Activation of the metabotropic glutamate receptor is neuroprotective during nitric oxide
巨噬细胞极化对心肌成纤维细胞活化的影响
天津医药2020年7月第48卷第7期巨噬细胞极化对心肌成纤维细胞活化的影响吴惠娟1,张盛昔1,杨潇2,胡因铭1,王乐旬1△,郭姣1摘要:目的观察巨噬细胞极化上清对心肌成纤维细胞活化的影响。
方法提取SD 大鼠的骨髓细胞和心肌成纤维细胞。
利用巨噬细胞集落刺激因子(M-CSF )处理骨髓细胞后,加入刺激因子:M0(无刺激因子)、M1(100μg/L 脂多糖+10μg/L 干扰素-γ)、M2(20μg/L 白细胞介素-4)诱导巨噬细胞极化。
将极化后的不同型别巨噬细胞及其培养上清分别与心肌成纤维细胞共培养,分别设空白对照组、M0组、M1组和M2组,通过细胞免疫荧光检测心肌成纤维细胞中纤维化蛋白的表达水平;实时荧光定量逆转录聚合酶链反应检测巨噬细胞和成纤维细胞特征分子的表达;Western blot 检测纤维化相关蛋白及转化生长因子β受体(TGFβR )、血小板衍生生长因子受体(PDGFRs )信号通路活化情况。
结果经M-CSF 及相应刺激因子诱导,成功获得M1和M2型巨噬细胞。
细胞共培养结果显示,与M0组相比,M1组上清培养的心肌成纤维细胞中胶原蛋白1(Col1a1)和Col3a1的mRNA 水平以及平滑肌肌动蛋白(α-SMA )表达水平显著降低(P <0.05),而M2组上清培养的心肌成纤维细胞中Col1a1和Col3a1的mRNA 水平以及α-SMA 、结缔组织生长因子(CCN2)表达水平显著升高(P <0.05)。
M1组上清培养的心肌成纤维细胞中PDGFRβ蛋白磷酸化水平显著低于M0组(P <0.01),而M2组上清培养的心肌成纤维细胞中PDGFRβ蛋白磷酸化水平显著高于M0组(P <0.05)。
结论M1型巨噬细胞上清能够抑制心肌成纤维细胞活化,而M2型巨噬细胞上清能够激活心肌成纤维细胞。
M1型巨噬细胞抑制纤维化的作用可能与抑制PDGFRβ通路的活化有关。
关键词:纤维化;心脏;成纤维细胞;巨噬细胞;受体,血小板源生长因子β中图分类号:R392.12文献标志码:ADOI :10.11958/20200169Effects of macrophage polarization on the activation of cardiac fibroblastWU Hui-juan 1,ZHANG Sheng-xi 1,YANG Xiao 2,HU Yin-ming 1,WANG Le-xun 1△,GUO Jiao 11Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine,Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine,Joint Laboratory of Guangdong,Hong Kong and Macao on Glycolipid Metabolic Diseases,Institute of Chinese Medicine Sciences,Guangdong Pharmaceutical University,Guangzhou 510006,China;2Department of Clinical Laboratory,Guangzhou First People's Hospital△Corresponding Author E-mail:*********************Abstract:ObjectiveTo observe the effects of macrophage polarization supernatant on the activation of cardiacfibroblasts.MethodsBone marrow cells and cardiac fibroblasts of SD rats were extracted.Bone marrow cells were inducedto M1and M2by treating with macrophage colony-stimulating factor (M-CSF),and cells were divided into M0group (no stimulating factor),M1group (100μg/L LPS+10μg/L INF-γ)and M2group (20μg/L IL-4).Different macrophages were co-cultured with cardiac fibroblasts,and different macrophage supernatants were collected to culture with cardiac fibroblasts.Immunofluorescence was performed to examine the fibrotic protein expression in cardiac fibroblasts.The mRNA levels of macrophage-specific molecules,fibrosis-related genes and signaling pathways were tested by real-time PCR.The fibrosis-related proteins and the activation of TGFβR and PDGFRs signal pathways were detected by Western blot assay.Results After treatment with M-CSF and stimulating factors,M1macrophages and M2macrophages were pared with the M0group,the mRNA levels of Col1a1and Col3a1and the protein level of α-SMA were significantly decreased in the cardiac fibroblasts treated by the supernatant of M1macrophage group (P <0.05),while the mRNA levels of Col1a1and基金项目:广东省自然科学基金博士启动纵向协同项目(2018A030310403);广东省自然科学基金(2018A0303130168);广东省医学科学技术研究基金(A2018068);广东省基础与应用基础研究基金项目(2020A1515010155)作者单位:1广东省代谢病中西医结合研究中心,广东省代谢性疾病中医药防治重点实验室,粤港澳联合代谢病重点实验室,广东药科大学中医药研究院(邮编510006);2广州市第一人民医院检验科作者简介:吴惠娟(1995),女,硕士在读,主要从事中医药防治糖脂代谢病方面研究△通信作者E-mail :*********************细胞与分子生物学611Tianjin Med J,July2020,Vol.48No.7《中国心血管病报告2018》显示我国心血管病现患人数约为2.9亿,其中90%以上与心脏有关[1]。
细胞信号转导进展G蛋白
GPCR Art
mAchR—A类GPCRs
• Belongs to subfamily A18 – M1 (CHRM1, ACM1) – M2 (CHRM2, ACM2) – M3 (CHRM3, ACM3) – M4 (CHRM4, ACM4) – M5 (CHRM5, ACM5)
• 代谢型离子通道
Linda B. Buck
其他G-蛋白
2.视觉感受器中的G蛋白
• 视紫红质为7次跨膜蛋白,由视蛋白和视黄醛组成。其信号途径为: 光信号→Rh激活→Gt活化→cGMP磷酸二酯酶激活→胞内cGMP减 少→Na+离子通道关闭→离子浓度下降→膜超极化→神经递质释放 减少→视觉反应。
G蛋白介导的信号转导
➢GPCR ➢G蛋白介导的跨膜信号转导 ➢小G蛋白
1. "The Biology of Cancer" , by Robert A. Weinberg, June 2006, (Garland Science Textbooks), 864pp. 2. "One Renegade Cell" (Science Masters) by Robert A. Weinberg, October 01, 1999, 170pp. 3. "Racing to the Beginning of the Road: The Search for the Origin of Cancer" , by: Robert A. Weinberg, May 01, 1996 (Harmony Books) 270pp. 4. "Genes and the Biology of Cancer" (Scientific American Library) by: Harold Varmus, Robert A. Weinberg, October 01, 1992, 215pp. 5. "Oncogenes and the Molecular Origins of Cancer" (Monograph Ser No. 18), March 01, 1990, Cold Spring Harbor (R.A.Weinberg, Editor) 270pp.
LPS诱导小胶质细胞活化的模型建立 (数学建模作业)
摘要
神经退行性疾病是影响人类健康的一类疾病的总称,包括阿尔茨海默病,帕金森病等。小胶质细胞在中枢神经系统的炎症进程中发挥重要作用。小胶质细胞的适度激活对神经元具有保护作用,然而过度激活的小胶质细胞释放大量的神经毒性因子,如一氧化氮(NO),而神经毒性因子会导致神经退行性疾病的发生。因此小胶质细胞的活化是多种神经退行性疾病的根本原因。
关键词:神经退行性疾病小胶质细胞NO优克那非N-乙酰半胱氨酸数学建模
ABSTRACT
Neurodegenerative diseases that affect human health, the general term for a class of diseases, including Alzheimer's disease, Parkinson's disease. Microglia in central nervous system play an important role in the inflammatory process. Moderate microglia activation has a protective effect on neurons, but excessive activation of microglia release large amounts of neurotoxic factors, such as nitric oxide (NO), the neural toxicity factor will lead to neurodegenerative diseases. Therefore, the activation of microglia is a variety of underlying causes of neurodegenerative diseases.
谷氨酸循环及谷氨酸兴奋性毒性
谷氨酸循环及谷氨酸兴奋性毒性众所周知,谷氨酸是中枢神经系统最重要的兴奋性神经递质。
谷氨酸不能通过血脑屏障。
在脑内合成Glu的途径有4条[1]:(1)α-酮戊二酸接受氨基产生Glu;(2)γ-氨基丁酸(γ-amino-bu-tyric acid,GABA)经GABA转氨酶形成Glu;(3)鸟氨酸在鸟氨酸转氨酶的作用下产生谷氨酸半醛,后者进一步生成Glu;(4)谷氨酰胺在谷氨酰胺酶的作用下水解成Glu。
而其中只有第4条途径来源的Glu发挥神经递质的作用。
一.谷氨酸—谷氨酰胺循环神经系统中,神经胶质细胞(主要是星型胶质细胞,AC)与神经元的比例约为10:1。
AC 介于神经元与毛细血管之间,是血脑屏障的重要组成部分。
正常状态下,神经元胞浆的Glu 浓度在10mM/L,AC胞浆的Glu浓度在50至几百μM/L,胞外则为0.6,突触间隙为1μM/L,而突触终端囊泡可达100mM/L,胞内外Glu的浓度相差万倍以上。
突触传递过程中,神经冲动传导至神经突触,神经末梢去极化,突触小泡通过突触囊泡和质膜融合而从神经元释放(即胞吐作用)。
囊泡释放的Glu可使突触间隙的浓度由静息的1μM/L升高到1.1 mM/L,维持在此峰值的时间约为1.2ms。
[2]作用于突触后膜的各型Glu受体,传递神经冲动,发挥生理作用,同时,触发负反馈调节,并由AC膜上的谷氨酸转运体摄取,神经胶质细胞具有很强的Glu摄取能力,并含有谷氨酰胺合成酶,能将Glu转变成谷氨酰胺,再转运至突触前神经末梢胞质中,经谷氨酰胺酶脱氨生成Glu。
同时,一部分经谷氨酸脱羧酶催化生成具有抑制作用的GABA。
接着,Glu通过位于囊泡上的谷氨酸转运体将其转位进入囊泡内腔,并储存于囊泡中。
在静息神经元(resting neuron)中,Glu在神经末梢的突触囊泡内以很小的膜结合细胞器形式储存。
由此形成神经元和胶质细胞之间的“谷氨酸-谷氨酰胺循环”(如图)二.谷氨酸受体GluR分为亲离子型受体和代谢型受体(mGluR)。
银杏达莫注射液联合骨髓间充质干细胞移植改善脑梗死后的神经功能
银杏达莫注射液联合骨髓间充质干细胞移植改善脑梗死后的神经功能杨朝阳【摘要】背景:通过细胞移植重建损伤脑组织成为治疗脑梗死的新途径,骨髓间充质干细胞成为近年来细胞移植治疗领域的研究热点。
<br> 目的:探讨银杏达莫注射液联合骨髓间充质干细胞移植对脑梗死大鼠神经功能的改善作用及相关机制。
<br> 方法:利用线栓法制作大鼠大脑中动脉闭塞模型,建模成功后60只SD大鼠随机分为对照组、细胞移植组及联合组。
对照组尾静脉注射PBS、细胞移植组尾静脉注射2.5×109 L-1的骨髓间充质干细胞悬液、联合组尾静脉注射2.5×109 L-1的骨髓间充质干细胞悬液和银杏达莫2 mL/kg,1次/d,连续注射5 d。
于移植后的1,3 d及1,2周进行mNSS行为学评分,以观察大鼠神经功能缺损状况。
移植后2周RT-PCR检测脑组织中脑源性神经生长因子、生长相关蛋白43基因表达变化,TUNEL法检测细胞凋亡情况,免疫组化法检测BrdU阳性细胞数。
<br> 结果与结论:移植后的1,3 d各组大鼠神经功能缺损评分差异无显著性意义(P >0.05),在移植后1,2周,联合组神经功能缺损评分低于细胞移植组及对照组(P <0.05);移植后2周,联合组脑源性神经生长因子、生长相关蛋白43 mRNA表达明显高于细胞移植组及对照组(P<0.05),联合组凋亡细胞数目明显少于细胞移植组及对照组(P <0.05),联合组BrdU阳性细胞数量明显多于细胞移植组及对照组(P <0.05)。
结果表明骨髓间充质干细胞联合银杏达莫干预能促进脑梗死组织脑源性神经生长因子、生长相关蛋白43 mRNA的表达,抑制细胞凋亡,改善大鼠神经功能。
%BACKGROUND:Reconstruction of damaged brain tissue through cel transplantation has become a new way to treat cerebral infarction. In recent years, bone marrow mesenchymal stem celshave become the new darling in cel transplantation therapy. <br> OBJECTIVE:To investigate the effect of ginkgo-damole injection combined with bone marrow mesenchymal stem cel transplantation to improve the neurological function of acute cerebral infarction rats and its mechanism. <br> METHODS:Animal models of middle cerebral artery occlusion were made in rats using suture method, and then 60 rat models were randomly divided into control group, cel transplantation group and combination group. The control group was given intravenous injection of PBSvia the tail vein; the cel transplantation group was given intravenous injection of bone marrow mesenchymal stem cel suspension (2.5×109/L) via the tail vein; the combination group was given intravenous injection of bone marrow mesenchymal stem cel suspension (2.5×109 /L) and ginkgo-damole injection (2 mL/kg, once a day, totaly 5 days)via the tail vein. Modified neurological severity scores were recorded at 1, 3 days and 1, 2 weeks after transplantation. At 2 weeks after transplantation, expressions of brain-derived neurotrophic factor and growth associated protein 43 in the brain were detected using RT-PCR; cel apoptosis detected using MTT assay; BrdU positive cels counted using <br> immunohistochemistry method.<br> RESULTS AND CONCLUSION:There were no differences in the modified neurologic severity scores among the three groups at 1, 3 days after transplantation (P > 0.05), but the modified neurological severity scores in the combination group were lower than those in the cel transplantation group and control group at 1, 2 weeks after transplantation (P < 0.05). The expressions of brain-derived neurotrophicfactor and growth associated protein 43 in the brain were significantly higher in the combination group than the other two groups at 2 weeks after transplantation (P < 0.05); compared with the other two groups, the number of apoptotic cels was less but the number of BrdU positive cels was higher in the combination group (P < 0.05). These findings indicate that the combination of ginkgo-damole injection and bone marrow mesenchymal stem cel transplantation can increase the expressions of brain-derived neurotrophic factor and growth associated protein 43 in the brain, inhibit cel apoptosis and improve neurological function in rats with cerebral infarction.【期刊名称】《中国组织工程研究》【年(卷),期】2015(000)050【总页数】6页(P8108-8113)【关键词】干细胞;移植;银杏达莫注射液;骨髓间充质干细胞;干细胞移植;脑源性神经生长因子;GAP-43;脑梗死【作者】杨朝阳【作者单位】济源市人民医院普内科,河南省济源市 454000【正文语种】中文【中图分类】R394.2文章亮点:1“干细胞循环”理论与中医理论中的“活血化瘀”法与有相通之处,银杏达莫注射液是从中药银杏叶中提取的复方制剂,能清除自由基、改善血液循环。
24-nervous system-2-协和基础医学院
Ligand of receptor
Agonist:
A substance that mimics a specific neurotransmitter, is able to attach to that neurotransmitter's receptor & thereby produces the same action that the neurotransmitter usually produces
Preganglionic neurons of autonomic NS Postganglionic neurons of parasympathetic NS Few postganglionic neurons of sympathetic NS
Central Nervous System (CNS) -- Widespread
G Protein-Coupled Receptor
(Second Messenger System)
1st Messenger: Neurotransmitter
2nd Messenger:
cAMP/cGMP IP3 DAG Ca2+
Slow & Prolonged Reaction
Otto Loewi
"for their discoveries of Ach as neurotransmitter”
Catecholamines
Dopamine Norepinerphrine Epinephrine
Synthesis
CNS CNS & PNS
CNS
Dopamine (DA)
Some substances e.g. CO & NO act as neurotransmitter
线粒体自噬的英语
