Mitochondrial Reactive Oxygen Species. Contribution to Oxidative Stress and Interorganellar
- 格式:pdf
- 大小:131.54 KB
- 文档页数:10
线粒体氧化应激线粒体形态Mitochondrial oxidative stress is a phenomenon that occurs when there is an imbalance between the production of reactive oxygen species (ROS) and the ability of the cell to detoxify them or repair the resulting damage. This can lead to damage to cellular components and is implicated in a range of diseases, including neurodegenerative disorders, cardiovascular diseases, and cancer. One of the key consequences of mitochondrial oxidative stress is damage to the mitochondrial DNA (mtDNA), which can lead to mutations and deletions that affect the function of the mitochondria and, in turn, the overall health of the cell.The morphology of mitochondria is also affected by oxidative stress. Under normal conditions, mitochondria have a tubular, elongated shape, but when exposed to oxidative stress, they undergo fragmentation and become more spherical. This change in morphology is thought to be a protective response, as it allows damaged mitochondria to be segregated and targeted for removal by the cell'squality control mechanisms. However, excessivefragmentation can also lead to impaired mitochondrial function and cell death.In addition to the structural and functional changes in mitochondria, oxidative stress can also lead to the activation of cell death pathways. This can occur through the release of pro-apoptotic factors from the mitochondria, such as cytochrome c, which triggers a cascade of eventsthat ultimately lead to cell death. In this way, mitochondrial oxidative stress can have profound effects on cell survival and tissue integrity.One of the major sources of mitochondrial oxidative stress is the electron transport chain (ETC), which is responsible for generating the majority of the cell'senergy in the form of adenosine triphosphate (ATP). During this process, electrons can leak from the ETC and reactwith molecular oxygen to form superoxide, a type of ROS. Under normal conditions, the cell has antioxidant defense systems in place to neutralize these ROS and prevent damage. However, when the production of ROS exceeds the capacity ofthese defenses, oxidative stress occurs.There are several factors that can contribute to mitochondrial oxidative stress, including environmental toxins, metabolic dysfunction, and genetic mutations. For example, exposure to environmental pollutants such as heavy metals and pesticides can increase ROS production and overwhelm the cell's antioxidant defenses. Similarly, metabolic disorders such as diabetes and obesity can lead to increased ROS production due to dysregulation ofcellular metabolism. Finally, mutations in genes encoding for antioxidant enzymes or mitochondrial proteins can also predispose individuals to mitochondrial oxidative stress.In conclusion, mitochondrial oxidative stress and its effects on mitochondrial morphology and function have far-reaching implications for human health. Understanding the mechanisms underlying mitochondrial oxidative stress and developing strategies to mitigate its effects are critical for the prevention and treatment of a wide range of diseases. By targeting the sources of oxidative stress and enhancing the cell's antioxidant defenses, it may bepossible to alleviate the burden of mitochondrial dysfunction and improve overall cellular and tissue health.。
