+ exchanger NHE1 in stress-induced signal transduction: Implications for cell proliferation and cell death. EurJ Physiol,2006,452:249-259.
    46. Shiflett M W, Brown R A, Balleine B W.Acquisition and Performance of Goal-Directed Instrumental Actions Depends on ERK Signaling in Distinct Regions of Dorsal Striatum in Rats.J. Neurosci February,2010,30:2951-2959.
    47. Donnellan F, Keating N, Geoghegan P, et al. JNK mitogen-activated protein kinase limits calcium-dependent chloride secretion across colonic epithelial cells.Am J Physiol Gastrointest Liver Physiol,2010,298:G37-G44.
    48. Xue J, Zhou D, Yao H, et al. Novel functional interaction between Na+/H+ exchanger 1 and tyrosine phosphatase SHP-2. Am. J. Physiol. Regul. Integr. Comp. Physiol,2007,292: R2406-R2416.
    49. Takewaki S, Kuro-OM, Hiroi Y, et al. Activation of Na+-H+ antiporter (NHE-1) gene expression during growth, hypertrophy and proliferation of the rabbit cardiovascular system. Journal of Molecular and Cellular Cardiology,1995,27:729-742.
    50. Yamazaki T, Komuro I, Kudoh S, et al. Role of ion channels and exchangers in mechanical stretch-induced cardiomyocyte hypertrophy. Circulation Research,1998,82:430-437.
    51. Zhao D, Seth D M, Navar LG.Enhanced Distal Nephron Sodium Reabsorption in Chronic Angiotensin II-Infused Mice.Hypertension,2009,54:120-126.
    52. Aker S, Snabaitis AK, Konietzka I, et al. Inhibition of the Na+/H+ exchanger attenuates the deterioration of ventricular function during pacing-induced heart failure in rabbits. Cardiovasc. Res,2004,63:273-282.
    53. Javadov S, Baetz D, Rajapurohitam V, et al. Antihypertrophic effect of Na+/H+ exchanger isoform 1 inhibition is mediated by reduced mitogen-activated protein kinase activation secondary to improved mitochondrial integrity and decreased generation of mitochondrial-derived reactive oxygen species. J. Pharmacol. Exp. Ther,2006, 317:1036-1043.
    54. Rotin D., Grinstein S. Impaired cell volume regulation in Na+-H+ exchange-deficient mutants. Am. J. Phyiol,1989;257:C1158-C1165.
    55. Nemeth ZH, Deitch E A, Szabo C, et al. Na+/H+ exchanger blockade inhibits enterocyte inflammatory response and protects against colitis. Am J Physiol Gastrointest Liver Physiol, 2002,283:G122-G132.
    56. Jin I, Huang H, Smith B, et al. Protein Tyrosine Kinase Involvement in Learning-Produced Changes in Hermissenda Type B Photoreceptors. J Neurophysiol,2009,102:3573-3595.
    57. Deng P, Pang ZP, Lei Z, et al. Excitatory Roles of Protein Kinase C in Striatal Cholinergic Interneurons.J Neurophysiol,2009,102:2453-2461.
    58.Turner J H, Garnovskaya MN, Coaxum SD, et al. Ca2+-Calmodulin and Janus Kinase 2 Are Required for Activation of Sodium-Proton Exchange by the Gi-Coupled 5-Hydroxytryptamine1a Receptor J. Pharmacol. Exp. Ther,2007,320:314-322.
    59.shrode LD, Rubie EA, Woodgett JR, Cytosolic alkalinization increases stress-activated protein kinase/c-Jun NH2-terminal kinase (SAPK/JNK) activity and p38 mitogen-activated protein kinase activity by a calcium-independent mechanism.J Biol Chem,1997,272: 13653-13659.
    60.Kuroki DW, Minden A, Sanchez I, et al. Regulation of a c-Jun amine-terminal kinase/stress-activated protein kinase cascade by a sodium-dependent signal transduction pathway. J Biol. Chem,1997,272:23905-23911.
    61.Yan W, Nehrke K, Choi J, et al. The Nck-interacting Kinase (NIK) Phosphorylates the Na+-H+ Exchanger NHE1 and Regulates NHE1 Activation by Platelet-derived Growth Factor.J. Biol. Chem,2001,276:31349-31356.
    62.Piechotta K, Garbarini N, England R, et al. Characterization of the interaction of the stress kinase SPAK with the Na+-K+-2C1-cotransporter in the nervous system:Evidence for a scaffolding role of the kinase J. Biol. Chem,2003,278:52848-52856.
    63.Khanna C, Wan X, Bose S, et al. The membrane-cytoskeleton linker ezrin is necessary for osteosarcoma metastasis. Nat. Med,2004,10:182-186.
    64.Touyz RM, Yao G, Schiffrin EL. Role of the actin cytoskeleton in angiotensin II signaling in human vascular smooth muscle cells. Can. J. Physiol. Pharmacol,2005,83; 91-97.
    65. Krepinsky JC, Li Y, Tang D, et al. Stretch-induced Raf-1 activation in mesangial cells requires actin cytoskeletal integrity.Cell Signal,2005,17:311-320.
    66.Pedersen SF, Owsianik G, Nilius B. TRP channels:An overview. Cell Calcium,2005, 38:233-252.
    67.Li C, Naren AP. Macromolecular complexes of cystic fibrosis transmembrane conductance regulator and its interacting partners. Pharmacol. Ther,2005,108:208-223.