线粒体自噬的英语Mitochondrial Autophagy: A Vital Process for Cellular HomeostasisMitochondria, often referred to as the "powerhouses" of cells, play a crucial role in cellular metabolism and energy production. These organelles are responsible for generating the majority of the cell's supply of adenosine triphosphate (ATP), the primary energy currency of the cell. However, mitochondria are not just passive energy producers; they are dynamic structures that undergo constant remodeling and maintenance to ensure optimal function. One of the key mechanisms involved in this process is mitochondrial autophagy, also known as mitophagy.Autophagy is a fundamental cellular process in which damaged or unwanted components are engulfed and degraded within the cell. This process serves as a quality control mechanism, removing dysfunctional organelles, misfolded proteins, and other cellular debris to maintain cellular homeostasis. Mitophagy, a specialized form of autophagy, specifically targets and removes damaged or dysfunctional mitochondria, ensuring the overall health and efficiency of the cellular energy production system.The importance of mitophagy cannot be overstated. Impaired mitophagy has been linked to a variety of disease states, including neurodegenerative disorders, cardiovascular diseases, and metabolic disorders. When mitochondria become damaged or dysfunctional, they can release reactive oxygen species (ROS) and pro-apoptotic factors, leading to cellular stress and potentially triggering programmed cell death (apoptosis). Mitophagy serves as a protective mechanism, selectively removing these damaged mitochondria and preventing the propagation of cellular damage.The process of mitophagy is a highly regulated and complex event, involving a series of coordinated steps. The initial step involves the identification of damaged or dysfunctional mitochondria. This is typically achieved through the detection of specific molecular signals, such as the loss of membrane potential or the accumulation of misfolded proteins within the mitochondria. These signals trigger the recruitment of specialized proteins, known as mitophagy receptors, which act as the "tags" that mark the damaged mitochondria for removal.Once the mitochondria have been identified, the next step is the formation of the autophagosome, a double-membrane vesicle that engulfs the targeted mitochondria. This process is facilitated by a group of proteins known as the autophagy-related (Atg) proteins, which coordinate the assembly and maturation of theautophagosome. The autophagosome then fuses with the lysosome, an organelle rich in digestive enzymes, resulting in the degradation of the mitochondrial contents.The regulation of mitophagy is a delicate balance, as the process must be precisely controlled to ensure the appropriate removal of damaged mitochondria without compromising the overall cellular function. This regulation is achieved through a complex network of signaling pathways and transcriptional programs that respond to various cellular cues, such as oxidative stress, nutrient availability, and energy status.One of the key regulators of mitophagy is the PINK1/Parkin pathway, which has been extensively studied in the context of Parkinson's disease. In this pathway, the PINK1 protein acts as a sensor, detecting the loss of mitochondrial membrane potential and recruiting the E3 ubiquitin ligase Parkin to the damaged mitochondria. Parkin then ubiquitinates specific mitochondrial proteins, marking them for degradation and triggering the mitophagy process.In addition to the PINK1/Parkin pathway, other signaling cascades, such as the AMPK (AMP-activated protein kinase) and mTOR (mechanistic target of rapamycin) pathways, also play crucial roles in the regulation of mitophagy. These pathways respond to changes incellular energy status and nutrient availability, respectively, and modulate the activity of mitophagy-related proteins to maintain cellular homeostasis.The importance of mitophagy extends beyond its role in maintaining cellular health. Emerging evidence suggests that mitophagy may also be involved in various physiological processes, such as development, aging, and adaptation to environmental stressors. For instance, during embryonic development, mitophagy is crucial for the elimination of paternal mitochondria, ensuring the exclusive inheritance of maternal mitochondrial DNA.Furthermore, the dysregulation of mitophagy has been implicated in the pathogenesis of various age-related diseases, including neurodegenerative disorders, cardiovascular diseases, and cancer. Understanding the mechanisms underlying mitophagy and its role in these disease states has become a major focus of research in the field of cellular and molecular biology.In conclusion, mitochondrial autophagy, or mitophagy, is a vital process that ensures the proper maintenance and function of mitochondria within the cell. By selectively removing damaged or dysfunctional mitochondria, mitophagy plays a crucial role in maintaining cellular homeostasis and preventing the propagation of cellular damage. The regulation of mitophagy is a complex anddynamic process, involving a network of signaling pathways and transcriptional programs that respond to various cellular cues. Ongoing research in this field continues to shed light on the importance of mitophagy in both physiological and pathological conditions, paving the way for the development of potential therapeutic interventions targeting this crucial cellular process.。
JNK-pathway
Review:MAPKs (Mitogen-Activated Protein Kinases) are Serine-threonine protein Kinases that are activated in response to a variety of extracellular stimuli and mediate signal transduction from the cell surface to the nucleus. MAPKs are expressed in multiple cell types including Cardiomyocytes, Vascular Endothelial cells, and Vascular Smooth Muscle Cells. Three major MAPKs include ERKs (Extracellular signal-Regulated Kinases), JNKs (c-Jun NH(2)-terminal protein Kinases), and p38 Kinases. Members of the JNK/SAPK (Stress-Activated Protein Kinase) family of MAPKs are strongly stimulated by numerous Environmental Stresses, but also more modestly stimulated by Mitogens, Inflammatory Cytokines, Oncogenes, and inducers of Cell differentiation and morphogenesis. Ten mammalian JNK isoforms have been identified and are encoded by three distinct genes, JNK1, JNK2, and JNK3, the transcripts of which are alternatively spliced to yield four JNK1 isoforms, four JNK2 isoforms, and two JNK3 isoforms. JNK1 and JNK2 are the products of alternative splicing of a single gene and are expressed in many tissues, but JNK3 is specifically expressed in brain. Members of the JNK family play crucial roles in regulatingresponses to various Stresses, and in Neural Development, Inflammation, and Apoptosis. JNK activation is much more complex than that of ERK1/ERK2 owing to inputs by a greater number of MAPKKKs (Mitogen-Activated Protein Kinase Kinase Kinases) (at least 13, including MEKK1 (MAP/ERK Kinase-Kinase-1)-MEKK4 (MAP/ERK Kinase-Kinase-4), ASK (Apoptosis Signal-regulating Kinase) and MLKs (Mixed-Lineage Kinases), which are activated by upstream Rho-family GTPases). These activate JNK MAPKKs MEK4 (MAPK/ERK Kinase-4) and MEK7 (MAPK/ERK Kinase-7), which further activate JNKs. The JNK MAPK modules are regulated by a number of different scaffold proteins, including JIP1 (JNK Interacting Protein-1), JIP2 (JNK Interacting Protein-2), JIP3 (JNK Interacting Protein-3), JIP4 (JNK Interacting Protein-4), Beta-Arrestin-2, Filamin and CrkII. The scaffold proteins presumably target the MAPK modules to different sites in the cell and play roles in kinase activation and/or substrate selection (Ref.1 & 2).Stress or Genotoxic agents are the most powerful inducers of JNK. Different forms of stress have been shown to mediate JNK activation via various cellular pathways. JNK activation in response to UV irradiation is mediated by upstream signaling components, including Rac (Ras-Related C3 Botulinum Toxin Substrate), CDC42 (Cell Division Cycle-42), PAK (p21/CDC42/Rac1-Activated Kinase), ASK1 (Apoptosis Signal-regulating Kinase-1), MLK, MEKK1, SEK1 (SAPK/ERK Kinase-1)/MKK4, MKK7 and p21Ras, in concert with nuclear DNA lesions. Besides Stress, JNKs can also be activated via GPCRs (G-Protein Coupled Receptors), RTKs (Receptor Tyrosine Kinases) and Cytokine Receptors. How GPCRs activate the JNKs is still an unanswered question. Free Beta-Gamma dimers and GN-Alpha12 and GN-Alpha13 proteins are able to activate JNK in a Rac1-CDC42 or p115RhoGEF and RhoA-dependent manner. However, the nature of the GEFs (Guanine nucleotide Exchange Factors) that connect Beta-Gamma and GN-Alpha12/ GN-Alpha13 to Rac1 and CDC2 is still unclear. Interestingly, GN-Alpha12 can also activate JNK by activating the MEKK (MEK kinase). The activation of JNK by Cytokine receptors appears to be mediated by the TRAF (TNF Receptor-Associated Factor) group of Adaptor proteins. Activation of the TNF receptor leads to recruitment of TRAF2 (TNF Receptor-Associated Factor-2), which is required for JNK activation. These adaptor proteins (TRADD (Tumor Necrosis Factor Receptor-1-Associated Death Domain Protein), RIP (Receptor-Interacting Protein) and Daxx) have been reported to bind MEKK1 and ASK1. TRAF2 activates MAPK4Ks like GCK (Germinal Center Kinase), GCKR (GCK-Related Kinase), GLK (GCK-Like Kinase) and HGK (HPK/GCK-like Kinase), which further activates JNKs via MEKK1 and MKK4/7 respectively. ASK1 also interacts with TRAF2 and activates JNK via MKK4/7 (Ref. 3, 4 & 5).Growth Factors also activate JNKs. Although the Signaling cascade from Growth Factor Receptors to ERKs is relatively well understood, the pathway leading to JNK activation is more obscure. Activation of JNK by EGF (Epidermal Growth Factor) or NGF (Nerve Growth Factor) is dependent on H-Ras activation. Growth Factors and Growth Factor Receptors stimulate Ras by recruiting SOS (Son of Sevenless), GRB2 (Growth Factor Receptor-Bound Protein-2) and SHC to the membrane. PI3K (Phosphatidylinositde-3-Kinase) also activate Ras. Ras activates two protein kinases, Raf1 and MEKK (MEK (MAPK, or ERK, kinase) Kinase). Raf1 contributes directly to ERK activation but not to JNK activation, whereas MEKK participated in JNK activation but caused ERK activation only after overexpression. Recently, Raf1 is found to interact with the proapoptotic, stress-activated protein kinase ASK1 in vitro and in vivo. This interaction allows Raf1 to act independently of the MEK–ERK pathway to activate JNK pathway (Ref.6 & 7). The Rho family GTPases, CDC42 (Cell Division Cycle-42) and Rac also initiate a cascade leading to JNK/SAPK,presumably by binding and activating the protein kinase PAK (p21-Activated Kinases), a kinase that phosphorylates and promotes activation of MEKK1. Rac/CDC42 are also involved in JNK activation via a pathway consisting of a sequential cascade MLKs and MKK4/7 (MAP Kinase Kinase-4/7. MLK2 (Mixed-Lineage Kinase-2) and MLK3 (Mixed-Lineage Kinase-3) interact with the activated (GTP-bound) forms of Rac and CDC42, with a slight preference for Rac. Besides MLKs, MEKK1/4 and Posh (Plenty of SH3) are also activated by Rac/CDC42 to activate MKK4/7 and thus JNKs. Adaptor proteins such as Crk (v-Crk Avian Sarcoma Virus Ct10 Oncogene Homolog) and CrkL (v-Crk Avian Sarcoma Virus Ct10 Oncogene Homolog-Like) also leads to activation of JNKs in response to RTK. HPK1 (Hematopoietic Progenitor Kinase-1) associates with Crk and CrkL through binding to the SH3 (Src-Homology Domain-3) of these proteins. Furthermore, association of HPK1 with these proteins increases HPK1's kinase activity. HPK1 then act as upstream of MEKK1 and TAK1 (Transforming Growth Factor-Beta-activated Kinase-1) in the JNK kinase cascade. JNKs are negatively regulated by MKP (MAP Kinase Phosphatase) (Ref.2, 8 & 9).The activated JNK/SAPKs translocate to the nucleus where they phosphorylate transcription factors