枸橼酸坦度螺酮组合物通过抑制神经元氧化损伤和调节小胶质细胞活化发挥抗抑郁作用邓岚1,曾娟1,王征琴1,唐新新1,陈刚2,吴建明1,吴安国11.西南医科大学药学院(泸州646000);2.四川科瑞德制药股份有限公司(泸州646000)【摘要】目的筛选不同组成的枸橼酸坦度螺酮组合物(tandospirone citrate compositions,TCCs ),以评估其改善小鼠抑郁症行为的效果,并探究枸橼酸坦度螺酮的神经保护作用。
方法采用动物行为实验和体外细胞实验的方法。
在动物实验中,通过强迫游泳测试(forced swim-ming test,FST )和尾悬挂测试(tail suspension test,TST ),评估五种TCCs 对小鼠抑郁样行为的影响。
在体外细胞实验中,研究表现最佳的组合物TCC-4在抑制过氧化氢(hydrogen peroxide,H 2O 2)诱导的PC-12细胞损伤和脂多糖(lipopolysaccharide,LPS )诱导的BV-2细胞神经炎症的作用。
结果在FST 和TST 实验中,TCCs 显著减少了小鼠的不动时间(P <0.05),显示出其抗抑郁样效果。
体外细胞实验中发现,表现最佳的TCC-4能有效抑制H 2O 2诱导的PC-12细胞死亡、活性氧(reactive oxygen species,ROS )的产生及线粒体膜电位(mitochondrial membrane po-tential,MMP )的降低(P <0.001),且减轻了LPS 诱导的BV-2细胞激活和吞噬作用(P <0.001)。
结论TCCs,尤其是TCC-4,显示出良好的抗抑郁效果。
其作用机制可能与抑制神经元的氧化损伤和调节小胶质细胞的活化有关。
这些发现为TCC-4作为新型抗抑郁药物的潜力提供了支持,并为其在临床应用的进一步研究奠定了理论基础。
【关键词】抑郁症;枸橼酸坦度螺酮组合物;氧化应激;神经炎症;小胶质细胞【中图分类号】R964文献标志码:A DOI :10.3969/j.issn.2096-3351.2024.02.014Tandospirone citrate composition exerts antidepressant effects by inhibitingneuronal oxidative damage and microglial activationDENG Lan 1,ZENG Juan 1,WANG Zhengqin 1,TANG Xinxin 1,CHEN Gang 2,WU Jianming 1,WU Anguo 11.School of Pharmacy ,Southwest Medical Univisity ,Luzhou 646000,China ;2.Sichuan Credit Pharmaceutical CO.,Ltd.,Luzhou 646000,China【Abstract 】Objective Different tandospirone citrate compositions (TCCs )were screened to assess their effectiveness in improv‑ing depressive behaviors in mice and to explore the neuroprotective effects of the most effective combinations.Methods Animal behav‑ioral experiments and in vitro cellular experiments were used.In animal experiments ,the effects of five TCCs on depression-like behav‑iors in mice were evaluated by the forced swim test (FST )and tail suspension test (TST ).In vitro experiments were conducted to inves‑tigate the effects of TCC-4,which exhibited the best performance ,on inhibiting hydrogen peroxide (H 2O 2)-induced damage in PC-12cells and lipopolysaccharide (LPS )-induced neuroinflammation in BV-2cells.Results In FST and TST experiments ,TCCs signifi‑cantly reduced the resting time of mice (P <0.05),showing their antidepressant-like effects.In H 2O 2-induced PC-12cells ,the best-performing TCC-4was found to effectively inhibit cell death ,reactive oxygen species (ROS )production ,and mitochondrial membrane potential (MMP )reduction (P <0.001).Additionally ,TCC-4could attenuate LPS-induced BV-2cell activation and phagocytosis (P <0.001).Conclusion TCCs ,especially TCC-4,showed favorable antidepressant effects.The mechanism of action might be related to the inhibition of oxidative damage in neurons and the regulation of microglia activation.These findings provided support for the poten‑tial of TCC-4as a novel antidepressant drug and laid the foundation for further studies on its clinical application.【Key words 】Depression ;Tandospirone citrate compositions ;Oxidative stress ;Neuroinflammation ;Microglia基金项目:国家自然科学基金青年项目(81903829)通信作者:吴安国,E-mail:****************.cn引用本文:邓岚,曾娟,王征琴,等.枸橼酸坦度螺酮组合物通过抑制神经元氧化损伤和调节小胶质细胞活化发挥抗抑郁作用[J].西南医科大学学报,2024,47(2):166-171.DOI:10.3969/j.issn.2096-3351.2024.02.014抑郁症作为全球性的公共卫生问题,严重影响数百万患者的生活质量,并对社会经济造成了沉重的负担[1]。