    68.Levitan IB. Signaling protein complexes associated with neuronal ion channels. Nat. Neurosci,2006,9:305-310.
    69. Shiflett MW, Martini R P, Mauna J C, Cue-Elicited Reward-Seeking Requires Extracellular Signal-Regulated Kinase Activation in the Nucleus Accumbens.J. Neurosci,2008,28:1434 1443.
    70.Pang T, Su X, Wakabayashi S, et al. Calcineurin homologous protein as an essential cofactor for Na+/H+ exchangers. J. Biol. Chem,2001,276:17367-17372.
    71.Chen S, Khan ZA, Karmazyn M, et al. Kinase-dependent modification of dendritic excitability after long-term potentiation. J. Physiol., January 1,2009,587:115-125.
    72.Li X, Liu Y, Kay CM, et al. The Na+/H+ exchanger cytoplasmic tail:structure, function, and interactions with tescalcin. Biochemistry,2003,42:7448-7456.
    73.Li X, Liu Y, Alvarez BV, et al. A novel carbonic anhydrase II binding site regulates NHE1 activity. Biochemistry,2006,45:2414-2424.
    74.Baumgartner M, Patel H, Barber DL. Na+/H+ exchanger NHE1 as plasma membrane scaffold in the assembly of signaling complexes. Am. J. Physiol. Cell Physiol, 2004,287:C844-C850.
    75.Aharonovitz O, Zaun HC, Balla T, et al. Intracellular pH regulation by Na+/H+ exchange requires phosphatidylinositol 4,5-bisphosphate. J. Cell Biol,2000,150:213-224.
    76.Glassberg AE, Luce JM, Matthay MA, et al.. Reasons for nonenrollment in a clinical trial of acute lung injury. Chest,2008,134(4):719-23.
    77. Ware LB, Matthay MA. The acute respiratory distress syndrome. N Engl J Med,2000,342: 1334-1349.
    78.Hughes M, Mackirdy FN, Ross J, et al. Acute respiratory distress syndrome:an audit of incidence and outcome in Scottish intensive care units. Anaesthesia,2003,58:838-845.
    79.Bhatia M, Moochhala S. Role of inflammatory mediators in the pathophysiology of acute respiratory distress syndrome. J Pathol,2004,202:145-156.
    80.Abraham E. Neutrophils and acute lung injury. Crit Care Med,2003,31:S195-199.
    81.Bachofen M, Weibel ER. Structural alterations of lung parenchyma in the adult respiratory distress syndrome. Clin Chest Med,1982,3:35-56.
    82.Steinberg KP, Milberg JA, Martin TR, et al. Evolution of bronchoalveolar cell populations in the adult respiratory distress syndrome. Am J Respir Crit Care Med,1994,150:113-122.
    83.Warshawski FJ, Sibbald W, Driedger A,et al. Abnormal neutrophil-pulmonary interaction in the adult respiratory distress syndrome:qualitative and quantitative assessment of pulmonary-neutrophil kinetics in humans with in vivo indium-111 neutrophil scintigraphy. Am Rev Respir Dis,1986,133:792-804.
    84.Laufe MD, Simon RH, Flint A,et al. Adult respiratory distress syndrome in neutropenic patients. Am J Med,1986,80:1022-1026.
    85.Matthay MA. Conference summary:acute lung injury. Chest,1999,116:119S-126S.
    86.Prescott SM, McIntyre TM, Zimmerman G. Two of the usual suspects, platelet-activating factor and its receptor, implicated in acute lung injury. J Clin Invest,1999,104:1019-1020.
    87.Hayashi H, Aharonovitz O, Alexander RT, et al. Na+/H+ exchange and pH regulation in the control of neutrophil chemokinesis and chemotaxis. Am J Physiol Cell Physiol, 2008,294:C526-C534.
    88.Doerschuk CM, Quinlan WM, Doyle NA. et al. The role of P-selectin and ICAM-1 in acute lung injury as determined using blocking antibodies and mutant mice. J Immunol,1996,157: 4609-4614.
    89.Puneet P, Moochhala S, Bhatia M. Chemokines in acute respiratory distress syndrome. Am J Physiol Lung Cell Mol Physiol,2005,288:L3-15.
    90.Moraes TJ, Zurawska JH, Downey GP. Neutrophil granule contents in the pathogenesis of lung injury. Curr Opin Hematol,2006,13:21-27.
    91.Tsushima K, King L S, Aggarwal NR, et al. Acute Lung Injury Review. Internal Medicine, 2009,48:621-630.
    92.Suzuki T, Moraes TJ, Vachon E, et al. Proteinase-activated receptor-1 mediates elastase-induced apoptosis of human lung epithelial cells. Am J Respir Cell Mol Biol, 2005,33:231-247.
    93.Gando S, Kameue T, Nanzaki S, et al. Increased neutrophil elastase, persistent intravascular coagulation, and decreased fibrinolytic activity in patients with posttraumatic acute respiratory distress syndrome. J Trauma,1997,42:1068-1072.
    94.Pugin J, Verghese G, Widmer M C, et al. The alveolar space is the site of intense inflammatory and profibrotic reactions in the early phase of ARDS. Crit Care Med,1999,27: 304-312.