such as c-Jun, Elk1, DPC4 (Deleted In Pancreatic Carcinoma 4)/ SMAD4 (Sma and MAD (Mothers Against Decapentaplegic) Related Protein-4), p53, ATF2 (Activating T ranscription Factor-2), NFAT4 (Nuclear Factor of Activated T-Cell-4) and NFAT1 (Nuclear Factor of Activated T-Cell-1). JNK1 directly phosphorylates Bcl2 (B-Cell CLL/Lymphoma-2) in vitro, co-localizes and collaborates with Bcl2 to mediate prolonged cell survival. JNK cascade also activates TCF (Ternary Complex Factor) protein. JNK also phosphorylate HSF1 (Heat Shock Factor-1) and JNK-mediated phosphorylation of HSF1 selectively stabilize the HSF1 protein and confers protection to cells under conditions of severe stress. DCX is also a substrate of JNK and interacts with both JNK and JIP. MAPs (Microtubule-Associated Proteins), both MAP1B and MAP2B are also found to be the substrates of JNK. Ser-727 phosphorylation of STAT3 (Signal Transducer and Activator of Transcription-3) can also be induced by JNK. JNK also regulates Insulin signaling by negatively regulating IRS1 (Insulin Receptor Substrate-1). JNK is generally thought to be involved in inflammation, proliferation and Apoptosis. Accordingly, its substrates are transcription factors and Anti-apoptotic proteins. However, JNK also phosphorylates Serine 178 on Paxillin and regulate cell migration. Despite extensive progress in the understanding of the JNK MAP kinase pathway, the mechanisms by which the pathway contributes to the many cellular programs where JNKs are activated are poorly defined. The JIP1 proteins have been proposed to act as molecular scaffolds that organize the JNK signal transduction pathway in response to specific stimuli. The JNK stress pathway is thought to be important in many pathological conditions including the progression of some neurodegenerative diseases su ch as Huntington’s and also in cancer. This pathway therefore offers potential targets for therapeutic intervention. The identification of critical components of this signaling pathway, such as JIP1, offers new routes to understand how this pathway is regulated and potential ways of manipulating it to combat disease (Ref.10, 11 & 12).References:1. Himes SR, Sester DP, Ravasi T, Cronau SL, Sasmono T, Hume DA.The JNK are important for development and survival of macrophages.J Immunol. 2006 Feb 15;176(4):2219-28.PubMed ID: 164559782. Moulin N, Widmann C.Islet-brain (IB)/JNK-interacting proteins (JIPs): future targets for the treatment of neurodegenerative diseases?Curr Neurovasc Res. 2004 Apr;1(2):111-27.PubMed ID: 161851883. Zhou JY, Liu Y, Wu GS.The role of mitogen-activated protein kinase phosphatase-1 in oxidative damage-induced cell death.Cancer Res. 2006 May 1;66(9):4888-94.PubMed ID: 166514454. Yang L, Mao L, Chen H, Catavsan M, Kozinn J, Arora A, Liu X, Wang JQ.A signaling mechanism from G alpha q-protein-coupled metabotropic glutamate receptors to gene expression: role of the c-Jun N-terminal kinase pathway.J Neurosci. 2006 Jan 18;26(3):971-80.PubMed ID: 164213175. Yang Q, Kim YS, Lin Y, Lewis J, Neckers L, Liu ZG.Tumour necrosis factor receptor 1 mediates endoplasmic reticulum stress-induced activation of the MAP kinase JNK.EMBO Rep. 2006 May 5;PubMed ID: 166800936. Kraus S, Benard O, Naor Z, Seger R.c-Src is activated by the epidermal growth factor receptor in a pathway that mediates JNK and ERK activation by gonadotropin-releasing hormone in COS7 cells.J Biol Chem. 2003 Aug 29;278(35):32618-30.PubMed ID: 127503727. Matsukawa J, Matsuzawa A, T akeda K, Ichijo H.The ASK1-MAP kinase cascades in mammalian stress response.J Biochem (Tokyo). 2004 Sep;136(3):261-5.PubMed ID: 155988808. Yamauchi J, Miyamoto Y, Kokubu H, Nishii H, Okamoto M, Sugawara Y, Hirasawa A, Tsujimoto G, Itoh H.Endothelin suppresses cell migration via the JNK signaling pathway in a manner depen dent upon Src kinase, Rac1, and Cdc42.FEBS Lett. 2002 Sep 11;527(1-3):284-8.PubMed ID: 122206759. Zhou JY, Liu Y, Wu GS.The role of mitogen-activated protein kinase phosphatase-1 in oxidative damage-induced cell death.Cancer Res. 2006 May 1;66(9):4888-94.PubMed ID: 1665144510. Baan B, van Dam H, van der Zon GC, Maassen JA, Ouwens DM.The role of JNK, p38 and ERK MAP-kinases in insulin-induced Thr69 and Thr71-phosphorylation of transcription factor ATF2.Mol Endocrinol. 2006 Apr 6;PubMed ID: 1660107111. Sprowles A, Robinson D, Wu YM, Kung HJ, Wisdom R.c-Jun controls the efficiency of MAP kinase signaling by transcriptional repression of MAP kinase phosphatases.Exp Cell Res. 2005 Aug 15;308(2):459-68.PubMed ID: 1595021712. Heasley LE, Han SY.JNK Regulation of Oncogenesis.Mol Cells. 2006 Apr 30;21(2):167-73.PubMed ID: 16682809。
glutamate词根 -回复
glutamate词根-回复Glutamate is a crucial neurotransmitter that plays a vital role in various physiological processes in the human body. It is derived from glutamic acid, an amino acid that is found abundantly in many foods. Glutamate is involved in neuronal communication, learning, memory, and several other essential functions in the central nervous system. Understanding the structure, function, and regulation of glutamate is crucial for comprehending its significance and implications in various health conditions and diseases.Glutamate is primarily synthesized through a process called glutaminolysis. In this process, glutamine, another amino acid, is converted into glutamate by the enzyme glutaminase. This conversion occurs mainly in cells such as neurons, astrocytes, and microglia, which are key players in maintaining brain function. Glutamate is then released into the synaptic cleft, where it binds to postsynaptic receptors, initiating the process of neuronal communication.Glutamate acts on three major receptor types: NMDA(N-methyl-D-aspartate) receptors, AMPA(alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) receptors, and metabotropic glutamate receptors (mGluRs). The activation of these receptors leads to various intracellular signaling pathways, resulting in neuronal excitability, synaptic plasticity, and ultimately, cognitive functions such as learning and memory.However, glutamate signaling must be regulated to maintain normal brain function. An imbalance in glutamate levels can lead to excitotoxicity, a process in which excessive glutamate causes neuronal damage and cell death. This phenomenon is implicated in several neurodegenerative disorders, such as Alzheimer's disease, Parkinson's disease, and stroke. Glutamate excitotoxicity occurs when there is an overactivation of receptors or a disruption in glutamate transporters, leading to an excessive buildup of glutamate in the synaptic cleft.To regulate glutamate levels, several mechanisms come into play. One critical mechanism is the reuptake of glutamate by transporters, primarily located on astrocytes. These transporters, such as excitatory amino acid transporters (EAATs), actively remove extracellular glutamate, preventing its accumulation and ensuring optimum neurotransmission. Dysfunction or impairment of thesetransporters can lead to glutamate dyshomeostasis and subsequent excitotoxicity.Moreover, glutamate can be converted back into glutamine through a process called the glutamate-glutamine cycle. In this cycle, astrocytes take up glutamate from the synaptic space and convert it into glutamine using the enzyme glutamine synthetase. Glutamine is then transported back to neurons, where it is converted back into glutamate. This cycle ensures a constant supply of glutamate and maintains neurotransmitter balance.Furthermore, glutamate signaling is tightly regulated by various other factors. For example, inhibitory neurotransmitters such as GABA (gamma-aminobutyric acid) counterbalance the excitatory effects of glutamate by inhibiting neuronal activity. Additionally, numerous modulatory systems, including those involving dopamine, serotonin, and norepinephrine, interact with glutamate signaling to fine-tune neuronal responses.Dysregulation of glutamate signaling has been implicated in the pathophysiology of various neurological and psychiatric disorders. For instance, abnormalities in glutamate transmission have beenobserved in schizophrenia, major depressive disorder, and addiction. Delineating the mechanisms underlying glutamate dysregulation may lead to the development of targeted therapeutic interventions for these conditions.In conclusion, understanding the role of glutamate in neuronal communication and its regulation is crucial for comprehending various physiological and pathological processes in the brain. From its synthesis to its receptors and regulation, glutamate is involved in a wide range of functions, including learning, memory, and cognition. Dysregulation of glutamate signaling can have profound implications for brain health, contributing to neurodegenerative disorders and psychiatric conditions. Further research into glutamate and its intricate mechanisms will undoubtedly continue to shed light on its significance and potential therapeutic interventions for related disorders.。
ampk激活通过调控氧化应激修复高糖诱导的人脑微血管内皮细胞损伤
中国组织化学与细胞化学杂志CHINESE JOURNAL OF HISTOCHEMISTRY AND CYTOCHEMISTRY第29卷第1期2020年02月V ol .29.No .1February .2020〔收稿日期〕2019-08-10 〔修回日期〕2020-02-10〔基金项目〕国家自然科学基金面上项目(81671534,81070514)〔作者简介〕杨梦珍,女(1994年),汉族,硕士生*通讯作者(To whom correspondence should be addressed):lin.wang@AMPK 激活通过调控氧化应激修复高糖诱导的人脑微血管内皮细胞损伤杨梦珍,汪琳*(武汉大学基础医学院组织学与胚胎学系,武汉430071)〔摘要〕目的 探究高糖条件下,腺苷酸活化蛋白激酶(adenine monophosphate activated protein kinase, AMPK )调控的氧化应激对血脑屏障通透性的影响。
方法 采用人脑微血管内皮细胞(human brain microvascular endothelial cells, HBMEC )作为体外血脑屏障细胞模型,采取如下两种方式处理:①不同浓度(5.5mmol/L 或27.5mmol/L )葡萄糖处理HBMEC 24h ;②AMPK 激动剂氨基咪唑-4-甲酰胺核苷酸(4-amino-1H-imida-zole-5-carboxamide, AICAR )预处理2h 后,再给予27.5mmol/L 葡萄糖处理24h 。
Western blot 及免疫荧光染色检测细胞间血脑屏障相关的闭锁连接蛋白(zonula occludens protein 1, ZO-1)水平,活性氧检测试剂盒检测细胞内活性氧( reactive oxygen species, ROS)水平,透射电镜观察细胞内线粒体超微结构改变。
结果 高糖处理后HBMEC 细胞ZO-1水平显著降低,ROS 水平升高,线粒体结构损伤;AICAR 预处理可显著逆转高糖处理引起的HBMEC 细胞中 ZO-1水平降低、ROS 水平升高和线粒体结构损伤。
铁死亡与自噬在疾病中的研究进展