晚期糖基化终产物诱导人主动脉内皮细胞氧化应激损伤及线粒体功能紊乱丁银慧;孙竞;钱元霞;李芸子;高静;陈赟【摘要】目的:观察晚期糖基化终产物(advanced glycation end products,AGEs)对内皮细胞氧化应激水平和线粒体功能的影响,以探讨糖尿病血管内皮细胞功能障碍可能的发生机制.方法:以不同质量浓度(50,100 μg/mL)的AGE修饰的牛血清白蛋白(AGE-bovine serum albumin,AGE-BSA)作用于人主动脉内皮细胞(human aortic endothelial cells,HAECs) 48 h,CCK-8法测定HAECs的增殖能力,黄嘌呤氧化酶法测定超氧化物歧化酶(superoxide dismutase,SOD)活力,应用DCFH-DA 探针检测细胞内活性氧(reactive oxygen species,ROS)的水平;JC-1法检测线粒体膜电位(mitochondrial membrane potential,MMP),荧光素-荧光素酶法和Clark 氧电极法测定细胞中ATP含量及细胞耗氧率.结果:AGE-BSA能显著性地抑制HAECs增殖,增加细胞内ROS水平,降低SOD活力,且高质量浓度作用更加明显.同时,AGE-BSA还能使MMP下降、ATP生成及耗氧减少.结论:AGE-BSA诱导HAECs产生氧化应激损伤,其机制与其造成线粒体功能紊乱相关.%Objective:To investigate whether advanced glycation end products (AGEs) can induce cell injury and mitochondrial dysfunction in human aortic endothelial cells (HAECs).Methods:HAECs were treated with increasing concentrations (50,1 00μg/mL) of AGE-bovine serum albumin (AGE-BSA) for 48 h.The proliferative inhibition of HAECs was measured by CCK-8 method.Contents of ATP and activity of superoxide dismutase (SOD) were determined by the luciferase assay and SOD kits.Reactive oxygen species (ROS) were determined by DCFH-DA staining.Mitochondrial membrane potential(MMP) was observed with JC-1 staining.Oxygen utilization was measured polarographically with a Clark oxygen electrode.Results:Compared with control group,AGE-BSA (50,100 μg/mL) significantly reduced HAECs proliferation.AGE-BSA significantly increased the levels of ROS,while decreased the activity of SOD compared to the controlgroup.Importantly,AGE-BSA-mediated oxidative stress was followed by a collapse of mitochondrial membrane potential (MMP),the inhibition of ATP generation,and the down-regulation of oxygen utilization.Conclusion:AGE-BSA induced oxidative stress in HAECs.Moreover,AGE-BSA-induced HAECs injury was related to mitochondrial dysfunction.【期刊名称】《江苏大学学报(医学版)》【年(卷),期】2014(024)003【总页数】5页(P185-189)【关键词】晚期糖基化终产物;氧化应激;线粒体;人主动脉内皮细胞【作者】丁银慧;孙竞;钱元霞;李芸子;高静;陈赟【作者单位】江苏大学药学院,江苏镇江212013;江苏大学药学院,江苏镇江212013;江苏大学药学院,江苏镇江212013;江苏大学药学院,江苏镇江212013;江苏大学药学院,江苏镇江212013;南京大学医学院附属鼓楼医院泌尿外科,江苏南京210008【正文语种】中文【中图分类】R365.587糖尿病是威胁人类健康的主要疾病之一,其慢性并发症可涉及全身所有组织和器官,其中血管(包括大血管和微血管)病变和神经病变表现最为明显和突出。
氧化应激的指标
氧化应激的指标有很多,以下列出了一些常见的指标:
1. ROS(Reactive Oxygen Species,活性氧):活性氧是氧化应激的主要物质,可以通过荧光染料或自由基捕捉剂等方法检测。
2. MDA(Malondialdehyde,丙二醛):MDA是膜脂过氧化反应的产物,是氧化应激的重要指标之一,可以通过比色法或荧光法检测。
3. SOD(Superoxide Dismutase,超氧化物歧化酶):SOD是一种重要的抗氧化酶,可以检测SOD的活性或基因表达水平来评估氧化应激的程度。
4. CAT(Catalase,过氧化氢酶):CAT也是一种重要的抗氧化酶,可以检测其活性或基因表达水平来评估氧化应激的程度。
5. GSH(Glutathione,谷胱甘肽):GSH是一种重要的抗氧化剂,可以通过比色法或荧光法等方法检测。
6. 线粒体膜电位(Mitochondrial membrane potential,MMP):MMP是线粒体功能的重要指标,氧化应激可改变MMP,可通过荧光染料检测。
7. DNA氧化损伤:DNA氧化损伤是氧化应激的重要标志之一,可以通过单核苷酸多态性(Single Nucleotide Polymorphism,SNP)或8-OHdG 等指标检测。
8. 炎症因子:氧化应激可引起炎症反应,相关炎症因子如TNF-α(Tumor Necrosis Factor-α)和IL-6(Interleukin-6)等可以作为氧化应激的指标之一。
线粒体氧化应激及其线粒体营养素干预机制摘要:线粒体在生物氧化和能量转换的过程中会产生活性氧,当活性氧的生成与机体抗氧化防御系统之间存在不平衡时,线粒体就会发生氧化应激。
线粒体氧化应激导致线粒体能量代谢失调,进一步损伤线粒体,从而促进神经退行性疾病的发生,发展。
研究表明,线粒体营养素既可以增强抗氧化防御系统功能,又能够减少线粒体活性氧的生成,从而修复线粒体的氧化损伤,进而改善线粒体的结构和功能。
本文将从线粒体氧化应激和线粒体营养素干预机制两方面做以综述。
关键词:线粒体氧化应激活性氧烟酸硫辛酸硫辛酰胺线粒体是真核动物细胞进行生物氧化和能量转换的主要场所,细胞生命活动所需能量的80%是由线粒体提供的,因此,有人将线粒体称为细胞的“动力工厂”。
线粒体生物氧化和能量转换的过程中伴随着活性氧(reactive oxygen species,ROS)的产生。
过量的ROS会引起线粒体损伤,促进神经退行性疾病的发生,发展。
由氧化应激引起的线粒体损伤是衰老及神经退行性病变的主要原因,并且严重影响运动能力。
线粒体损伤可导致关键的线粒体酶功能障碍。
酶的功能障碍主要是由于底物和辅酶的结合不足,而这种结合不足在补充足够的底物或辅酶及其前体后可以得到改善,长期补充线粒体营养素(mt-nutrients)可以有效地保护线粒体功能的完整,修复线粒体的损伤。
Liu[1]等把线粒体营养的功能定义为:①可以提高线粒体酶底物和辅酶的水平;②诱导二相酶增强细胞内的抗氧化防御能力;③清除自由基及防止氧化剂的生成;④修复线粒体膜损伤。
现就线粒体氧化应激和线粒体营养素对其干预机制两方面做简要综述。
1 线粒体氧化应激氧化应激是指活性氧生成与抗氧化防御系统之间的不平衡状态,氧化应激可在活性氧生成超过抗氧化防御系统时或者在抗氧化剂活性降低时发生。