    95.Jin SW, Zhang L, Lian QQ, et al. Posttreatment with Aspirin-Triggered Lipoxin A4 Analog Attenuates Lipopolysaccharide-Induced Acute Lung Injury in Mice:The Role of Heme Oxygenase-1. Anesth & Analg,2007,104:369-377.
    96. Suter PM, Suter S, Girardin E, et al. High bronchoalveolar levels of tumor necrosis factor and its inhibitors, interleukin-1, interferon, and elastase in patients with adult respiratory distress syndrome after trauma, shock, or sepsis. Am Rev Respir Dis,1992,145:1016-1022.
    97.Geerts L, Jorens PG, Willems J, et al. Natural inhibitors of neutrophil function in acute respiratory distress syndrome. Crit Care Med,2001,29:1920-1924.
    98.Gadek JE, Pacht ER. The interdependence of lung antioxidants and antiprotease defense in ARDS. Chest,1996,110:273S-277S.
    99.Lesur O, Kokis A, Hermans C, et al. Interleukin-2 involvement in early acute respiratory distress syndrome:relationship with polymorphonuclear neutrophil apoptosis and patient survival. Crit Care Med,2000,28:3814-3822.
    100.Sookhai S, Wang JJ, McCourt M, et al. A novel therapeutic strategy for attenuating neutrophil-mediated lung injury in vivo. Ann Surg,2002,235:285-291.
    101.Goodman R, Pugin J, Lee JS, et al. Cytokine mediated inflammation in acute lung injury. Cytokine Growth Factor Rev,2003,14:523-535.
    102.Miller EJ, Cohen AB, Matthay MA. Increased interleukin-8 concentrations in the pulmonary edema fluid of patients with acute respiratory distress syndrome from sepsis. Crit Care Med,1996,24:1448-1454.
    103.Yokoi K, Mukaida N, Harada A, et al. Prevention of endotoxemia-induced acute respiratory distress syndrome-like lung injury in rabbits by a monoclonal antibody to IL-8. Lab Invest,1997,76:375-384.
    104.Martin TR. Cytokines and the acute respiratory distress syndrome (ARDS):a question of balance. Nat Med,1997,3:272-273.
    105.Parsons PE. Interleukin-10:the ambiguity in sepsis continues. Crit Care Med,1998,26: 818-819.
    106.Kurdowska A, Noble JM, Steinberg KP, et al. Anti-interleukin 8 autoantibody:interleukin 8 complexes in the acute respiratory distress syndrome. Am J Respir Crit Care Med, 2001,163:463-468.
    107.Fan J, Ye RD, Malik AB. Transcriptional mechanisms in acute lung injury. Am J Physiol Lung Cell Mol Physiol,2001,281:L1037-L50.
    108.Christman JW, Sadikot RT, Blackwell T. The role of nuclear factor-κB in pulmonary diseases. Chest,2000,117:1482-1487.
    109.Jhumka Z, Pervaiz S, Clement MV. Resveratrol regulates the expression of NHE-1 by repressing its promoter activity:critical involvement of intracellular H2O2 and caspases 3 and 6 in the absence of cell death. Int J Biochem Cell Biol,2009,41:945-56.
    110.Lum H, Roebuck KA. Oxidant stress and endothelial cell dysfunction. Am J Physiol Cell Physiol,2001,280:C719-C741.
    111.Waters CM, Savla U, Panos RJ. KGF prevents hydrogen peroxide-induced increases in airway epithelial cell permeability. Am J Physiol,1997,272:L681-L9.
    112.Chabot F, Mitchell JA, Gutteridge JMC, et al. Reactive oxygen species in acute lung injury. Eur Respir J,1998,11:745-757.
    113.Orlowski J, Kandasamy RA, Shull GE. Molecular cloning of putative members of the Na/H exchanger gene family. cDNA cloning, deduced amino acid sequence, and mRNA tissue expression of the rat Na/H exchanger NHE-1 and two structurally related proteins. J Biol Chem,1992,267:9331-9339.
    114.Akram S, Teong HF, Fliegel L, et al. Reactive oxygen species-mediated regulation of the Na+-H+ exchanger 1 gene expression connects intracellular redox status with cells' sensitivity to death triggers. Cell Death Differ,2006,13:628-641.
    115.Orlinska U, Newton RC. Modification of tumor necrosis factor-alpha (TNF-alpha) production by the Na(+)-dependent HCO3- cotransport in lipopolysaccharide-activated human monocytes. Immunopharmacology,1995,30:41-50.
    116.Nemeth ZH, Deitch EA, Lu Q, et al. NHE blockade inhibits chemokine production and NF-kappaB activation in immunostimulated endothelial cells. Am J Physiol Cell Physiol,2002,283:C396-403.
    117.Kamachi F, Ban HS, Hirasawa N, et al. Inhibition of lipopolysaccharide-induced prostaglandin E2 production and inflammation by the Na+/H+ exchanger inhibitors.J Pharmacol Exp Ther,2007,321:345-352.
    118.Nemeth ZH, Deitch EA, Szabo C, et al. Na+/H+ exchanger blockade inhibits enterocyte inflammatory response and protects against colitis. Am J Physiol Gastrointest Liver Physiol, 2002,283:G122-132.
    119.Modelska K, Matthay MA, McElroy MC, et al.Upregulation of alveolar liquid clearance after fluid resuscitation for hemorrhagic shock in rats. Am J Physiol,1997,273:L305-314.