㊀㊀ʌ摘㊀要ɔ㊀铁死亡是一种铁依赖的㊁以谷胱甘肽过氧化物酶4活性丧失㊁脂质过氧化物沉积为特点的细胞死亡方式ꎮ自噬是一种高度保守的㊁用于降解和回收利用生物大分子或受损细胞器的过程ꎮ当自噬过度激活时ꎬ也会引起细胞的自噬性死亡ꎮ文章就铁死亡与自噬的相互关系及其在神经系统疾病㊁循环系统疾病和肿瘤中的研究进展作一综述ꎬ以期加深对铁死亡及自噬关系的认识ꎮʌ关键词ɔ㊀铁死亡ꎻ自噬ꎻ相互关系ꎻ铁离子ʌDOIɔ㊀10.3969/j.issn.1671 ̄6450.2019.12.023Researchprogressofirondeathandautophagyindiseases㊀PENGXiaꎬFANGCongcongꎬXUYuming.DepartmentofPediatricsꎬRenminHospitalofWuhanUniversityꎬHubeiProvinceꎬWuhan430060ꎬChinaCorrespondingauthor:YAOBaozhenꎬE ̄mail:professoryao@aliyun.com㊀㊀ʌAbstractɔ㊀Irondeathisaniron ̄dependentcelldeathmodecharacterizedbylossofglutathioneperoxidase4activityandlipidperoxidedeposition.Autophagyisahighlyconservativeprocessusedtodegradeandrecyclebiologicalmacromole ̄culesordamagedorganelles.Whenautophagyisoveractivatedꎬautophagicdeathofcellsisalsocaused.Thisarticlereviewstherelationshipbetweenirondeathandautophagyanditsresearchprogressinnervoussystemdiseasesꎬcirculatorysystemdis ̄easesandtumorsinordertodeepentheunderstandingoftherelationshipbetweenirondeathandautophagy.㊀㊀ʌKeywordsɔ㊀FerroptosisꎻAutophagyꎻCorrelationꎻIronion㊀㊀早在铁死亡被命名之前ꎬ研究者们就已经发现了erastin㊁RSL3可以诱导细胞以一种不同于凋亡的方式死亡ꎬ且这种细胞死亡方式可以被铁螯合剂及抗氧化剂抑制ꎬ从而明确了这种细胞死亡方式与细胞内铁和活性氧(ROS)有关ꎮ2012年ꎬDixon等[1]发现这种细胞死亡方式在形态学㊁生化和遗传学上均与凋亡㊁坏死及自噬不同ꎮ由于这种细胞死亡方式特异地依赖于细胞内铁ꎬ故将其命名为铁死亡ꎮ铁死亡的机制尚未完全阐明ꎬ研究发现铁死亡存在复杂的细胞内调控机制ꎬ铁代谢㊁氨基酸代谢及脂质代谢都参与其中ꎮ自噬广泛存在于真核生物中ꎬ是依赖于细胞内溶酶体分解衰老或损伤的大分子或细胞器的过程ꎬ从而维持细胞内稳态[2]ꎮ然而ꎬ当细胞面对各种不利因素刺激时ꎬ自噬会过度激活ꎮ过度激活的自噬可以导致细胞发生自噬性死亡[3]ꎮ铁死亡和自噬作为2种不同的生物过程ꎬ在细胞内发挥着各自重要的作用ꎮ在铁死亡发现之初ꎬ研究者们认为这是一种与自噬截然不同的细胞死亡方式ꎮ近年来ꎬ虽然仍然存在很多争议ꎬ但是越来越多的研究在逐渐揭示铁死亡与自噬的关系ꎬ并在不同的疾病中证实ꎮ1㊀铁死亡的发生机制1.1㊀细胞内铁与铁死亡㊀铁作为生命必需的微量元素之一ꎬ主要以二价和三价铁离子的形式存在于机体内ꎮ正常情况下ꎬ小肠吸收或红细胞降解释放出的亚铁离子(Fe2+)被氧化为三价铁离子(Fe3+)ꎬFe3+经膜上转铁蛋白(transferrinꎬTF)㊁膜蛋白转铁蛋白受体1(transferrinreceptorprotein1ꎬTFR1)的作用后内吞入胞体ꎮ内吞入细胞内的Fe3+被还原成亚铁离子Fe2+ꎬ然后由二价金属离子转运蛋白1(divalentmetaltrans ̄porter1ꎬDMT1)或锌铁调控蛋白家族8/14(ZRT/IRT ̄likeproteins8/14ꎬZIP8/14)介导Fe2+储存到细胞内不稳定的铁池(labileironpoolꎬLIP)中和铁蛋白轻链多肽(ferritinlightchainꎬFTL)与铁蛋白重链多肽1(ferritinheavychain1ꎬFTH1)组成的铁储存蛋白复合物中ꎮ剩余部分亚铁离子将被氧化成Fe3+出胞ꎬ参与体内铁再循环ꎬ严格把控细胞内铁稳态ꎮ㊀㊀铁参与了生物体很多重要的功能ꎬ包括新陈代谢㊁氧转运㊁抗氧化反应及DNA合成等一系列生物过程ꎮFe ̄S是线粒体电子传递链中氧化还原酶类的重要辅助因子ꎬ如NADH㊁辅酶Q等ꎮ活性氧(reactiveoxygenspeciesꎬROS)的产生与铁稳态的扰动密切相关ꎬ而铁硫团簇(iron ̄sulfurclusterꎬISC)机械系统功能障碍明显加剧了ROS的产生[4]ꎮ然而ꎬ细胞内过多的铁通过芬顿反应产生羟自由基ꎬ从而促进不饱和脂肪酸(PU ̄FAs)的羟化ꎬ由此产生的脂质过氧化物和氢过氧化物严重影响细胞膜的结构和功能[5]ꎬ这个过程就是铁死亡ꎮ铁死亡过程可以被铁螯合剂阻断ꎬ说明细胞内铁与铁死亡密切相关[6]ꎮ而这些有毒的脂质过氧化物ꎬ在谷胱甘肽和谷胱甘肽过氧化物酶4(GPX4)的作用下才可以转变为为无毒的醇类物质ꎬ从而避免其对细胞的杀伤作用[7]ꎮ㊀㊀虽然细胞内铁与铁死亡密切相关ꎬ但是细胞内铁在铁死亡中的具体作用机制至今仍不明确ꎮDixon等[8]认为铁螯合剂抑制铁死亡最有可能的解释是阻止了铁向氧化物传递电子ꎬ从而抑制活性氧的生成ꎮ也有报道提出ꎬ铁螯合剂能够直接作用于含铁离子的酶ꎬ其中以脂氧合酶的可能性最大[9]ꎮ1.2㊀XC ̄系统与铁死亡㊀胱氨酸 谷氨酸反向转运系统(XC ̄系统)是一个广泛分布于磷脂双分子层的氨基酸逆向转运体ꎬ由轻链xCT(SLC7A11)和重链4F2(SLC3A2)组成[10]ꎮ通过XC ̄系统ꎬ胱氨酸与谷氨酸(GLu)以1ʒ1的比例在细胞内外进行交换ꎮ在细胞内ꎬ胱氨酸被谷胱甘肽(GSH)或硫氧还蛋白还原酶1还原为半胱氨酸ꎬ在γ ̄谷氨酰半胱氨酸合成酶和谷胱甘肽合成酶的作用下进一步合成GSH[11]ꎮ细胞摄取半胱氨酸是谷胱甘肽合成的关键步骤ꎬGSH的产生和维持对保护细胞免受氧化应激反应所造成的损伤至关重要ꎬ而半胱氨酸的含量需要转硫途径的调节[12 ̄13]ꎮKang等[14]发现高浓度Glu孵育神经细胞可以抑制XC ̄系统ꎬ从而抑制胱氨酸的摄入ꎬ引起细胞内谷胱甘肽减少和ROS的聚集ꎬ而抑制铁死亡可以抑制Glu引起的神经元死亡ꎮ这些研究都表明XC ̄系统参与了铁死亡过程ꎮ当XC ̄系统被抑制时ꎬ胱氨酸不能转入细胞内ꎬ谷胱甘肽合成减少ꎬ不能将有毒的脂质过氧化物还原成无毒的醇类物质ꎬ进而诱导了铁死亡的发生ꎮ1.3㊀GPX4与铁死亡㊀抑制XC ̄系统导致GSH减少和活性氧的聚集ꎮ当XC ̄系统功能正常且GSH正常合成时ꎬGSH也必须在GPX4的作用下将有毒的脂质过氧化物还原成无毒的醇类物质ꎮGPX4是铁死亡关键的调节因子ꎬ在预防脂质过氧化中起着至关重要的作用[15]ꎮ研究发现ꎬGPX4是唯一一种能够降低生物膜内脂质过氧化氢的酶ꎬ所以当GPX4功能受限时ꎬGSH并未耗竭ꎬ但脂质ROS明显升高ꎻ其次ꎬGSH是GPX4活性的一个必要的辅助因子[16]ꎮGPX4被抑制时将导致脂质ROS的形成及脂质过氧化ꎬ最后诱导铁死亡的发生[17]ꎮ1.4㊀脂质过氧化物与铁死亡㊀不饱和脂肪酸在氧存在情况下很容易发生脂质过氧化ꎬ这种过氧化反应在铁的存在下会加剧[18]ꎬ其中与铁死亡密切相关的不饱和脂肪酸包括花生四烯酸和肾上腺酸ꎮ酯酰基辅酶A合成酶长链家族成员4(AcylCoAsynthetaselong ̄chainfamilymember4ꎬACSL4)和脂质重塑相关的溶血卵磷脂酰基转移酶3(lysophosphatidyl ̄cholineacyl ̄transferase3ꎬLPCAT3)是参与脂质过氧化物形成的2种关键酶ꎬ在它们的作用下ꎬ不饱和脂肪酸转化成脂质过氧化物[19 ̄20]ꎮ脂质过氧化的积累会不可避免地会造成很大的损伤ꎮ丙二醛(MDA)是活性氧作用于生物膜不饱和脂肪酸而产生的脂质过氧化反应的最终产物ꎮ其积累可引起蛋白质与核酸的交联聚合ꎬ导致膜结构的不可逆破坏ꎬ最终导致细胞死亡[21]ꎮ2㊀自噬的过程㊀㊀相较于铁死亡ꎬ自噬的研究则更深入ꎮ目前研究认为ꎬ自噬主要有3种形式:(1)巨自噬ꎬ细胞内损坏的蛋白质㊁细胞器及胞内病原体等被细胞质产生的膜结构包裹ꎬ形成自噬体ꎬ最终与溶酶体融合后被降解ꎻ(2)微自噬ꎬ被降解物直接被溶酶体通过变形运动进行内吞ꎬ使其降解ꎬ这个过程不形成自噬体ꎻ(3)分子伴侣介导的自噬ꎬ待降解物需要与分子伴侣结合ꎬ然后被溶酶体上的溶酶体相关膜蛋白(lysosomalassociatedmembraneproteinꎬLAMPs)识别ꎬ并最终被溶酶体降解[22]ꎮ目前研究较多的自噬类型为巨自噬(以下简称为 自噬 )ꎮ自噬是一个复杂的过程ꎬ具体包括4个步骤:自噬的诱发ꎻ隔膜的延伸㊁闭合ꎬ形成自噬体ꎻ自噬体与溶酶体结合ꎬ形成自噬溶酶体ꎻ自噬体和内部物质的降解[23]ꎮ㊀㊀随着对自噬研究的深入ꎬ人们发现自噬也是有选择性的ꎮ细胞内存在这一些特殊的自噬ꎬ它们在某些特定的条件下对某种大分子或者细胞器进行特定的降解[24]ꎬ如线粒体自噬㊁内质网自噬㊁过氧化物酶体自噬㊁核糖体自噬和脂类自噬ꎮ3㊀铁死亡与自噬的关系㊀㊀铁死亡在发现之初ꎬ人们认为它是一种在生化㊁形态及基因水平与凋亡㊁坏死及自噬不同的细胞死亡途径[25]ꎬ然而随着研究的不断推进ꎬ越来越多的证据表明铁死亡的发生需要自噬机制的参与[26]ꎮHou等[27]通过在永生化小鼠胚胎成纤维细胞(MEFs)㊁人胰腺癌细胞系(PANC1和PANC2.03)和人纤维肉瘤细胞系HT ̄1080中敲低ATG5和ATG7抑制自噬后发现细胞内游离铁水平和脂质过氧化终产物(如MDA)水平均显著下降ꎬ而细胞内稳定铁蛋白标志物FTH1的表达显著上升ꎬ该研究首次从基因层面揭示了自噬和铁死亡关系ꎮZhou等[28]通过实验进一步证实了这种关系ꎬ发现这种特殊的自噬过程是以铁蛋白为底物的ꎮ之后ꎬ许多研究证实与铁死亡密切相关的大分子物质参与自噬的发生过程中ꎮGSH㊁GPX4及脂质过氧化物都是铁死亡过程中关键的大分子物质ꎬ研究表明在饥饿和氧化应激等条件下ꎬ自噬发生时伴随着GSH的下降[29]ꎻ而GPX4过表达可抑制ROS介导的自噬的发生ꎬ脂质过氧化物可以促进自噬体的形成[30 ̄31]ꎮ核受体辅激活因子4(NCOA4)是铁自噬过程中的选择性受体[32]ꎬ而下调NCOA4可以抑制erastin诱导的铁死亡ꎮ脂质自噬介导的脂滴降解可以促进铁死亡的发生[33]ꎮ其他如SLC7A11㊁NRF2㊁p53㊁HSPB1和ACSL4等铁死亡调控因子已经被证实是自噬的潜在调控因子ꎮ目前越来越多的研究证实过度的自噬可以促进铁死亡ꎮ然而ꎬ也有一些学者通过实验证实ꎬ铁死亡的发生可独立于自噬ꎮ4㊀铁死亡与自噬在疾病中的研究4.1㊀铁死亡与自噬在神经系统疾病中的研究㊀蛛网膜下腔出血(subarachnoidhemorrhageꎬSAH)是神经系统疾病中重要的一种ꎮ梁译丹等[33]研究证实ꎬ在SAH的大鼠模型中ꎬ通过侧脑室注射慢病毒沉默ATG5mRNA的表达从而抑制自噬ꎮ与单纯SAH组相比ꎬ慢病毒干预组PCR显示ATG5mRNA显著降低ꎻWestern ̄blot检测结果显示ATG5和LC3Ⅱ/Ⅰ蛋白表达量显著降低(P<0.05)ꎬ表明自噬被成功抑制ꎮ与此同时ꎬ与SAH组相比ꎬ慢病毒干预组的铁死亡标志物FTH1和GPX4表达升高(P<0.05)ꎬGSH含量提高ꎬ细胞内铁沉积减少ꎬ铁含量㊁MDA减少ꎮ上述研究结果表明自噬通过降解铁蛋白ꎬ促进铁死亡ꎮ该研究认为铁死亡的发生依赖于自噬的介导ꎬ在这之中铁蛋白发挥重要的中介作用ꎮ与之前的研究结果一致[28 ̄29]ꎬ即自噬通过降解铁蛋白ꎬ增加细胞内游离铁离子的浓度ꎬ从而促进铁死亡ꎮ这项研究为铁死亡和自噬的相关性提供了实验依据ꎮ4.2㊀铁死亡与自噬在循环系统疾病中的研究㊀Chen等[34]研究证实ꎬ在心力衰竭模型中ꎬ自噬铁死亡是同时发生的ꎮ激活的自噬与铁死亡与心力衰竭的发生发展密切相关ꎮ而Baba等[35]在心肌梗死的研究中得出了不同的结论ꎬ认为雷帕霉素机制靶点(mTOR)可以抑制心脏细胞中铁死亡的发生ꎬ从而发挥保护性作用ꎮ与此同时ꎬmTOR介导的对铁死亡的抑制过程中并没有伴随着自噬标志物的改变ꎬ这提示铁死亡可独立于自噬发生ꎮ4.3㊀铁死亡与自噬在肿瘤中的研究㊀香蒲新苷(typhaneoside)是一种可显著降低急性髓系白血病(acutemyeloidleukemiaꎬAML)细胞活力的药物ꎮ研究表明ꎬ香蒲新苷可通过作用于AMP活化蛋白激酶(AMP ̄activatedproteinkinaseꎬAMPK)信号通路激活自噬ꎬ从而进一步激活自噬依赖的铁蛋白的降解ꎬ最终诱导铁死亡的发生ꎬ而抑制自噬后则可以抑制这种细胞死亡方式[36]ꎮ这充分说明在香蒲新苷促进AML细胞发生铁死亡的过程是依赖于自噬的激活的ꎮ该研究再次为铁死亡和自噬的相互关系提供了充分的实验依据ꎮ而铁死亡与自噬在乳腺癌中的研究得出了不一样的结论ꎮ联合使用西拉米辛和拉帕替尼可以显著诱导乳腺癌细胞发生铁死亡ꎬ也可诱导细胞发生自噬性死亡ꎮ但是Ma等[37]通过实验证实这种联合用药诱导乳腺癌细胞发生自噬性死亡在时间上是落后于铁死亡的ꎮ这表明在西拉米辛和拉帕替尼治疗乳腺癌的过程中ꎬ铁死亡与自噬是独立发生的ꎮ铁死亡的发生并不总是依赖于自噬ꎬ铁死亡与自噬的关系仍未明确ꎬ其内在联系更为复杂ꎬ需要学者们更多的研究与探索ꎮ5㊀小㊀结㊀㊀铁死亡作为一种新型的细胞死亡方式ꎬ与多种疾病的发生发展密切相关ꎮ自噬作为一种保守的大分子或细胞器的降解过程ꎬ在病理情况下也会导致细胞的死亡ꎮ关于铁死亡与自噬的关系ꎬ目前尚无统一结论ꎮ虽然越来越多的研究证实铁死亡的发生伴随着自噬的激活ꎬ但是也有不少的研究表明铁死亡可独立于自噬的激活而存在ꎮ铁死亡与自噬的关系目前仍有许多未知的问题ꎬ且它们在疾病中的关系也处在研究的初级阶段ꎬ未来将在进一步的研究中深入揭示两者间的联系ꎮ参考文献[1]㊀DixonSꎬLembergKꎬLamprechtMꎬetal.Ferroptosis:AnIron ̄De ̄pendentFormofNonapoptoticCellDeath[J].Cellꎬ2012ꎬ149(5):1060 ̄1072.DO:10.1016/j.cell.2012.03.042.[2]㊀TurcoEꎬFracchiollaDꎬMartensS.RecruitmentandActivationoftheULK1/Atg1KinaseComplexinSelectiveAutophagy[J].JMolBi ̄olꎬ2019ꎬPii:S0022 ̄2836(19)30471 ̄1.DOI:10.1016/j.jmb.2019.07.027.[3]㊀HouJꎬRaoMꎬZhengWꎬetal.AdvancesonCellAutophagyandItsPotentialRegulatoryFactorsinRenalIschemia ̄ReperfusionInjury[J].DNACellBiolꎬ2019ꎬ38(9):895 ̄904.DOI:10.1089/dna.2019.4767.[4]㊀YangLꎬMihNꎬAnandAꎬetal.Cellularresponsestoreactiveoxygenspeciesarepredictedfrommolecularmechanisms[J].ProcNatlAcadSciUSAꎬ2019ꎬ116(28):14368 ̄14373.DO:10.1073/pnas.1905039116.[5]㊀ZhuTꎬShiLꎬYuCꎬetal.FerroptosisPromotesPhotodynamicThera ̄py:SupramolecularPhotosensitizer ̄InducerNanodrugforEnhancedCancerTreatment[J].Theranosticsꎬ2019ꎬ9(11):3293 ̄3307.DOI:10.7150/thno.32867.[6]㊀YuHꎬGuoPꎬXieXꎬetal.Ferroptosisꎬanewformofcelldeathꎬanditsrelationshipswithtumourousdiseases[J].JournalofCellularandMolecularMedicineꎬ2016.21(4):648 ̄657.DOI:10.1111/jcmm.13008.[7]㊀GongYꎬWangNꎬLiuNꎬetal.LipidPeroxidationandGPX4Inhibi ̄tionAreCommonCausesforMyofibroblastDifferentiationandFer ̄roptosis[J].DNACellBiolꎬ2019ꎬ38(7):725 ̄733.DOI:10.1089/dna.2018.4541.[8]㊀DixonSJꎬStockwellBR.Theroleofironandreactiveoxygenspeciesincelldeath[J].NatureChemicalBiologyꎬ2013ꎬ10(1):9 ̄17.DOI:10.1038/nchembio.1416.[9]㊀WangHꎬLiJꎬFollettPLꎬetal.12 ̄Lipoxygenaseplaysakeyroleincelldeathcausedbyglutathionedepletionandarachidonicacidinratoligodendrocytes[J].EuropeanJournalofNeuroscienceꎬ2004ꎬ20(8):2049 ̄2058.DOI:10.1111/j.1460 ̄9568.2004.03650.x. [10]㊀PitmanKEꎬAlluriSRꎬKristianAꎬetal.Influxrateof18F ̄fluoroamin ̄osubericacidreflectscystine/glutamateantiporterexpressionintumourxenografts[J].EurJNuclMedMolImagingꎬ2019ꎬ40(10):2190 ̄2198.DOI:10.1007/s00259 ̄019 ̄04375 ̄8.[11]㊀BridgesRJꎬNataleNRꎬPatelSA.Systemxc( ̄)cystine/glutamateantiporter:anupdateonmolecularpharmacologyandroleswithintheCNS[J].BrJPharmacolꎬ2012ꎬ165(1):20 ̄34.DOI:10.1111/j.1476 ̄5381.2011.01480.x.[12]㊀WangLꎬLiuYꎬDuTꎬetal.ATF3promoteserastin ̄inducedferropto ̄sisbysuppressingsystemXc[J].CellDeathDifferꎬ2019.DOI:10.1038/s41418 ̄019 ̄0380 ̄z.[13]㊀YuHꎬGuoPꎬXieXꎬetal.Ferroptosisꎬanewformofcelldeathꎬanditsrelationshipswithtumourousdiseases[J].JournalofCellularandMolecularMedicineꎬ2016ꎬ21(4):648 ̄657.DOI:10.1111/jcmm.13008.[14]㊀KangYꎬTizianiSꎬParkGꎬetal.CellularprotectionusingFlt3andPI3Kαinhibitorsdemonstratesmultiplemechanismsofoxidativeglu ̄tamatetoxicity[J].NatureCommunicationsꎬ2014ꎬ5:3672.DOI:10.1038/ncomms4672.[15]㊀ForcinaGCꎬDixonSJ.GPX4attheCrossroadsofLipidHomeostasisandFerroptosis[J].Proteomicsꎬ2019ꎬ19(18):e1800311.DOI:10.1002/pmic.201800311.[16]㊀Brigelius ̄FloheRꎬMaiorinoM.Glutathioneperoxidases[J].BiochimBiophysActaꎬ2013ꎬ1830(5):3289 ̄3303.DOI:10.1016/j.bba ̄gen.2012.11.020.[17]㊀ForcinaGCꎬDixonSJ.GPX4attheCrossroadsofLipidHomeostasisandFerroptosis[J].Proteomicsꎬ2019ꎬ19(18):e1800311.DOI:10.1002/pmic.201800311.[18]㊀PrattDAꎬTallmanKA.Freeradicaloxidationofpolyunsaturatedlip ̄ids:Newmechanisticinsightsandthedevelopmentofperoxylradicalclocks[J].AccChemResꎬ2011ꎬ44(6):458 ̄467.DOI:10.1021/ar200024c.[19]㊀DixonSJꎬWinterGEꎬMusaviLSꎬetal.HumanHaploidCellGeneticsRevealsRolesforLipidMetabolismGenesinNonapoptoticCellDeath[J].ACSChemicalBiologyꎬ2015ꎬ10(7):1604 ̄1609.DOI:10.1021/acschembio.5b00245.[20]㊀CongcongFꎬLijuanGꎬDanielSꎬetal.TheInterrelationbetweenRe ̄activeOxygenSpeciesandAutophagyinNeurologicalDisorders[J].OxidativeMedicineandCellularLongevityꎬ2017ꎬ2017:8495160.DOI:10.1155/2017/8495160.[21]㊀QomaladewiNPꎬKimMYꎬChoJY.Autophagyanditsregulationbyginsengcomponents[J].JGinsengResꎬ2019ꎬ43(3):349 ̄353.DO:10.1016/j.jgr.2018.12.011.[22]㊀杨静亚ꎬ高俊玲.自噬及其在疾病中的研究进展[J].世界最新医学信息文摘ꎬ2019ꎬ19(52):29 ̄30ꎬ34.DOI:10.19613/j.cnki.1671 ̄3141.2019.52.016.[23]㊀JohansenTꎬLamarkT.SelectiveAutophagy:ATG8FamilyProteinsꎬLIRMotifsandCargoReceptors[J].JMolBiolꎬ2019ꎬpiiꎬS0022 ̄2836(19):30445.DOI:10.1016/j.jmb.2019.07.016.[24]㊀YuHꎬGuoPꎬXieXꎬetal.Ferroptosisꎬanewformofcelldeathꎬanditsrelationshipswithtumourousdiseases[J].JournalofCellularandMolecularMedicineꎬ2016ꎬ21(4):648 ̄657.DOI:10.1111/jcmm.13008.[25]㊀KangRꎬTangD.AutophagyandFerroptosis ̄What'stheConnection[J].CurrentPathobiologyReportsꎬ2017ꎬ5(2):153 ̄159.DOI:10.1007/s40139 ̄017 ̄0139 ̄5.[26]㊀GaoMꎬMonianPꎬPanQꎬetal.Ferroptosisisanautophagiccelldeathprocess[J].Cellresearchꎬ2016ꎬ26(9):1021 ̄1032.DOI:10.1038/cr.2016.95.[27]㊀HouWꎬXieYꎬSongXꎬetal.Autophagypromotesferroptosisbydeg ̄radationofferritin[J].Autophagyꎬ2016ꎬ12(8):1425 ̄1428.DOI:10.1080/15548627.2016.1187366[28]㊀ZhouQꎬFuXꎬWangXꎬetal.AutophagyplaysaprotectiveroleinMn ̄inducedtoxicityinPC12cells[J].Toxicologyꎬ2017ꎬ394:45 ̄53.DOI:10.1016/j.tox.2017.12.001.[29]㊀GargADꎬDudekAMꎬFerreiraGBꎬetal.ROS ̄inducedautophagyincancercellsassistsinevasionfromdeterminantsofimmunogeniccelldeath[J].Autophagyꎬ2013ꎬ9(9):1292 ̄307.DOI:10.4161/auto.25399.[30]㊀HillBGꎬHaberzettlPꎬAhmedYꎬetal.Unsaturatedlipidperoxida ̄tion ̄derivedaldehydesactivateautophagyinvascularsmooth ̄musclecells[J].BiochemicalJournalꎬ2008ꎬ410(3):525 ̄534.DOI:10.1042/BJ20071063.[31]㊀QuilesDelReyMꎬManciasJD.NCOA4 ̄MediatedFerritinophagy:APotentialLinktoNeurodegeneration[J].FrontNeurosciꎬ2019ꎬ13:238.DOI:10.3389/fnins.2019.00238.[32]㊀MouYꎬWangJꎬWuJꎬetal.Ferroptosisꎬanewformofcelldeath:op ̄portunitiesandchallengesincancer[J].JournalofHematology&Oncologyꎬ2019ꎬ12(1):34.DOI:10.1186/s13045 ̄019 ̄0720 ̄y.(下转1292页)10.011.[12]㊀BrazNFTꎬRochaNPꎬVieiraELMꎬetal.Musclestrengthandpsychi ̄atricsymptomsinfluencehealth ̄relatedqualityoflifeinpatientswithmyastheniagravis[J].JClinNeurosciꎬ2018ꎬ50:41 ̄44.DOI:10.1016/j.jocn.2018.01.011.[13]㊀GuptillJTꎬSoniMꎬMeriggioliMN.CurrentTreatmentꎬEmergingTranslationalTherapiesꎬandNewTherapeuticTargetsforAutoim ̄muneMyastheniaGravis[J].Neurotherapeuticsꎬ2016ꎬ13(1):118 ̄131.DOI:10.1007/s13311 ̄015 ̄0398 ̄y.[14]㊀DhawanPSꎬGoodmanBPꎬHarperCMꎬetal.IVIGVersusPLEXintheTreatmentofWorseningMyastheniaGravis:WhatistheEvi ̄dence[J].ACriticallyAppraisedTopicNeurologistꎬ2015ꎬ19(5):145 ̄148.DOI:10.1097/NRL.0000000000000026.