众所周知,线粒体是真核动物细胞进行生物氧化和能量转换的主要场所,但在线粒体生物氧化和能量转化的过程中会产生活性氧(reactive oxygen species,ROS),由于活性氧的活性非常高,过量的活性氧会进攻线粒体DNA及线粒体内蛋白质,脂类等生物大分子物质,从而损伤线粒体使其能量合成受到障碍,最终导致线粒体功能下降,线粒体氧化应激导致线粒体能量代谢失调,进一步损伤线粒体,从而促进神经退行性疾病的发生,发展。
线粒体氧化应激和细胞氧化应激英文回答:Mitochondrial oxidative stress refers to the accumulation of reactive oxygen species (ROS) within the mitochondria, which can lead to damage to the mitochondrial DNA, proteins, and lipids. This can disrupt the normal functioning of the mitochondria and contribute to the development of various diseases, including neurodegenerative disorders, cardiovascular diseases, and cancer.ROS are produced as natural byproducts of cellular metabolism, particularly during the process of oxidative phosphorylation in the mitochondria. However, excessive production of ROS or impaired antioxidant defense mechanisms can result in an imbalance between ROS production and removal, leading to oxidative stress.Mitochondrial oxidative stress can be caused by variousfactors, including environmental toxins, radiation, inflammation, and genetic mutations. These factors can increase the production of ROS or impair the function of antioxidant enzymes, such as superoxide dismutase, catalase, and glutathione peroxidase, which are responsible for neutralizing ROS.The consequences of mitochondrial oxidative stress are diverse and can affect various cellular processes. For example, it can lead to mitochondrial dysfunction, characterized by a decrease in ATP production, impaired calcium homeostasis, and increased production of ROS. This can further exacerbate oxidative stress and create avicious cycle of mitochondrial damage.Cellular oxidative stress, on the other hand, refers to the accumulation of ROS within the entire cell, not justthe mitochondria. It can be caused by similar factors as mitochondrial oxidative stress, but the consequences can be more widespread. Cellular oxidative stress can lead to damage to cellular components, including DNA, proteins, and lipids, and can result in cellular dysfunction and celldeath.To counteract oxidative stress, cells have developed various defense mechanisms, including antioxidant enzymes, such as superoxide dismutase, catalase, and glutathione peroxidase, as well as non-enzymatic antioxidants, such as vitamins C and E, glutathione, and coenzyme Q10. These antioxidants help neutralize ROS and prevent oxidative damage.In conclusion, mitochondrial oxidative stress and cellular oxidative stress are both significant factors in the development of various diseases. Understanding the underlying mechanisms and developing strategies to mitigate oxidative stress can have important implications for the prevention and treatment of these diseases.中文回答:线粒体氧化应激是指线粒体内反应性氧化物质(ROS)的积累,这可能导致线粒体DNA、蛋白质和脂质的损伤。
Product InformationStorage upon receipt:• ≤–20ºC• Protect from light • Desiccate• Avoid freeze-thaw cyclesEx/Em: 510/580 nm Note:To preserve the activity of the MitoSOX™ Red reagent, minimize exposure to air.Quick FactsRevised: 03–March–2005IntroductionMitochondrial superoxide is generated as a byproduct of oxi-dative phosphorylation. In an otherwise tightly coupled electron transport chain, approximately 1–3% of mitochondrial oxygen consumed is incompletely reduced; those “leaky” electrons can quickly interact with molecular oxygen to form superoxide anion, the predominant reactive oxygen species (ROS) in