    120.Pittet JF, Wiener-Kronish JP, McElroy MC, et al. Stimulation of lung epithelial liquid clearance by endogenous release of catecholamines in septic shock in anesthetized rats. J Clin Invest,1994,94:663-671.
    121.Bhatia M, Moochhala S. Role of inflammatory mediators in the pathophysiology of acute respiratory distress syndrome. J Pathol,2004,202:145-156.
    122.Kaba NK, Schultz J, Law FY, et al. Inhibition of Na+/H+ exchanger enhances low pH-induced L-selectin shedding and β2-integrin surface expression in human neutrophils.Am J Physiol Cell Physiol,2008,295:C1454-1463.
    123.Patel S, Vemula J, Konikkat S, et al. Ion channel modulators mediated alterations in NO-induced free radical generation and neutrophil membrane potential. Free Radic Res,2009,43:514-521.
    124.Nakamura N, Tanaka S, Teko Y, et al. Four Na+/H+ exchanger isoforms are distributed to Golgi and post-Golgi compartments and are involved in organelle pH regulation. J Biol Chem,2005,280:1561-1572.
    125.Mastronarde JG, Monick MM, Gross TJ, et al. Amiloride inhibits cytokine production in epithelium infected with respiratory syncytial virus. Am J Physio 1,1996,271:L201-207.
    1. Kyriakis J M., Avruch J. Mammalian mitogen-activated protein kinase signal transduction pathways activated by stress and inflammation. Physiol. Rev,2001,81:807-869.
    2. Huang C, Jacobson K, Schaller M.D. MAP kinases and cell migration. J.Cell Sci,2004, 117:4619-4628.
    3. Morrison D.K, Davis R.J. Regulation of MAP Kinase Signaling Modules by Scaffold Proteins in Mammals. Annu. Rev. Cell Dev, Biol,2003,19:91-118.
    4. Murphy L.O, Blenis J. MAPK signal specificity:the right place at the right time.Trends Biochem. Sci,2006,31:268-275.
    5.Werry T.D, Christopoulos A, Sexton P.M. Mechanisms of ERK1/2 regulation by seven-transmembrane-domain receptors. Curr. Pharm. Des,2006,12:1683-1702.
    6. Hoffmann E.K, Pedersen S.F. Sensors and signal transduction pathways in vertebrate cell volume regulation. Contrib. Nephrol,2006,152:54-104.
    7. Pedersen S.F, O'Donnell M.E, Anderson S.E., Cala P.M. Physiology and pathophysiology of Na+/H+ exchange and Na+-K+-2Cl-cotransport in the heart, brain, and blood. Am. J. Physiol. Regul. Integr. Comp. Physiol,2006,291:R1-R25.
    8. Putney L.K, Denker S.P, Barber D.L. The changing face of the Na+/H+ exchanger, NHE1: Structure, regulation, and cellular actions. Annu. Rev. Pharmacol. Toxicol,2002,42: 527-552.
    9. Orlowski J, Grinstein S. Diversity of the mammalian sodium/proton exchanger SLC9 gene family. J. Euro Pflugers Archiv Physiology,2004,447:549-565.
    10. Pedersen S.F.The Na+/H+ exchanger NHE1 in stress-induced signal transduction: Implications for cell proliferation and cell death. J. Euro Pflugers Archiv Physiology,2006, 452:249-259.
    11. Bianchini L, L'Allemain G, Pouyssegur J. The p42/p44 mitogen-activated protein kinase cascade is determinant in mediating activation of the Na+/H+ exchanger (NHE1 isoform) in response to growth factors. J. Biol. Chem,1997,272:271-279.
    12. Takahashi E, Abe J, Gallis B, et al. p90(RSK) is a serum-stimulated Na+/H+ exchanger isoform-1 kinase. Regulatory phosphorylation of serine 703 of Na+/H+ exchanger isoform-1. J. Biol. Chem,1999,274:20206-20214.
    13.Mukhin YV, Garnovskaya MN, Ullian ME, et al. ERK Is Regulated by Sodium-Proton Exchanger in Rat Aortic Vascular Smooth Muscle Cells. J Biol Chem,2004,279:1845-1852.
    14. Foutz R M,Grimm P R, Sansom S C.Insulin increases the activity of mesangial BK channels through MAPK signaling. Am J Physiol Renal Physiol,2008,294:F1465-F1472.
    15. Qadri Y J, Song Y, Fuller CM, et al. Amiloride Docking to Acid-sensing Ion Channel-1. J Biol Chem,2010,285:9627-9635.
    16. Cooper JA, Bowen-Pope DF, Raines E, et al. Similar effects of platelet-derived growth factor and epidermal growth factor on the phosphorylation of tyrosine in cellular proteins. Cell,1982,31:263-273.
    17.Rossomando AJ, Payne DM, Weber MJ, et al. Evidence that pp42, a major tyrosine kinase target protein, is a mitogen-activated serine/threonine protein kinase. Proc Natl Acad Sci U S A,1989,86:6940-6943.
    18.Kazlauskas A, Copper J A. Autophosphorylation of the pdgf receptor in the kinase insert region regulates interactions with cell-proteins. Cell,1989,58:1121-1133.
    19.Ray L B, Sturgill T W.Insulin-stimulated microtubule-associated protein kinase is phosphorylated on tyrosine and threonine in vivo. Proc Natl Acad Sci U S A,1988, 85:3753-3757.