[15]㊀刘敏ꎬ邢昂ꎬ丛志强.血浆交换与大剂量丙种球蛋白治疗重症肌无力危象疗效的评价[J].中华神经科杂志ꎬ2005ꎬ10(38):658 ̄659.DOI:10.3760/j.issn:1006 ̄7876.2005.10.016.[16]㊀GamezJꎬSalvadoMꎬCasellasMꎬetal.Intravenousimmunoglobu ̄linasmonotherapyformyastheniagravisduringpregnancy[J].Jour ̄naloftheNeurologicalSciencesꎬ2017ꎬ383:118 ̄122.DOI:10.1016/j.jns.2017.10.037.[17]㊀BharathVꎬEckertKꎬKangMꎬetal.Incidenceandnaturalhistoryofintravenousimmunoglobulin ̄inducedasepticmeningitis:aretrospec ̄tivereviewatasingletertiarycarecent[J].Transfusionꎬ2015ꎬ55(11):2597 ̄2605.DOI:10.1111/trf.13200.[18]㊀StetefeldHRꎬSchroeterM.MyastheneCrisis[J].FortchrNeurolPsychiatrꎬ2018ꎬ86(5):301 ̄307.DOI:10.1055/a ̄0599 ̄0811. [19]㊀黄玲ꎬ王磊ꎬ尹东涛ꎬ等.环磷酰胺序贯治疗伴胸腺瘤的重症肌无力50例临床分析[J].神经疾病与精神卫生ꎬ2018ꎬ18(8):583 ̄586.DOI:10.3969/j.issn.1009 ̄6574.2018.08.012.[20]㊀QiGꎬLiuPꎬDongHꎬetal.MetastaticThymoma ̄AssociatedMyas ̄theniaGravis:FavorableResponsetoSteroidPulseTherapyPlusIm ̄munosuppressiveAgent[J].MedicalScienceMonitorꎬ2017ꎬ23ꎬ1217 ̄1223.DOI:10.12659/MSM.902442.[21]㊀冯慧宇ꎬ刘卫彬ꎬ邱力ꎬ等.中剂量环磷酰胺联合甲泼尼龙治疗重症肌无力危象的随机对照临床研究[J].中华医学杂志ꎬ2012ꎬ92(35):2473 ̄2476.DOI:10.3760/cma.j.issn.0376 ̄2491.2012.35.007.[22]㊀RodaRHꎬDohertyLꎬCorseAM.stoppingoralsteroidsparingagentatinitiationofrituximabinmyastheniagravis[J].NeuromusculDis ̄ordꎬ2019ꎬ29(7):554 ̄561.DOI:10.1016/j.nmd.2019.06.002. [23]㊀IorioRꎬDamatoVꎬAlboiniPEꎬetal.Efficacyandsafetyofritux ̄imabformyastheniagravis:asystematicreviewandmetaanalysis[J].JNeurolꎬ2015ꎬ262(5):1115 ̄1119.DOI:10.1007/s00415 ̄014 ̄7532 ̄3.[24]㊀KoulRꎬFutaisiAꎬAbdwaniR.Rituximabinsevereseronegativeju ̄venilemyastheniagravis:reviewoftheliterature[J].PediatriNeu ̄rolꎬ2012ꎬ47(3):209 ̄212.DOI:10.1016/j.pediatrneurol.2012.05.017.[25]㊀JingSꎬSongYꎬSongJꎬetal.Responsivenesstolowdoserituximabinrefractorygeneralizedmyastheniagravis[J].JNeuroimrnunolꎬ2017ꎬ311:14 ̄21.DOI:10.1016/j.jneuroim.2017.05.021. [26]㊀TopakianRꎬZimprichFꎬIglsederSꎬetalꎬHighefficacyofrituximabformyastheniagravis:acomprehensivenationwidestudyinAustria[J].JNeurolꎬ2019ꎬ266(3):699 ̄706.DOI:10.1007/s00415 ̄019 ̄09191 ̄6.[27]㊀OnukiTꎬUedaSꎬOtsuSꎬetal.ThymectomyduringMyasthenicCrisisunderArtificialRespiration[J].AnnThoracCardiovascSurgꎬ2019ꎬ25(4):215 ̄218.DOI:10.5761/atcs.cr.17 ̄00176.[28]㊀薛银萍ꎬ乞国艳ꎬ董会民ꎬ等.参芪扶正注射液在重症肌无力激素冲击治疗过程中的应用[J].广东医学ꎬ2016ꎬ37(24):3764 ̄3756.DOI:10.3969/j.issn.1001 ̄9448.2016.24.035.[29]㊀徐鹏ꎬ吕志国ꎬ王健ꎬ等.重症肌无力中医循证性临床诊疗指南修订实践研究[J].中华中医药杂志ꎬ2018ꎬ33(5):1979 ̄1983.DOI:CNKI:SUN:BXYY.0.2018 ̄05 ̄066.[30]㊀吴周烨ꎬ吴颢昕ꎬ何骁隽ꎬ等.益气升提法治疗实验性自身免疫性重症肌无力大鼠免疫机制研究[J].中华中医药杂志ꎬ2017ꎬ32(6):2746 ̄2749.DOI:CNKI:SUN:BXYY.0.2017 ̄06 ̄112. [31]㊀方学君ꎬ梁艺ꎬ刘晓曼ꎬ等.强肌健力方对重症肌无力大鼠Th17/Treg平衡的影响[J].广州中医药大学学报ꎬ2019ꎬ36(2):240 ̄245.DOI:10.13359/j.cnki.gzxbtcm.2019.02.019.[32]㊀王艳君ꎬ孟庆芳ꎬ王思青ꎬ等.蒿素对实验性自身免疫性重症肌无力大鼠R97 ̄116抗体及细胞因子的影响[J].中国神经免疫学和神经病学杂志ꎬ2016ꎬ23(3):167 ̄170.DOI:10.3969/J.issn.1006 ̄2963.2016.03.002.(收稿日期:2019-06-14)(上接1287页)[33]㊀梁译丹ꎬ覃王ꎬ黄豪ꎬ等.自噬通过降解铁蛋白促进神经元铁死亡参与蛛网膜下腔出血后早期脑损伤[J].第三军医大学学报ꎬ2019ꎬ41(15):1407 ̄1414.DOI:10.16016/j.1000 ̄5404.201902124.[34]㊀ChenXꎬXuSꎬZhaoCꎬetal.RoleofTLR4/NADPHoxidase4path ̄wayinpromotingcelldeaththroughautophagyandferroptosisduringheartfailure[J].BiochemBiophysResCommunꎬ2019ꎬ516(1):37 ̄43.DOI:10.1016/j.bbrc.2019.06.015.[35]㊀BabaYꎬHigaJKꎬShimadaBKꎬetal.ProtectiveEffectsoftheMecha ̄nisticTargetofRapamycinagainstExcessIronandFerroptosisinCardiomyocytes[J].AJPHeartandCirculatoryPhysiologyꎬ2017ꎬ314(3):H659 ̄H668.DOI:10.1152/ajpheart.00452.2017. [36]㊀ZhuHYꎬHuangZXꎬChenGQꎬetal.Typhaneosidepreventsacutemyeloidleukemia(AML)throughsuppressingproliferationandin ̄ducingferroptosisassociatedwithautophagy[J].BiochemBiophysResCommunꎬ2019ꎬ516(4):1265 ̄1271.DOI:10.1016/j.bbrc.2019.06.070.[37]㊀MaSꎬDielschneiderRFꎬHensonESꎬetal.Ferroptosisandautophagyinducedcelldeathoccurindependentlyaftersiramesineandlapatinibtreatmentinbreastcancercells[J].PLoSOneꎬ2017ꎬ12(8):e0182921.DOI:10.1371/journal.pone.0182921.(收稿日期:2019-05-27)。
尼古丁对慢性铅暴露大鼠海马CA1区NMDA受体介导的突触可塑性的修复作用
Reparatory Effects of Nicotine on NMDA Receptor-mediated Synaptic Plasticity in the Hippocampal CA1 Region of Chronically Lead-exposed RatsWang Huili Ruan Diyun *E-mail : Whl1028@ Ruandy@Department of Neurobiology and Biophysics,School of Life Science ,University of Science & Technology of China,Hefei, Anhui 230027, P.R. ChinaAbstractActivation of neuronal nicotinic acetylcholine receptors (nAChRs) modulates the induction of long-term potentiation (LTP), one of the possible cellular mechanisms for learning. To investigate the effect of nicotine on synaptic plasticity in chronically lead-exposed rats, field excitatory postsynaptic potentials (fEPSPs) and paired-pulse facilitation (PPF) were recorded in the CA1 area of hippocampal slices from chronically lead-exposed rats of 23-30 days old. The results showed that: (1) 1µM nicotine facilitated the induction of LTP in CA1 area of the hippocampus by a weak tetanic stimulation (100 Hz, 20 Pulses), which does not by itself produce LTP in lead-exposed rats. This effect was significantly suppressed by mecamylamine (MEC), a nonselective nicotinic antagonist, suggesting that the facilitation of LTP was through nAChRs. (2) The nicotine-mediated LTP was blocked by dihydro-β-erythroidine (DhβE), a non-α7 nAChR antagonist, while long-term depression (LTD) produced by the combination of nicotine and methyllycaconitine (MLA), a α7-nAChR antagonist and neither LTP nor LTD observed by the combination of nicotine, MLA and DHβE. It implied that several nAChRs subtypes were involved in the nicotine-mediated synaptic plasticity. (2) Nicotine enhanced PPF in hippocampal CA1 region, and the nicotine-mediated LTP in lead-exposed rats was blocked by either D-(-)-2-amino-5-phosphonopentanoic acid (D-AP5), the NMDAs receptor antagonist, or picrotoxin (PTX), an antagonist of γ-aminobutyric acid (GABA)A receptor. It suggested that nicotine-mediated synaptic plasticity was due to the activation of NMDARs by disinhibition of pyramidal cells through presynaptic nAChRs. This may represent the cellular basis of nicotine-mediated cognitive enhancement observed in chronically lead-exposed rats.Key words: chronic lead exposure; nicotine; nAChRs; synaptic plasticity; LTP*Corresponding Author:This work was supported by the National Basic Research Program of China (No. 2002CB512907), the National Natural Science Foundation of China (No., 30300288), Specialized Research Fund for the Doctoral Program of Higher Education (No. 20020358053), IntroductionLead is a well-known environmentally toxic agent that causes severe cognitive impairments in children (Lidsky & Schneider, 2003; Meng et al., 2005). Previous studies have reported that chronic lead exposure during development raises the threshold for and impairs the induction of LTP of excitatory postsynaptic potentials (EPSPs) in rat hippocampus (Lasley & Gilbert, 1999; Ruan et al., 2000). In addition, lead directly inhibits nicotinic acetylcholine receptors (nAChRs) function in the hippocampus (Mike et al., 2000). A substantial loss of cholinergic receptors has also been found in lead-exposed rats (Costa & Fox, 1983; Bielarczyk et al., 1994; Bielarczyk et al., 1996; Bourjeily & Suszkiw, 1997; Reddy et al., 2003). Because nAChRs play an important role in enhancing cognitive functions (Jones et al., 1999; Levin, 2002) and nicotine facilitates the induction of LTP (Fujii et al., 1999; Fujii & Sumikawa, 2001; Mansvelder et al., 2002), we asked whether nicotine can be used as a potential therapeutic agent, with nAChRs as therapeutic target, to treat impairments in plasticity caused by lead. Although it has been reported that nicotine attenuates the spatial learning deficits in rats chronically exposed to lead (Zhou & Suszkiw, 2004), whether nAChRs are directly involved in the induction of LTP in lead-exposed rats remains unclear.nAChRs are expressed abundantly in the hippocampus, a cerebral structure involved in learning and memory (Fabian-Fine et al., 2001). While most of nAChRs are expressed in GABAergic interneurons, some are also found in the pyramidal neurons as well. Two of the most abundant and well-characterized subtypes of nAChRs in the hippocampus are the heteromeric and homomeric α7-bearing receptors (Alkondon & Albuquerque, 1993; Role & Berg, 1996). Both receptor subtypes can modulate the induction of synaptic plasticity (Mann & Greenfield, 2003; Yamazaki et al., 2005), which may explain the effect of nicotinic agonists on learning and memory. In addition, nicotine at low concentration also enhances the release of excitatory,inhibitory and modulatory neurotransmitters by acting on presynaptic nAChRs in both synaptosome preparation (Wonnacott, 1997) and in the hippocampus (Gray et al., 1996).In the present study, we investigated the nicotinic modulation of synaptic transmission and plasticity in the hippocampus of rats chronically exposed to lead. We also examined whether this nicotinic modulation is through nAChRs and which subtypes of nAChRs are involved in the nicotine-mediated synaptic plasticity. In the CA1 region of the hippocampus, NMDA receptors (NMDARs) play an important role in the induction and maintenance of LTP. In order to explore whether any nicotinic augmentation of LTP in the lead-exposed rats is due to NMDAR activation, we repeated the same experiments in the presence of the NMDARs antagonist, D-(-)-2-amino-5-phosphonopentanoic acid (D-AP5). Finally, given that nAChRs are widely expressed in GABAergic interneurons in the hippocampus, we also examined whether the observed effects of nicotine resulted from modulation of GABAergic transmission using a GABA A receptors antagonist picrotoxin (PTX). In this study, we investigated the reparatory effects of nicotine on NMDA receptor-mediated synaptic plasticity in the hippocampal CA1 region of chronically lead-exposed rats and discussed the mechanism of its effects. Materials and methods1. Experimental animalsThe Wistar rats were obtained from the Laboratory Animal Center, University of Science and Technology of China, P. R. China, and were maintained in accordance with Guide for the Care and Use of Laboratory Animals (US National Research Council, 1996).All efforts were made to minimize the number of animals used and their suffering.The protocol for chronic exposure to lead has been described previously (Ruan et al., 1998; Ruan et al., 2000; Cai et al., 2001). Wistar dams were fed on standard laboratory chow and distilled water ad libitum at carefully controlled ambient temperature (24 ± 1°C) and relative humidity (50 ± 10%). On parturition day, the dams were randomly divided into two groups: control and lead-exposed. The lead-exposed pups acquired lead via milk of dams whose drinking water contained 0.2% (1090 ppm) lead acetate from parturition to weaning, while the control dams remained on distilled water throughout the lactation period. Litters were culled to eight pups with both sexes. At the age of 21 days, offspring were weaned, housed in a colony room with a 12:12 light:dark schedule and permitted free access to food and distilled water. Extracellular recordings were carried out at the age of 23–30 days (weight: 