mitochon-dria.1–4 Increases in cellular superoxide production have been implicated in cardiovascular diseases, including hypertension, atherosclerosis, and diabetes-associated vascular injuries,5–7 as well as in neurodegenerative diseases such as Parkinson ʼs, Alzheimer ʼs, and amyotrophic lateral sclerosis (ALS).8–12The assumption that mitochondria serve as the major intracel-lular source of ROS has been based largely on experiments with isolated mitochondria rather than direct measurements in living cells. MitoSOX ™ Red mitochondrial superoxide indicator is a novel fluorogenic dye for highly selective detection of super-oxide (Figure 1) in the mitochondria of live cells. MitoSOX™ Red reagent is live-cell permeant and is rapidly and selec-tively targeted to the mitochondria. Once in the mitochondria, MitoSOX™ Red reagent is oxidized by superoxide and exhibits red fluorescence. MitoSOX™ Red reagent is readily oxidized by superoxide but not by other ROS- or reactive nitrogen species (RNS)–generating systems, and oxidation of the probe is pre-vented by superoxide dismutase. The oxidation product becomes highly fluorescent upon binding to nucleic acids.This reagent may enable researchers to distinguish artifacts of isolated mitochondrial preparations from direct measurements of superoxide generated in the mitochondria of live cells. It may also provide a valuable tool in the discovery of agents that modu-late oxidative stress in various pathologies.MaterialsContentsMitoSOX™ Red mitochondrial superoxide indicator (MW = 759) is supplied in 10 vials, each containing 50 μg.Storage and HandlingUpon receipt, the MitoSOX™ Red reagent should be stored upright, desiccated, and protected from light at ≤–20˚C. Avoid freeze-thaw cycles. MitoSOX™ Red reagent is packaged with an oxygen scavenging pouch that will extend the shelf life of the product. After removing vials for use, seal the remaining vials into the pouch to preserve the activity of the reagent. Vialsshould be allowed to warm to room temperature before opening. When stored properly, components should be stable for at least 6 months. Note: MitoSOX™ Red reagent is a derivative of ethid-ium bromide and should be treated with the appropriate caution.MitoSOX™ Red mitochondrial superoxide indicator *for live-cell imaging* (M36008)Figure 1. Selectivity of the MitoSOX™ Red mitochondrial superoxide indicator. Cell-free systems were used to generate a variety of reactive oxygen species (ROS) and reactive nitrogen species (RNS); each oxidant was then added to a separate 10 µM solution of MitoSOX™ Red reagent and incubated at 37˚C for 10 minutes. Excess DNA was added (unless otherwise noted) and the samples were incubated for an additional 15 minutes at 37˚C before fluorescence was measured. The Griess nitrite determina-tion kit (for nitric oxide, peroxynitrite, and nitrite standards only; gray bars) anddihydrorhodamine 123 (DHR 123; white bars) were employed as positive controls for oxidant generation. Superoxide dismutase (SOD), a superoxide scavenger, was used as a negative control for superoxide. The results show that the MitoSOX™ Red probe (black bars) is readily oxidized by superoxide but not by the other oxidants.副产品线粒体超氧化物作为氧化磷酸化的副产物产生。
线粒体功能障碍的原因及其对肿瘤作用的研究进展李琪;陈斌(综述);秦泽莲(审校)【摘要】线粒体是机体能量产生的主要细胞器,在有氧呼吸、物质代谢、氧化应激、凋亡、Ca2+稳态等方面发挥重要的功能。
越来越多研究表明线粒体功能障碍与肿瘤密切相关,线粒体代谢异常、活性氧增多、线粒体基因突变、Ca2+超载、凋亡异常影响多种肿瘤发生、生长、侵袭、转移。