    20.Cyer M S.Regulation of nuclear localization during signaling. J Biol Chem,2001, 276:20805-20808.
    21.Aplin A E, Stewart S A, Assoian R K, et al. Integrin2 mediated adhesion regulates ERK nuclear translocation and phosphorylation of Elk21. J Cell Biol,2001,153:273-281.
    22.Khokhlatchev AV, Canagarajah B,Wilsbacher J, et al. Phosphorylation of the MAP kinase ERK2 promotesits Homo dimerization and nuclear translocation.Cell,1998,93:605-615.
    23.Gonzalez MV, JimenezB, BercianoMT, et al. Glucocorticoids antagonizeAP21 by inhibiting the activation/phosphorylation of JNK without affecting its subcellular distribution. J Cell Biol,2000,150:1199-1207.
    24.Kyriakis JM,AvruchJ.Mammalian mitogen 2 activated protein Kinase signal transduction pathways activated by stress and inflammation. PhysiolRev,2001,812:807-869
    25.Nishimoto S, Nishida E. MAPK signalling:ERK5 versus ERK1/2.EMBO Rep,2006, 7:782-786.
    26.Bogoyevitch M.A, Court N.W. Counting on mitogen-activated protein kinases-ERKs 3,4, 5,6,7 and 8. Cell Signal,2004,16:1345-1354.
    27.Canagarajah B J, Khokhlatchev A, Cobb M H, et al. Activation mechanism of the MAP kinase ERK2 by dual phosphorylation. Cell,1997,90:859-869.
    28.Bradham C, McClay D R. p38 MAPK in development and cancer. Cell Cycle, 2006,5:824-828.
    29.Arimura N, Kaibuchi K. Neuronal polarity:from extracellular signals to intracellular mechanisms. Nat Rev Neurosci,2007,8:194-205.
    30.Engelberg D. Stress-activated protein kinases-Tumor suppressors or tumor initiators? Semin. Cancer Biol.2004,14:271-282.
    31. Hagemann C, Blank J L. The ups and downs of MEK kinase interactions. Cell Signal,2001, 13:863-875.
    32. Uhlik M.T, Abell A.N, Cuevas B.D, et al. Wiring diagrams of MAPK regulation by MEKK1,2, and 3. Cell.Biol,2004,82:658-663.
    33. Johnson G.L, Lapadat R. Mitogen-activated protein kinase pathways mediated by ERK, JNK, and p38 protein kinases. Science,2002,298:1911-1912.
    34.Baumgartner M, Patel H, Barber D.L. Na+/H+ exchanger NHE1 as plasma membrane scaffold in the assembly of signaling complexes. Am. J. Physiol. Cell Physiol,2004, 287:C844-C850.
    35.Uhlik M.T, Abell A.N, Johnson N.L, et al. Rac-MEKK3-MKK3 scaffolding for p38 MAPK activation during hyperosmotic shock. Nat. Cell Biol,2003,5:1104-1110.
    36.Kolch W. Coordinating ERK/MAPK signalling through scaffolds and inhibitors.Nat.Rev.Mol.Cell Biol,2005,6:827-837.
    37. Zhang H., Shi X., Hampong M, et al. Stress-induced Inhibition of ERK1 and ERK2 by Direct Interaction with p38 MAP Kinase. J. Biol. Chem,2001,276:6905-6908.
    38. Friis M.B, Friborg C.R, Schneider L, et al.Cell shrinkage as a signal to apoptosis in NIH 3T3 fibroblasts. J. Physiol,2005,567:427-443.
    39.Marshall C J. Specificity of receptor tyrosine kinase signaling:transient versus sustained extracellular signal-regulated kinase activation. Cell,1995,80:179-185.
    40. Schliess F., Heinrich S. Haussinger D. Hyperosmotic induction of the mitogen-activated protein kinase phosphatase MKP-1 in H4IIE rat hepatoma cells. Arch. Biochem. Biophys, 1998,351;35-40.
    41. Lornejad-Schafer M., Schafer C., Richter L, et al. Osmotic regulation of MG-132-induced MAP-kinase phosphatase MKP-1 expression in H4IIE rat hepatoma cells. Cell Physiol. Biochem,2005,16:193-206.
    42. Farooq A, Zhou M.M. Structure and regulation of MAPK phosphatases. Cell Signal,2004, 16:769-779.
    43. Alessi DR, Gomez N, Moorhead G, et al. Inactivation of p42 MAP kinase by protein phosphatase 2A and a protein tyrosine ph osphatase, but not CL100, in various cell lines. Curr Biol,1995,5:283-295.
    44. Slepkov ER,Rainey JK,Sykes BD, et al. Structural and functional analysis of the Na+/H+exchanger. Biochem J,2007,401(Pt3):623-633.
    45. Orlowski J., Grinstein S. Diversity of the mammalian sodium/proton exchanger SLC9 gene family. Pflugers Arch,2004,447:549-565.
    46.Meima M E,Mackley J R,Barter D L. Beyond ion translocation:structural functions of the sodium-hydrogen exchanger isoform-1. Curr Opin Nephrol Hypertens,2007,16:365-72.