40–50 g) in a total of 32 control (16 males and 16 females; mean weight: 44.6 g) and 56 lead-exposed rats (28 males and 28 females; mean weight: 43.8 g). For any given experimental measure, equal numbers of females and males were used, andno more than two animals were selected from each litter.2. Blood and hippocampal lead determinationsBlood and hippocampal lead determinations were made in littermates of the same animals utilized for electrophysiology on the day that recordings were made. Blood samples (2 ml per animal) were collected in heparinized syringes via cardiac puncture of anesthetized rats. After decapitation, the two hippocampi were collected. Lead concentrations were measured by a plasmaQuad3 plasma mass spectrograph (VG Elemental, UK) after the tissues were digested with an organic tissue solubilizer.3. Slice preparations and recordingsThe experiments were performed on transverse slices prepared from the hippocampi at the age of 23-30 days. The rats were anesthetized with anhydrous diethyl ether and killed by decapitation. Then the brains were quickly removed and immersed in cold (nearly 0°C) oxygenated (95% O2/5% CO2) artificial cerebrospinal fluid (ACSF) containing (in mM): NaCl 124, KCl 5, NaH2PO4 1.25, NaHCO3 26, MgCl2 1.25, CaCl2 2.5 and dextrose 10, pH=7.30-7.45.The hippocampus in one hemisphere was dissected free and sectioned into about 400 µm thick slices by using a manual chopper. The slices were incubated for at least 1h in ACSF at room temperature (21–25◦C). Then one hippocampal slice was transferred to the recording chamber (BSC-HT Med. Sys., USA) in which it was continuously perfused at a rate of 1 ml/min with 30–32°C ACSF saturated with 95% O2/5% CO2. After at least 1 hequilibration in the slice chamber, a bipolar stimulating electrode was located in Schaffer/commissural fibers. The recording electrode, a glass micropipette (resistance: 3–5 MΩ) filled with ACSF, was positioned in the dendritic layer of the area CA1. Testing single shocks of 0.2 ms duration at 0.05 Hz evoked EPSPs. At the start of each experiment, a full input–output (I/O) curve was constructed. The stimulus intensity that yielded 1/2 and 2/3 of the maximal response was selected for baseline measurements. After recording the baseline responses for 20 min, LTP was induced by the application of a weak tetanic stimulation (TS, 100 Hz, 20 Pulses). Testing with single shocks was continued for at least 60 min after LTP was induced. LTP was presented as the increase in EPSPs slopes in relation to the baseline response (100%) after the weak tetanic stimulation application, and its amplitude was the mean of relative EPSPs slopes in 45–60 min. At the end of each experiment, the I/O curve was reconstructed. Pharmacological agents, when used, were administered 10 min prior to TS, unless otherwise noted.4. Drug applicationDrugs used were nicotine, mecamylamine (MEC), methyllycaconitine (MLA), dihydro-β-erythroidine (DHβE), D-(-)-2-amino-5-phosphonopentanoic acid (D-AP5) and picrotoxine (PTX) (all purchased from Sigma-Aldrich, USA). A stock solution of℃each drug was made up, aliquoted and frozen at -20. Aliquots were thawed only once and diluted in oxygenated ACSF immediately prior to application via a perfusion system. 5. Data analysisData were recorded using Igor Pro 4.05 software (Wave Metrics Inc, OR, USA) and analyzed with Origin 7.5 (OriginLab Corporation, MA, USA). The time scale in each experiment was converted to time from the onset of the weak tetanic stimulation. Results were expressed as means ± S.E.M, and n represents the number of the animals that were sampled. Statistical differences were assessed by one-way analyses of variance (ANOVA) and two-tailed Student’s t-tests for post hoc multiple comparisons. Probabilities less than 0.05 were considered significant.ResultsIn order to rule out the potential difference associated with gender, blood and hippocampal lead levels in male and female rats were measured and no significant differences between them were found both in control and lead-exposed groups. So were the amplitudes of nicotine-mediated LTP (n = 8 for each group).1. Blood and hippocampal lead levelsLead concentrations in blood and hippocampus were 6.52 ± 0.43 µg/dl and 0.86 ± 0.11 µg/g, respectively, in 8 control rats, and 43.0 ± 13.23 µg/dl and 3.8 ± 0.5 µg/g, respectively, in 8 lead-exposed rats. Lead levels in blood and hippocampus of the lead-exposed rats were significantly higher than those of the controls (n = 8,P < 0.01). 2. Effect of nicotine on LTP in chronically lead-exposed rat hippocampusWe chose to use 1µM nicotine throughout our study because this dose has been shown to facilitate the induction of LTP in the hippocampus (Fujii et al., 1999; Fujii et al., 2000b; Yamazaki et al., 2005) without affecting single-pulse field EPSPs (fEPSPs) in both control and lead-exposed slices (data not shown), which is consistent with previous experiments (Fujii et al., 1999). After a 20-min baseline field EPSPs recordings was established, 1µM nicotine was bath-applied for 10 min, followed by a weak tetanic stimulation (100 Hz, 20 pulses) to induce LTP. fEPSPs were continuously recorded for more than 60 min after tetanic stimulation. Fig.1 shows that with this weak tetanic stimulation, LTP was successfully induced in control rats, but not in the lead-exposed rats without nicotine treatment. Our previous work (Cai et al., 2001; Yu et al., 2005) demonstrated that a stronger stimulation (100 Hz, 100 pulses) could induce LTP in rats chronically exposed to lead. Thus, lead raised the threshold for LTP induction. However, with nicotine treatment, the same weak tetanic stimulation (100 Hz, 20 Pulses) induced LTP in lead-exposed rats (Fig.1). The magnitude of LTP was significantly increased in nicotine-treated lead-exposed slice (130.85 ±6.36%) compared with lead-exposed slice values (100.8 ± 4.6%) (n=8, P<0.01).3. Effect of MEC on the nicotine-mediated synaptic plasticity in rats chronically exposed with lead Mecamylamine (MEC) is recognized as a non-selective nicotinic antagonist. Superfusion of 5µM MEC alone to the lead-exposed slice produced no significant effect on fEPSPs or LTP (data not shown). In addition, MEC completely blocked LTP in slices that were treated with 1µM nicotine (5-min MEC treatment prior to nicotine application, n=8, Fig.2). This result confirmed that nicotine facilitated the induction of LTP by activating nAChRs.4. Effects of MLA and DHβE on the nicotine-mediated synaptic plasticity in rats chronically exposed to leadTo explore which subtype of nAChRs is involved in nicotine-mediated synaptic plasticity, we conducted the experiments in the presence of α7 and non-α7 nAChR antagonists: MLA (10 nM) and DHβE (1µM), respectively. Superfusion of MLA or DHβE alone to the lead-exposed slice caused no significant difference in LTP by the weak tetanic stimulation (data not shown). DHβE blocked nicotine-induced LTP (n = 8). However, MLA treatment after nicotine produced LTD in slices from lead-exposed rats (n = 8, Fig.3). This type of LTD was further blocked by DHβE (n= 8). These results suggest that multiple subtypes of nAChRs are involved in the modulatory effect of nicotine on synaptic plasticity.5. Nicotine application enhanced paired-pulse facilitation (PPF) in lead-exposed ratsThe predominant role of nAChRs in the brain is to modulate neurotransmitter release at presynaptic terminals (McGehee et al., 1995; Wonnacott, 1997). To examine whether nicotine altered postsynaptic responses through a mechanism involving presynaptic nAChRs, we used paired-pulse stimulation protocol and analyzed the ratio of the second postsynaptic response to the first postsynaptic response. PPF is a presynaptic form of short-term synaptic plasticity resulting from enhanced transmitter release revealed by the second of a pair of closely spaced stimulation pulses (Zucker, 1989).We evoked fEPSPs with half-maximum stimulating current intensities and measured the magnitude of PPF of fEPSPs slopes at different inter-pulse intervals (IPIs) from control and lead-exposed slices (Fig. 4). 1µM nicotine perfusion induced significant enhancement of PPF for the 20–180 ms IPIs in both control and lead-exposed groups (n = 8, P<0.05). The four groups all exhibited a maximal facilitation at an inter-pulse interval of 30 ms. The average peak facilitation was 172.35%, 185.1%, 120.6% and 150.8% (n = 8 for each group) in the control, nicotine, lead-exposed and lead plus 1µM nicotine groups, respectively.6. Effects of D-AP5 and picrotoxin on the nicotine-mediated synaptic plasticity in lead-exposed ratNMDA receptors act in the induction and maintenance of LTP in the CA1 area of hippocampus. To investigate the role of NMDA receptors in nicotine-mediated LTP in slices from lead-exposed rats, we applied 50µM D-AP5 5 min prior to the nicotine treatment. D-AP5 prevented the nicotine-induced LTP in these slices (Pb: 100.8±4.6%, n=8; Pb + nicotine: 130.8±5.5%, n=8; Pb + nicotine + D-AP5: 99.5±5.6%, n=8, P<0.05 cf. Pb + nicotine group; Fig. 5). This result suggests that the 1µM nicotine-induced LTP in hippocampal CA1 region of lead-exposed rats was also NMDA-dependent.Most of the nAChRs are located in GABAergic interneurons in CA1 area of hippocampus, so any observed effect of nicotine on LTP induction in lead-exposed rats could result from the GABA receptors. To test the hypothesis, the effect of the GABA A receptor antagonist picrotoxin (PTX, 50µM) on LTP in lead-exposed rats was examined. PTX blocked the nicotine-induced enhancement of LTP (Pb: 100.8±4.6%, n=8; Pb + nicotine: 130.8±5.5%, n=8, P<0.05 cf. Pb; Pb + nicotine + PTX: 105.2±6.4%, n=8, P<0.05 cf. Pb + nicotine group; Fig. 6).DiscussionWe studied the nicotine-mediated synaptic plasticity in hippocampal slices from rats chronically exposed to lead during development and demonstrated that nicotine affects a number of functions in hippocampal CA1 region.1. Nicotine lowered the threshold for the induction of LTP in lead- exposed rats.Acetylcholine receptors play important roles in learning and memory. Several reports attested the effectiveness of nicotine in ameliorating cognitive dysfunction and reducing memory impairments associated with various neurological disorders in humans as well as animals with lesions to the hippocampus and/or basal forebrain cholinergic system (Levin & Simon, 1998; Rezvani & Levin, 2001).Our study demonstrated for the first time that nicotine facilitated the induction of LTP in CA1 area of the hippocampus in chronically lead-exposed rat. Previous studies have shown that there was a significant loss of the septo-hippocampal cholinergic afferents following developmental lead exposure (Bielarczyk et al., 1994; Bielarczyk et al., 1996; Bourjeily & Suszkiw, 1997). Intrahippocampal transplants of cholinergic-rich septal and nucleus basalis tissue reduced lead-induced deficits in spontaneous locomotor activity (Adhami et al., 2000). Acute nicotine treatment improved performance of lead-exposed rats in