本文就线粒体功能障碍发生机制及其与肿瘤的关系进行文献总结。
%As a main cellular organelle for bioenergy production , the mitochondrion plays a pivotal role in aerobic respiration , substance metabolism , oxidative stress , apoptosis and calcium homeostasis .Increasingly studies have shown a close relationship between mitochondrial dysfunction and cancer .Mitochondrial metabolic disturbance , reactive oxygen species ( ROS ) increase, mitochondrial gene mutation , calcium overload and abnormal apoptosis can influence tumorigenesis , growth, invasiveness and metastasis of multipletumors .We aimed to summarize the mechanisms and influences of mitochondrial dysfunction on cancer .【期刊名称】《中国微创外科杂志》【年(卷),期】2016(016)012【总页数】5页(P1150-1154)【关键词】线粒体;基因;功能障碍;肿瘤【作者】李琪;陈斌(综述);秦泽莲(审校)【作者单位】北京大学第三医院成形外科,北京 100083;北京大学第三医院成形外科,北京 100083;北京大学第三医院成形外科,北京 100083【正文语种】中文线粒体是细胞进行有氧呼吸的主要场所,在有氧条件下,通过一系列生化反应和电子传递,将糖和脂肪酸氧化过程中释放的自由能转变为ATP中的化学能。
本外文翻译含中英文,前面英文,后面中文。
Mobile Phone Radiation Induces Reactive OxygenSpecies Production and DNA Damage in HumanSpermatozoa In VitroAbstractBackgroundIn recent times there has been some controversy over the impact of electromagnetic radiation on human health. The significance of mobile phone radiation on male reproduction is a key element of this debate since several studies have suggested a relationship between mobile phone use and semen quality. The potential mechanisms involved have not been established, however, human spermatozoa are known to be particularly vulnerable to oxidative stress by virtue of the abundant availability of substrates for free radical attack and the lack of cytoplasmic space to accommodate antioxidant enzymes. Moreover, the induction of oxidative stress in these cells not only perturbs their capacity for fertilization but also contributes to sperm DNA damage. The latter has, in turn, been linked with poor fertility, an increased incidence of miscarriage and morbidity in the offspring, including childhood cancer. In light of these associations, we have analyzed the influence of RF-EMR on the cell biology of human spermatozoa in vitro.Principal FindingsPurified human spermatozoa were exposed to radio-frequency electromagnetic radiation (RF-EMR) tuned to 1.8 GHz and covering a range of specific absorption rates (SAR) from 0.4 W/kg to 27.5 W/kg. In step with increasing SAR, motility and vitality were significantly reduced after RF-EMR exposure, while the mitochondrial generation of reactive oxygen species and DNA fragmentation were significantly elevated (P<0.001). Furthermore, we also observed highly significant relationships between SAR, the oxidative DNA damage bio-marker, 8-OH-dG, and DNA fragmentation after RF-EMR exposureConclusionsRF-EMR in both the power density and frequency range of mobile phones enhances mitochondrial reactive oxygen species generation by human spermatozoa, decreasing the motility and vitality of these cells while stimulating DNA base adduct formation and, ultimately DNA fragmentation. These findings have clear implications for the safety of extensive mobile phone use by males of reproductive age, potentially affecting both their fertility and the health and wellbeing of their offspringlistIntroduction (1)Results (2)RF-EMR disrupts human sperm motility and vitality and induces intracellular reactive oxygen species (ROS) production (2)RF-EMR has a negative impact on human