    47.Pittet J F, Wiener-Kronish J P, McElroy M C, et al. Stimulation of lung epithelial liquid clearance by endogenous release of catecholamines in septic shock in anesthetized rats. J Clin Invest,1994,94:663-671
    48. Modelska K, Matthay M A, McElroy M C, et al.Upregulation of alveolar liquid clearance after fluid resuscitation for hemorrhagic shock in rats. Am J Physiol,1997,273:L305-314.
    49.Orlinska U, Newton R C. Modification of tumor necrosis factor-alpha (TNF-alpha) production by the Na(+)-dependent HCO3- cotransport in lipopolysaccharide-activated human monocytes. Immunopharmacology,1995,30:41-50.
    50.Wakabayashi S, Pang T, Su X, et al. A novel topology model of the human Na+/H+ exchanger isoform 1. J Biol Chem,2000,275:7942-7949.
    51.Slepkov E R, Rainey J K, Li X, et al. Structural and Functional Characterization of Transmembrane Segment IV of the NHE1 Isoform of the Na+/H+ Exchanger. J Biol Chem, 2005,280:17863-17872.
    52.Slepkov E R, Chow S, Lemieux M J, et al. Proline residues in transmembrane segment IV are critical for activity, expression and targeting of the Na+/H+ exchanger isoform 1. J Biol Chem,2004,379(Pt1):31-38.
    53.He B.Deng C.Zhang M, et al. Reduction of intracellular pH inhibits the expression of VEGF in K562 cells after targeted inhibition of the Na+/H+ exchanger. Leukemia Research,2007, 31:507-514.
    54. Pang T, Su X, Wakabayashi S, et al. Calcineurin homologous protein as an essential cofactor for Na+/H+ exchangers. J. Biol. Chem,2001,276:17367-17372.
    55. Lin X, Barber DL. A calcineurin homologous protein inhibits GTPase-stimulated Na-H exchange. J. Biol. Chem,1996,93:12631-12636.
    56. Wakabayashi S, Bertrand B, Ikeda T, et al. Mutation of calmodulin-binding site renders the Na+/H+ exchanger (NHE1) highly H+-sensitive and Ca2+ regulation-defective. J. Biol. Chem,1994,269:13710-5.
    57. Chen S, Khan ZA, Karmazyn M, et al. Kinase-dependent modification of dendritic excitability after long-term potentiation. J. Physiol., January 1,2009,587:115-125.
    58. Pang THT, Mori H, Shigekawa M, et al. Role of calcineurin B homologous protein in pH regulation by the Na+/H+ exchanger 1:tightly bound Ca2+ ions as important structural elements. Biochemistry,2004,43:3628-3636.
    59. Li X, Liu Y, Kay CM, et al. The Na+/H+ exchanger cytoplasmic tail:structure, function, and interactions with tescalcin. Biochemistry,2003,42:7448-7456.
    60. Mailander J, Muller-Esterl W, Dedio J. Human homolog of mouse tescalcin associates with Na+/H+ exchanger type-1. FEBS Lett,2001,507:331-335.
    61. Li X, Liu Y, Alvarez BV, et al. A novel carbonic anhydrase Ⅱ binding site regulates NHE1 activity. Biochemistry,2006,45:2414-2424.
    62.Li X, Alvarez B, Casey JR, et al. Carbonic anhydrase Ⅱ binds to and enhances activity of the Na+/H+ exchanger. J. Biol. Chem,2002,277:36085-36091.
    63. Cechin SR, Dunkley PR, Rodnight R. Signal transduction mechanisms involved in the proliferation of C6 glioma cells induced by lysophosphatidic acid. Neurochemical Research, 2005,30:603-611.
    64. Moor AN, Fliegel L. Protein kinase mediated regulation of the Na+/H+ exchanger in the rat myocardium by MAP-kinase-dependent pathways. J. Biol. Chem,1999,274:22985-22992.
    65. Wang H, Silva NLCL, Lucchesi PA, et al. Phosphorylation and regulation of the Na+/H+ exchanger through mitogen-activated protein kinase. Biochemistry,1997,36:9151-9158.
    66. Snabaitis AK, Yokoyama H, Avkiran M. Roles of mitogen-activated protein kinases and protein kinase C in al A-adrenoreceptor-mediated stimulation of the sarcolemmal Na+-H+ exchanger. Circ. Res,2000,86:214-220.
    67. Khaled AR, Moor AN, Li A, et al. Trophic factor withdrawal:p38 mitogen-activated protein kinase activates NHE1, which induces intracellular alkalinization. Mol. Cell. Biol, 2001,21:7545-7557.
    68. Takahashi E, Abe J, Berk BC. Angiotensin Ⅱ stimulates p90rsk in vascular smooth muscle cells:a potential Na+/H+ exchanger kinase. Hypertension,1997,29:1265-1272.
    69. Takahashi E, Abe J, Gallis B, et al. p90RSK is a serum-stimulated Na+/H+ exchanger isoform-1 kinase. Regulatory phosphorylation of serine 703 of Na+/H+ exchanger isoform-1. J. Biol. Chem,1999,274:20206-20214.
    70. Tominaga T, Ishizaki T, Narumiya S, et al. p160ROCK mediates RhoA activation of Na-H exchange. EMBO J,1998,17:4712-4722.
    71. Yan W, Nehrke K, Choi J, et al. The Nck-interacting kinase (NIK) phosphorylates the Na+-H+ exchanger NHE1 and regulates NHE1 activation by platelet-derived growth factor. J. Biol. Chem,2001,276:31349-31356.