memory tasks (Adhami et al., 2000; Zhou & Suszkiw, 2004). Our results provided a cellular mechanism for the above findings. Similar to a previous study showing that nicotine enhanced the induction of long-term potentiation in the hippocampus by compensating for 192-IgG-saporin-induced loss of cholinergic function (Yamazaki et al., 2002), our study suggests that nicotine compensates for the diminished activation of nAChRs caused by lead exposure, and rescues impaired LTP in lead-exposed rats.2. Multiple nAChRs are involved in the nicotine-mediated synaptic plasticity in rats chronically exposed to leadFunctional nAChRs in the mammalian brain consist of heteromeric and homomeric α7-bearing receptors subtypes, both play important roles in memory tasks, particularly those that involve the hippocampus (Matsuyama & Matsumoto, 2003). The α7-containing nAChR displays distinctive features, including high Ca2+ permeability and high single-channel conductance (Seguela et al., 1993). Mike et al. found that lead accelerated the rate of receptor desensitization by reducing the frequency of opening and the mean open time of α7-containing nAChR channels.Nicotine application reversed the receptor desensitization, leading to the rise of [Ca2+]i (Mike et al., 2000).Our study showed that MEC, a nonselective nicotinic receptor antagonist, completely abolished the acute nicotine-induced LTP in hippocampal slices from rats chronically exposed to lead. It suggests that nAChRs are involved in the nicotine-mediated synaptic plasticity in lead-exposed rats. Also, in order to explore the subtypes of nAChRs being involved, we performed experiments in the presence of methyllycaconitine (MLA), an antagonist of nAChRs containing α7 subunits, and dihydro-β-erythroidine (DHβE), a potent antagonist of the nAChRs containing non-α7 subtypes, especially α4β2 subunit. In this study, MLA or DHβE caused no significant change in LTP induction in the chronically lead-exposed animals in the absence of exogenously added nicotine. This result is different from previous reports showing that MLA induced LTP in normal hippocampal slices, which suggests that inactivation of α7-containing nAchR plays a role in the induction of LTP (Fujii et al., 2000a; Yamazaki et al., 2005). It remains to be determined whether this discrepancy is due to lead-induced inhibition of α7-containing nAChRs.The LTD produced by nicotine and MLA co-treatment is an interesting observation. It is possible that several nAChR subtypes are involved in different forms of nicotine-mediated synaptic plasticity, such as LTP and LTD. The interaction between lead and cholinergic system, in particular with nicotinic receptors, has been reported (Oortgiesen et al., 1997). The bi-directional model of plasticity describes a calcium dependent induction of synaptic plasticity (Connor et al., 1999; Nishiyama etal., 2000). Small increase in intracellular calcium concentration leads to LTD and large increase leads to LTP. In the absence of a significant calcium increase or with intermediate levels of calcium influx, neither LTP nor LTD is induced. What we have observed in the present study may result from regulation of calcium influx due to differential activation of different nAChR subtypes. In the absence of nicotine, the calcium influx produced by depolarization and glutamate receptor activation resulting from the weak tetanic stimulation (100Hz, 20pulses) is insufficient to induce LTP in lead-exposed hippocampus. When the weak tetanic stimulation is applied in the presence of nicotine, activation of nAChRs boosts the calcium influx to a level high enough to produce LTP. Although α7-containing nAChRs are susceptible to desensitization and lead may accelerate the rate of desensitization, it seems that the additional calcium influx due to activation of these receptors is sufficient to induce the cascade of events that lead to LTP. MLA application blocks calcium-permeable AChRs such as α7-containing receptors (Seguela et al., 1993; Castro & Albuquerque, 1995), therefore at relatively low levels of intracellular calcium the weak tetanic stimulation leads to LTD. Alternatively, MLA at the concentration used in this study may also be acting on more than one receptor subtypes, which leads to a net decrease in calcium levels and results in LTD. When using nicotine combined with DHβE, which more effectively blocks non-α7-containing receptors, part of the calcium influx was reduced. The levels of calcium under the experimental condition may be at an intermediate level which is neither low enough to produce LTD nor high enough to produce LTP. When nicotine is combined with two antagonists, MLA and DHβE, multiple nAChRs are blocked and the calcium level is further reduced to below the threshold for LTD induction.3. Nicotine-mediated synaptic plasticity is due to the activation of NMDA receptor through nicotine-induced disinhibition of pyramidal cellsIn the presence of D-AP5, a specific antagonist of NMDA receptor, the nicotine-mediated synaptic plasticity in lead-exposed rats was completely blocked, suggesting that this type of LTP is also NMDA receptor-dependent. Furthermore, our study demonstrated that acute nicotine treatment enhanced PPF in lead-exposed rats. PPF is commonly accounted for by presynaptic residual calcium. Since presynaptic calcium influx is crucial to PPF and Pb2+ reduces presynaptic Ca2+ entry through presynaptic α7-containing nAChRs (Mike et al., 2000; Braga et al., 2004), our result suggest that acute nicotine treatment in lead-exposed rats enhances presynaptic Ca2+ levels through presynaptic nAChRs. It is possible that activation of presynaptic nAChRs leads to more glutamate, which strongly activates postsynaptic NMDA receptors, resulting in a higher postsynaptic calcium influx and LTP. More investigation of the molecular mechanism is required.Hippocampal nAChRs are predominantly located on GABAergic interneurons (Sudweeks & Yakel, 2000). In this study, a blockade of GABA A receptor with PTX prevented the induction of LTP produced by nicotine in the lead-exposed rats. This provides strong evidence supporting that GABAergic responses are crucially involved in the augmentation effect of nicotine on the induction of LTP. In conclusion, acute nicotine treatment facilitated the induction of LTP through nAChRs in hippocampal slice from rats chronically exposed to lead. Several subtypesof nAChRs were involved in the nicotine-mediated synaptic plasticity. Moreover, this effect may be due to the enhancement of NMDAR responses mediated by nicotine-induced disinhibition of pyramidal cells, probably through presynaptic nAChRs.AcknowledgementsWe are grateful to Professor Franco Lepore for critical reading and valuable comments for the manuscript. This work was supported by the National Basic Research Program of China (No. 2002CB512907), Academic Sinica (No. KZCX3-SW-437, KZCX3-SW-151), the National Natural Science Foundation of China (No. 40231002 , 30300288),Specialized Research Fund for the Doctoral Program of Higher Education (No.20020358053), Anhui Provincial Natural Science Fundation (No. 050430801), and University of Science and Technology of China (No. KB0833).AbbreviationsACh, acetylcholine; ACSF, artificial cerebrospinal fluid; D-AP5, D-(-)-2-amino-5-phosphonopentanoic acid; DHβE, dihydro-β-erythroidine; fEPSPs, field excitatory postsynaptic potentials; GABA, γ-aminobutyric acid; LTP, long-term potentiation; MEC, mecamylamine; MLA, methyllycaconitine; nAChRs, nicotinic acetylcholine receptors; NIC, nicotine; PPF, paired-pulse facilitation; PTX, picrotoxin; TS, tetanic stimulation.ReferencesAdhami, V.M., Husain, R., Agarwal, A.K. & Seth, P.K. (2000) Intrahippocampal cholinergic-rich transplants restore lead-induced deficits: a preliminary study in rats. Neurotoxicol. Teratol., 22, 41-53.Alkondon, M. & Albuquerque, E.X. (1993) Diversity of nicotinic acetylcholine receptors in rat hippocampal neurons. I. Pharmacological and functional evidence for distinct structural subtypes. J. Pharmacol. Exp. Ther., 265, 1455-1473.Bielarczyk, H., Tian, X. & Suszkiw, J.B. (1996) Cholinergic denervation-like changes in rat hippocampus following developmental lead exposure. Brain Res., 708, 108-115. Bielarczyk, H., Tomsig, J.L. & Suszkiw, J.B. (1994) Perinatal low-level lead exposure and the septo-hippocampal cholinergic system: selective reduction of muscarinic receptors and cholineacetyltransferase in the rat septum. Brain Res., 643, 211-217.Bourjeily, N. & Suszkiw, J.B. (1997) Developmental cholinotoxicity of lead: loss of septal cholinergic neurons and long-term changes in cholinergic innervation of the hippocampus in perinatally lead-exposed rats. Brain Res., 771, 319-328.Braga, M.F., Pereira, E.F., Mike, A. & Albuquerque, E.X. (2004) Pb2+ via protein kinase C inhibits nicotinic cholinergic modulation of synaptic transmission in the hippocampus. J. Pharmacol.Exp. Ther., 311, 700-710.Cai, L., Ruan, D.Y., Xu, Y.Z., Liu, Z., Meng, X.M. & Dai, X.Q. (2001) Effects of lead exposure on long-term potentiation induced by 2-deoxy-D-glucose in area CA1 of rat hippocampus in vitro. Neurotoxicol. Teratol., 23, 481-487.Castro, N.G. & Albuquerque, E.X. (1995) alpha-Bungarotoxin-sensitive hippocampal nicotinic receptor channel has a high calcium permeability. Biophys. J., 68, 516-524.。
n氨基甲酰基l天冬氨酸的英文
n氨基甲酰基l天冬氨酸的英文N-Acetylaspartylglutamic acid (NAAG) is a peptide neurotransmitter found in the central nervous system. It plays a role in modulating synaptic transmission and is involved in various physiological processes such as learning, memory, and pain sensation. NAAG is synthesized from N-acetylaspartate (NAA) and glutamate through the enzymatic action of NAAG synthetase.The presence of NAAG in the brain suggests its importance in neurotransmission. It acts as a neurotransmitter by binding to specific receptors known as metabotropic glutamate receptors (mGluRs), particularly mGluR3. Activation of mGluR3 by NAAG leads to various cellular responses, including inhibition of neurotransmitter release and regulation of ion channels.Research has shown that NAAG levels are altered incertain neurological disorders. For example, decreased NAAG levels have been observed in conditions such as Alzheimer's disease and schizophrenia, while increased levels have been reported in epilepsy. These findings suggest that NAAG mayplay a role in the pathophysiology of these disorders andcould potentially serve as a therapeutic target.In addition to its role as a neurotransmitter, NAAG has also been investigated for its potential therapeutic applications. Studies have explored the use of NAAG and NAAG-based compounds in the treatment of various neurological conditions, including neuropathic pain, stroke, and traumatic brain injury. Preclinical studies have shown promising results, but further research is needed to determine the efficacy and safety of NAAG-based therapies in humans.Overall, N-acetylaspartylglutamic acid is a fascinating molecule with diverse functions in the central nervous system.Its role as a neurotransmitter and its potential therapeutic applications make it an intriguing target for further research in neuroscience and drug development.。