spermatozoa over a range of SAR values (4)Reactive Oxygen Species are central to the RF-EMR response (5)RF-EMR induces oxidative DNA damage (8-OH-dG) (6)RF-EMR induces DNA fragmentation in human spermatozoa (7)Discussion (9)Methods (12)Ethics Statement (12)Reagents and Solutions (12)Human spermatozoa (12)Radio Frequency Electromagnetic Radiation and Waveguide (13)Dihydroethidium Assay (14)MitoSOX Red (MSR) Assay (15)Assay for 8-hydroxy-2′-deoxyguanosine (8-OH-dG) (15)TUNEL Assay (16)Analysis by Flow Cytometry (16)Statistics (17)References (17)IntroductionMale infertility is a distressingly common condition affecting about 1 in 20 of the male population [1]. In a majority of cases, the male partner produces sufficient numbers of spermatozoa to achieve fertilization but there are functional defects in these cells that prevent conception from occurring [2]. Despite several decades of research, the causes of such functional deficiencies in human spermatozoa remain largely unresolved. However, one contributory factor that has recently emerged is the quality of the sperm DNA delivered to the oocyte at the moment of fertilization [3]. Fragmentation of DNA in the male germ line has been associated with impaired fertilization, poor embryonic development, high rates of miscarriage and an increased incidence of morbidity in the offspring, including childhood cancer [3], [4]. In view of the seriousness of these clinical outcomes, attention has recently focused on the environmental and genetic factors that might be involved in the aetiology of DNA damage in the male germ line.These investigations have suggested that one of the environmental factors potentially involved in the etiology of DNA damage in human spermatozoa is an increased exposure to radio-frequency electromagnetic radiation (RF-EMR) emitted from mobile phones. This association was initially suggested by an epidemiological study which found negative correlations between mobile phone usage and various attributes of semen quality, particularly motility [5]. This was immediately followed by an experimental study involving exposure of male mice to RF-EMR, which revealed a significant impact on the integrity of both the mitochondrial and nuclear genomes [6]. Recently, the negative impact of mobile phone usage on semen quality in human males was confirmed in a study that found the duration of exposure to be correlated with defects in sperm count, motility, viability, and normal morphology [7]. In light of these data, there is now an urgent need to determine whether exposure of human spermatozoa to RF-EMR can also induce DNA damage and to resolve the cellular mechanisms involved.Several studies have found an association between human health and exposure to RF-EMR, with emphasis on a range of clinical conditions including childhood leukaemia, brain tumours, genotoxicity and neurodegenerative disease [8], [9]. While the cellular mechanisms underpinning these effects have not been completely resolved, it has been suggested that oxidative stress could be a key factor [10]. However, extensive analysis of the importance of oxidative stress in mediating the pathological effects of RF-EMR has generated conflicting results, possibly due to differences in the fundamental redox susceptibility of the cell lines employed in these analyses [11]. In this context, it is significant that human