    72. Fliegel L, Walsh MP, Singh D, et al. Phosphorylation of the carboxyl-terminal domain of the Na+/H+ exchanger by Ca2+/calmodulin-dependent protein kinase Ⅱ. Biochem. J, 1992,282:139-145.
    73. Haworth RS, Sinnett-Smith J, Rozengurt E, et al. Protein kinase D inhibits plasma membrane Na+/H+ exchanger activity. Am. J. Physiol,1999,277:C1202-C1209.
    74. Sauvage M, Maziere P, Fathallah H, et al. Insulin stimulates NHE1 activity by sequential activation of phosphatidylinositol 3-kinase and protein kinase Cζ in human erythrocytes. Eur. J. Biochem,2000,267:955-962.
    75. Sardet C, Fafournoux P, Pouyssegur J. a-Thrombin, epidermal growth factor, and okadaic acid activate the Na+/H+ exchanger, NHE-1, by phosphorylating a set of common sites. J. Biol. Chem,1991,266:19166-19171.
    76. Misik AJ, Perreault K, Holmes CF, et al. Protein phosphatase regulation of Na+/H+ exchanger isoform I. Biochemistry,2005,44:5842-5852.
    77. Hoffmann EK, Pedersen SF. Sensors and signal transduction pathways in vertebrate cell volume regulation. Contributions to Nephrology,2006,152:54-104.
    78. Pedersen SF, O'Donnell ME, Anderson SE, et al. Physiology and pathophysiology of Na+/H+ exchange and Na+-K+-2Cl-cotransport in the heart, brain, and blood. Am J Physiol Regul Integr Comp Physiol.2006,91:R1-25.
    79. Putney LK, Denker SP, Barber DL. The changing face of the Na+/H+ exchanger, NHE1: Structure, regulation, and cellular actions. Annual Review of Pharmacology and Toxicology,2002,42:527-552.
    80. Pedersen SF. The Na+/H+ exchanger NHE1 in stress-induced signal transduction: Implications for cell proliferation and cell death. EurJ Physiol,2006,452:249-259.
    81. Bianchini L, L'Allemain G, Pouyssegur J. The p42/p44 mitogen-activated protein kinase cascade is determinant in mediating activation of the Na+/H+ exchanger (NHE1 isoform) in response to growth factors Journal of Biological Chemistry,1997,272:271-279.
    82. Takahashi E, Abe J.-I, Gallis B, et al. p90(RSK) is a serum-stimulated Na+/H+ exchanger isoform-1 kinase. Regulatory phosphorylation of serine 703 of Na+/H+ exchanger isoform-1 Journal of Biological Chemistry,1999,274:20206-20214.
    83. Klein JD, Lamitina ST, O'Neill WC. JNK is a volume-sensitive kinase that phosphorylates the Na-K-2C1 cotransporter in vitro Am J Physiol Cell Physiol,1999,277:C425-C431.
    84. Hu Z, Wang Y, Graham WV. MAPKAPK-2 is a critical signaling intermediate in NHE3 activation following Na+-glucose cotransport Journal of Biological Chemistry,2006, 281:24247-24253.
    85. Shiflett M W, Brown R A, Balleine B W. Acquisition and Performance of Goal-Directed Instrumental Actions Depends on ERK Signaling in Distinct Regions of Dorsal Striatum in Rats.J. Neurosci February,2010,30:2951-2959.
    86. Donnellan F, Keating N, Geoghegan P, et al. JNK mitogen-activated protein kinase limits calcium-dependent chloride secretion across colonic epithelial cells. Am J Physiol Gastrointest Liver Physiol,2010,298:G37-G44.
    87. Khaled AR., Moor AN, Li A, et al. Trophic factor withdrawal:p38 mitogen-activated protein kinase activates NHE1, which induces intracellular alkalinization. Molecular and Cellular Biology,2001,21:7545-7557.
    88. Pederson SF, Varming C, Christensen ST, et al. Mechanisms of activation of NHE by cell shrinkage and by calyculin A in Ehrlich ascites tumor cells. J membrane biology,2002, 189:67-81.
    89. Xue J, Zhou D, Yao H, et al. Novel functional interaction between Na+/H+ exchanger 1 and tyrosine phosphatase SHP-2. Am. J. Physiol. Regul. Integr. Comp. Physiol,2007,292: R2406-R2416.
    90. Takewaki S, Kuro-OM, Hiroi Y, et al. Activation of Na+-H+ antiporter (NHE-1) gene expression during growth, hypertrophy and proliferation of the rabbit cardiovascular system. Journal of Molecular and Cellular Cardiology,1995,27:729-742.
    91. Yamazaki T, Komuro I, Kudoh S, et al. Role of ion channels and exchangers in mechanical stretch-induced cardiomyocyte hypertrophy. Circulation Research,1998,82:430-437.
    92.Mukhin Y.V, Garnovskaya M.N, Ullian M.E, et al. ERK Is Regulated by Sodium-Proton Exchanger in Rat Aortic Vascular Smooth Muscle Cells. J.Biol.Chem,2004,279:1845-1852.
    93. Aker S, Snabaitis AK, Konietzka I, et al. Inhibition of the Na+/H+ exchanger attenuates the deterioration of ventricular function during pacing-induced heart failure in rabbits. Cardiovasc. Res,2004,63:273-282.