【铁死亡】非典型的谷氨酸-半胱氨酸连接酶活性防止铁死亡
【铁死亡】非典型的谷氨酸-半胱氨酸连接酶活性防止铁死亡2020年12月,来自佛罗里达H.Lee. Moffitt肿瘤研究中心的Gina M. DeNicola课题组在Cell Metabolism上发表了题为“Non-canonical Glutamate-Cysteine Ligase Activity Protects against Ferroptosis”的文章,报道了在NRF2激活的细胞中,谷氨酸-半胱氨酸连接酶催化亚基(GCLC)通过合成γ-谷氨酰肽保护细胞免受由半胱氨酸饥饿诱导的铁死亡,这种保护作用不依赖于谷胱甘肽。
——背景——铁死亡是一种铁依赖性的非凋亡细胞死亡,由细胞内脂质氢过氧化物累积所致。
半胱氨酸是维持细胞氧化还原稳态所必需的,半胱氨酸剥夺会诱导铁死亡。
GSH是由半胱氨酸、谷氨酸和甘氨酸组成的三肽,GSH的合成分两步进行。
首先,通过GCLC连接谷氨酸和半胱氨酸,生成γ-谷氨酰-半胱氨酸(γ-Glu-Cys)。
然后γ-Glu-Cys与甘氨酸连接,生成三肽GSH。
虽然半胱氨酸与GSH合成直接相关,会影响ROS水平和脂质过氧化物酶GPX4的活性,但除了用于GSH合成,对胱氨酸饥饿的代谢后果了解甚少。
——结果——1.转硫途径不能支持NSCLC细胞系的半胱氨酸水平首先,对NSCLC细胞系进行胱氨酸饥饿处理导致细胞死亡,细胞死亡伴随着脂质过氧化物的积累,并可被铁死亡抑制剂所抑制。
转硫途径可以使细胞从头合成半胱氨酸。
通过碳同位素标记,发现在正常及饥饿条件下同位素标记半胱氨酸含量都极低,因此在胱氨酸饥饿处理4h后,细胞内半胱氨酸强烈耗竭。
这些结果表明,转硫途径无法支持NSCLC细胞系中的半胱氨酸水平,从而导致在胱氨酸饥饿条件下半胱氨酸耗竭,GSH合成受损。
图1 转硫途径不能支持NSCLC的半胱氨酸池A. 在胱氨酸缺乏和正常条件下,用DMSO或 Fer-1处理NSCLC后的细胞死亡。
B. 同位素标记丝氨酸示意图。
alternative activation of macrophages
alternative activation of macrophages1. 引言1.1 概述在免疫系统中,巨噬细胞作为一类重要的免疫细胞,具有清除病原体和调节免疫应答的功能。
长期以来,人们习惯将巨噬细胞分为两种状态:经典型激活(M1)和替代型激活(M2)。
替代型巨噬细胞激活是指在一些特定的微环境条件下,巨噬细胞表现出一种不同于M1状态的极化状态。
相比于M1型巨噬细胞,替代型巨噬细胞具有促进修复和再生、抑制炎症反应和调节免疫应答等特点。
1.2 文章结构本文将分为以下几个部分进行探讨。
首先,在第二部分中,我们将详细介绍替代型巨噬细胞激活的定义及其机制。
其次,在第三部分中,我们将探究影响替代型巨噬细胞激活的因素及其调节机制。
随后,在第四部分中,我们将探讨替代型巨噬细胞激活在免疫调节和治疗中的应用前景。
最后,在第五部分中,我们将对本文进行总结,并展望替代型巨噬细胞激活领域未来的研究方向。
1.3 目的本文旨在全面了解和探讨替代型巨噬细胞激活的定义、机制以及其与疾病之间的关联性。
同时,本文也将探索影响替代型巨噬细胞激活的因素及其调节机制,并重点关注替代型巨噬细胞激活在免疫调节和治疗中的应用前景。
通过这些内容的详细介绍,我们旨在提供更多关于替代型巨噬细胞激活领域的基础知识,并为未来相关领域的深入研究提供参考。
2. 替代型巨噬细胞激活的定义与机制2.1 替代型巨噬细胞激活的基本概念:替代型巨噬细胞是一种特殊类型的巨噬细胞,其功能和表型具有明显的区别于典型巨噬细胞(经典激活)状态下的巨噬细胞。
替代型巨噬细胞通常通过一系列特定信号转导途径的调控而被激活,并表现出不同于典型巨噬细胞的生物学行为。
替代型巨噬细胞激活的主要特征包括基因表达谱的变化、分泌因子的变化以及各种功能相关分子和受体表达谱上的改变。
2.2 替代型巨噬细胞激活的信号通路与途径:替代型巨噬细胞可以通过多条转录因子介导调控其驶向而实现激活状态。
其中,IL-4和IL-13等Th2类型淋巴细胞分泌的细胞因子是促使替代性悦升中介处理器产生高积极性的重要因素。
前额叶皮层的5-HT谷氨酸:一个新的抗精神病药的靶点
前额叶皮层的5-HT谷氨酸:一个新的抗精神病药的靶点张策【期刊名称】《山西医科大学学报》【年(卷),期】2000(031)0Z1【摘要】Activation of neocortical 5 hydroxytryptamine 2A (5-HT2A) re ceptors is thought to mediate the profoud psychominetic effects of hallucinogeni c drugs such as lysergic acid diethylamide(LSD).Coversely, blockade of neocortic al 5-HT2A receptor may be related to the thymoleptic effects of newly rele ased antidepressant especially atypical antipsychotic drugs. Electrophsiological experiments using in vitro rat slices of the medial prefrontal cortex has f ound that activation of 5-HT2A receptors results in glutamate release fr om thalamocortical terminals.In addition to activation of 5-HT2A receptors , metabotropic glutamate (mGluR),and neuropeptide (μ-opioid) receptors suppress this release of glutamate that is induced by 5-HT2A receptor activation. Recent clinical reports have found clear structural change in the prefrontal cor tex of depressed patients. Furthermore, a number of post-mortem studies of suici des from different laboratories have found an increased number of 5-HT2A b inding sites in certain regions of the prefrontal cortex.Thus,understanding the effects of 5-HT2A receptor activation in prefrontal cortex is likely to b e relevant for the development of antidepressant drugs.【总页数】3页(P60-62)【作者】张策【作者单位】美国耶鲁大学医学院精神病学系【正文语种】中文【中图分类】R338.2+5【相关文献】1.磷酸二酯酶7:一个新的抗炎免疫药物靶点研究进展 [J], 张莉;杜冠华2.IL-35/iTr35,一个新的急性肾损伤治疗靶点? [J], 钱铖;许贤林;嵇小兵3.上海交通大学科学家发现一个治疗神经源性疼痛的新的分子靶点 [J],4.驱动蛋白家族成员11作为一个新的caspase-1靶点嵌合到caspase-1依赖性的细胞死亡 [J], 王欣欣;郑桂玉;王芳;林立5.谷氨酸与精神分裂症:寻找新的抗精神病药物 [J], 唐劲松;陈晓岗因版权原因,仅展示原文概要,查看原文内容请购买。
camp signaling pathway γ-氨基丁酸
camp signaling pathway γ-氨基丁酸The γ-aminobutyric acid (GABA) signaling pathway is a critical inhibitory neurotransmitter system in the brain. It is involved in regulating neuronal excitability and plays important roles in various physiological processes, including motor control, cognition, and mood regulation.The GABA pathway begins with the synthesis of GABA from glutamate, which is catalyzed by the enzyme glutamate decarboxylase. GABA is then released from presynaptic terminals into the synaptic cleft, where it binds to GABA receptors on the postsynaptic membrane.There are two types of GABA receptors: GABA_A receptors and GABA_B receptors. GABA_A receptors are ion channels that allow chloride ions to flow into the cell, causing hyperpolarization and inhibiting neuronal activity. GABA_B receptors are metabotropic receptors coupled to G-proteins, which modulate the activity of ion channels and intracellular signaling pathways.The GABA_A receptor is a ligand-gated ion channel that consists of five subun its, typically consisting of two α, two β, and one γ subunit. Activation of GABA_A receptors by GABA leads to the opening of the chloride ion channel, resulting in an increase in chloride ion influx. This hyperpolarizes the postsynaptic membrane, making it less likely for an action potential to be generated.On the other hand, the GABA_B receptor is a G-protein coupled receptor (GPCR) that consists of two subunits, GABA_B1 andGABA_B2. Activation of GABA_B receptors by GABA leads to the activation of G-proteins, which then modulate the activity of ion channels, such as potassium channels, calcium channels, and others. This modulation of ion channels results in a decrease in neuronal excitability.Overall, the GABA signaling pathway plays a crucial role in maintaining the balance between excitation and inhibition in the brain. Dysregulation of this pathway has been implicated in various neurological and psychiatric disorders, such as epilepsy, anxiety, and depression.。
代谢型谷氨酸受体在胶质瘤发展中的作用及其作为潜在治疗靶点的研究进展
代谢型谷氨酸受体在胶质瘤发展中的作用及其作为潜在治疗靶
点的研究进展
孙中钱;李浩鸣;申强;张斌;张军臣
【期刊名称】《医学综述》
【年(卷),期】2024(30)9
【摘要】代谢型谷氨酸受体(mGluRs)是中枢谷氨酸能系统中重要的受体之一,其广泛参与调控突触传递、突触可塑性、神经兴奋性和抑制性平衡等生理过程。
mGluRs在介导胶质瘤的发生和发展中起重要作用,其介导的细胞内信号通路参与胶质瘤的进展、侵袭和复发过程,因此mGluRs作为胶质瘤的潜在药物靶点越来越受到关注。
其中,mGluR1和mGluR3的体外药物阻断已被证明对于胶质瘤细胞具有抗细胞增殖和迁移的作用,mGluR3拮抗剂对于替莫唑胺有增加敏感性的作用,mGluR4和mGluR8可能成为治疗恶性胶质瘤的潜在靶点,但需要广泛的研究验证。
未来应对不同类型的mGluRs在胶质瘤细胞中的作用进行深入探索,以为临床药物的研发提供理论依据。
【总页数】6页(P1080-1085)
【作者】孙中钱;李浩鸣;申强;张斌;张军臣
【作者单位】济宁医学院临床医学院;潍坊医学院管理学院;济宁医学院附属医院神经外科
【正文语种】中文
【中图分类】R739.41
【相关文献】
1.代谢型谷氨酸受体在突触可塑性中的作用研究进展
2.MCT1和MCT4在肿瘤发展中的作用及作为肾癌潜在治疗靶点的研究进展
3.Ⅱ、Ⅲ组亲代谢型谷氨酸受体激动剂逆转1-甲基-4-苯基吡啶离子抑制C6胶质瘤细胞摄取谷氨酸
4.代谢型谷氨酸受体可作为一种新型恶性胶质瘤治疗靶点(英文)
5.II、III组亲代谢型谷氨酸受体激动剂对脂多糖抑制C6胶质瘤细胞摄取谷氨酸的影响
因版权原因,仅展示原文概要,查看原文内容请购买。
- 1、下载文档前请自行甄别文档内容的完整性,平台不提供额外的编辑、内容补充、找答案等附加服务。
- 2、"仅部分预览"的文档,不可在线预览部分如存在完整性等问题,可反馈申请退款(可完整预览的文档不适用该条件!)。
- 3、如文档侵犯您的权益,请联系客服反馈,我们会尽快为您处理(人工客服工作时间:9:00-18:30)。
0304-3940/95/$09.50 0 1995 Elsevier Science Ireland Ltd. All rights reserved SSDI 0304-3940(95)11’ 753-G
174
K. Maiese et al. /Neuroscience
Letters 194 (1995) 173-l 76
medium and incubated in dissociation medium containing trypsin inhibitor (10-20 U/ml) three times, for 5 min each. The cells were washed in growth medium (Leibovitz’ s L- 15 Medium (Gibco BRL, Gaithersburg, MD) with 6% sterile rat serum (Bioproducts for Science, Indianapolis, IN), 150 mM NaHCOs, 2.25 mg/ml transferrin, 2.5 ,ug/ml insulin, 10 nM progesterone, 90pM putrescine, 15 nM selenium, 35 mM glucose, 1 mM Lglutamine, penicillin (50 U/ml), streptomycin (5Opg/ml), and supplemented with vitamins. The dissociated cells were plated at a density of approximately 15 x lo5 cells/ mm* in 35 mm* polylysine-laminin coated plates (Falcon Labware, Lincoln Park, NJ). Cells were maintained in growth medium at 37°C in a humidified atmosphere of 5% CO2 and 95% room air. All experiments were performed with cultured neurons that had been in culture for 2-3 weeks. Non-neuronal cells accounted for less than 20% of the total cell population. NO administration was performed by replacing the culture media with media containing 3OOpM sodium
Neuroscience Letters 194 (1995) 173-176
Activation of the metabotropic glutamate receptor is neuroprotective during nitric oxide toxicity in primary hippocampal neurons of rats
60 -
?? 30
L-AP4 and SNP
SNP Only -D
20
0 I 1 .oo (10 04
1
1.50 (30 W
2.00 (100 114
2 50 (250 PM)
3 00 (1000 PM)
Log [L-AP4J @Ml Fig. 1. Reduction in NO toxicity by L-AP4. Hippocampal cultures were pretreated with increasing concentrations of L-API1 1 h prior to a 5-min exposure of sodium nitroprusside (SNP) (3OOpM) (closed symbols) or in the absence of SNP (open symbols). Each data point represents the mean and SEM of n = 6 determinations (culture plates) from three separate experimental preparations. Neuronal survival was based on the percentage of the total number of neurons (viable + non-viable) and determined by trypan blue exclusion 24 h following exposure to SNP or 25 h following exposure to LAP4 without SNP administration. In cultures receiving only SNP, 25 * 7% (mean and SEM of n = 6 determinations (culture plates) from three separate experimental preparations) of the neurons were viable.
duced in hippocampal astrocytes [3] and mutant mice deficient in neuronal NOS activity have been shown to experience reduced infarct volumes [7]. Glutamate receptors have recently been reported to stimulate NO production in neurons [6,16]. Modulation of metabotropic glutamate receptor activity in the hippocampus [5] may represent one of the pathways that mediates NO toxicity during cerebral ischemia. Therefore, we examined whether the neuroprotective effect of mGluR activation was dependent upon the NO pathway. We focused on the mGluR4 receptor since this subtype has been associated with the inhibition of calcium currents [23] which could subsequently alter the generation and toxicity of NO [ 151. The hippocampi were obtained from l-day-old Sprague-Dawley rat pups and incubated in dissociation medium (90 mM Na$O+ 30 mM KzS04, 5.8 mM MgCl*, acid, and 1 mM 0.25 mM CaC12, 10 mM kynurenic HEPES with pH adjusted to 7.4) containing papain (10 U/ml) and cysteine (3 mmol/l) for two 20-min periods. The hippocampi were then rinsed in dissociation
Received 11 April 1995; revised version received 12 June 1995; accepted 12 June 1995
Abstract Metabotropic glutamate receptors (mGluRs) can influence neuronal survival and have been shown to be neuroprotective during glutamate toxicity in retinal cells and in cortical neurons. The mechanisms that mediate protection by this group of receptors are not clear. Since nitric oxide (NO) production can lead to neuronal cell death during excessive glutamate release, we examined whether neuronal survival was directly linked to mGluR activity and the NO pathway. Treatment with the mGluR4 receptor subtype agonist, L(+)-2-amino-4-phosphonobutyric acid, in hippocampal cell cultures protected neurons during NO exposure. Treatment with L-(+)-2amino-3-phosphonopropionic acid, an antagonist of the mGluRI receptr of inositol trisphosphate formation, did not significantly alter neuronal survival during NO administration. We conclude that activation of the mGluR4 receptor protects hippocampal neurons from NO toxicity and that the mechanism of NO induced neurodegeneration does not appear to involve inhibition of the mGluR1 receptor subtype activity or the phosphoinositide system.