    94. Javadov S, Baetz D, Rajapurohitam V, et al. Antihypertrophic effect of Na+/H+ exchanger isoform 1 inhibition is mediated by reduced mitogen-activated protein kinase activation secondary to improved mitochondrial integrity and decreased generation of mitochondrial-derived reactive oxygen species. J. Pharmacol. Exp. Ther,2006, 317:1036-1043.
    95. Rotin D., Grinstein S. Impaired cell volume regulation in Na+-H+ exchange-deficient mutants. Am J.Phyiol,1989,257:C1158-C1165.
    96. Nemeth ZH, Deitch E A, Szabo C, et al. Na+/H+ exchanger blockade inhibits enterocyte inflammatory response and protects against colitis. Am J Physiol Gastrointest Liver Physiol, 2002,283:G122-G132.
    97. Jin I, Huang H, Smith B, et al. Protein Tyrosine Kinase Involvement in Learning-Produced Changes in Hermissenda Type B Photoreceptors.J Neurophysiol,2009,102:3573-3595.
    98. Ortiz-Acevedo A, Rigor R R, Maldonado H M, et al. Coordinated control of volume regulatory Na+/H+ and K+/H+ exchange pathways in Amphiuma red blood cells. Am. J. Physiol. Cell Physiol,2010,298:C510-C520.
    99. Pedersen S F, Darborg B V, Rasmussen M, et al. The Na+/H+ exchanger, NHE1, differentially regulates mitogen-activated protein kinase subfamilies after osmotic shrinkage in Ehrlich Lettre Ascites cells. Cell Physiol Biochem,2007,20:735-750.
    100. shrode LD, Rubie EA, Woodgett JR, Cytosolic alkalinization increases stress-activated protein kinase/c-Jun NH2-terminal kinase (SAPK/JNK) activity and p38 mitogen-activated protein kinase activity by a calcium-independent mechanism. J Biol Chem,1997,272: 13653-13659.
    101. Kuroki DW, Minden A, Sanchez I, et al. Regulation of a c-Jun amine-terminal kinase/stress-activated protein kinase cascade by a sodium-dependent signal transduction pathway. J Biol. Chem,1997,272:23905-23911.
    102. Yan W, Nehrke K, Choi J, et al. The Nck-interacting Kinase (NIK) Phosphorylates the Na+-H+ Exchanger NHE1 and Regulates NHE1 Activation by Platelet-derived Growth Factor. J. Biol. Chem,2001,276:31349-31356.
    103. Su Y-C, Han J, Xu S, et al. NIK is a new Ste20-related kinase that binds NCK and MEKK1 and activates the SAPK/JNK cascade via a conserved regulatory domain. EMBO J, 1997,16:1279-1290.
    104. Piechotta K, Garbarini N, England R, et al. Characterization of the interaction of the stress kinase SPAK with the Na+-K+-2Cl-cotransporter in the nervous system:Evidence for a scaffolding role of the kinase J. Biol. Chem,2003,278:52848-52856.
    105. Khanna C, Wan X, Bose S, et al. The membrane-cytoskeleton linker ezrin is necessary for osteosarcoma metastasis. Nat. Med,2004,10:182-186.
    106. Touyz RM, Yao G, Schiffrin EL. Role of the actin cytoskeleton in angiotensin II signaling in human vascular smooth muscle cells. Can. J. Physiol. Pharmacol,2005,83; 91-97.
    107. Krepinsky JC, Li Y, Tang D, et al. Stretch-induced Raf-1 activation in mesangial cells requires actin cytoskeletal integrity.Cell Signal,2005,17:311-320.
    108. Fanger GR, Widmann C, Porter AC, et al. R14-3-3 proteins interact with specific MEK kinases. J. Biol. Chem,1998,273:3476-3483.
    109. Pedersen SF, Owsianik G, Nilius B. TRP channels:An overview. Cell Calcium,2005, 38:233-252.
    110. Li C, Naren AP. Macromolecular complexes of cystic fibrosis transmembrane conductance regulator and its interacting partners. Pharmacol. Ther,2005,108:208-223.
    111. Levitan IB. Signaling protein complexes associated with neuronal ion channels. Nat. Neurosci,2006,9:305-310.
    112. Shiflett M W, Martini R P, Mauna J C, Cue-Elicited Reward-Seeking Requires Extracellular Signal-Regulated Kinase Activation in the Nucleus Accumbens.J. Neurosci, 2008,28:1434-1443.
    113. Hegle A.P, Marble D.D, Wilson Proc GF. A voltage-driven switch for ion-independent signaling by Ether-go-go K+ channels. Natl. Acad. Sci. USA,2006,103:2886-2891.
    114. Xie Z, Askari A. Na+/K+-ATPase as a signal transducer. Eur. J. Biochem,2002, 269:2434-2439.
    115. Cardone R.A, Casavola V, Reshkin S.J. The role of disturbed pH dynamics and the NA+/H+ exchanger in metastasis. Nat. Rev. Cancer,2005,5:786-795.
    116. Aguirre-Ghiso J.A, Estrada Y, Liu D, et al. ERKMAPK activity as a determinant of tumor growth and dormancy; regulation by p38MAPK. Cancer Res,2003,63;1684-1695.
    117. Bradham C, McClay D.R. p38 MAPK in development and cancer. Cell Cycle,2006, 5:824-828.

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700