spermatozoa are uniquely sensitive to oxidative stress for a variety of reasons. Firstly, these cells arelargely devoid of the cytoplasm that in somatic cells houses the antioxidant enzymes that offer a first line of defense against free radical attack [12]. Secondly, these cells possess abundant targets for the induction of peroxidative damage including polyunsaturated fatty acids and DNA [12]–[14]. Thirdly, these cells are professional generators of reactive oxygen species, that appear to emanate largely from the sperm mitochondria and, possibly, plasma membrane NAD(P)H oxidases [15], [16]. Thus if any cell type would be vulnerable to the oxidative stress reportedly generated on exposure to RF-EMR, it would be human spermatozoa.In light of these considerations, we have conducted a careful analysis of the biological consequences of exposing human spermatozoa to RF-EMR. The study design involved overnight exposure to RF-EMR at a defined frequency (1.8 GHz), over a range of SAR values that both covered the emission characteristics of mobile phones and generated sufficient dose-response data to shed light on the underlying pathophysiological mechanisms. Moreover, the temperature of the incubations was maintained at 21°C to avoid any secondary heating effects. The results clearly demonstrate that exposure to this type of radiation not only stimulates free radical generation by the sperm mitochondria but also creates a state of oxidative stress characterized by the formation of oxidative base adducts and DNA fragmentation. These data clearly have important implications for the safety of mobile phone use and highlight the potential importance of RF-EMR in the etiology of male infertility and childhood disease.ResultsRF-EMR disrupts human sperm motility and vitality and induces intracellular reactive oxygen species (ROS) productionIn an initial experiment, functional human spermatozoa isolated from the high density region of Percoll gradients and suspended in BWW medium were exposed to RF-EMR at an SAR of 27.5 W/kg. This exposure induced a highly significant decline in both vitality (p<0.001; Figure 1A) and motility (p<0.01; Figure 1B) compared with the unexposed controls. Exposed spermatozoa also produced significantly higher amounts of ROS than background levels as measured by both the dihydroethidium (DHE) (p<0.001; Figure 1C) and MitoSOX red (MSR) probes (p<0.001; Figure 1D) suggesting that free radical generation had been initiated as a consequence of RF-EMR and that the mitochondria were significantly involved in this response.Figure 1. RF-EMR exposure decreases motility and vitality of human sperm while also inducing intracellular ROS.Percoll-purified spermatozoa (5×106 cells) were suspended in 1 ml BWW in a 35 mm Petri dish and placed within the waveguide while control cells placed outside the waveguide. A frequency of 1.8 GHz at a SAR of 27.5 W/kg was used and all samples were incubated for 16 h at 21°C. A, Sperm vitality was significantly reduced from the control value of 82%±4% to 29%±4% for the exposed cells (***p<0.001). B, Sperm motility was also significantly reduced from the control value of 82%±4% to 28%±1% (**p<0.01). C, ROS production was increased after RF-EMR exposure such that 28%±1% of the cells were producing ROS, while only 7%±0.4% of the controls contributed to ROS production (***p<0.001). D, 24%±1% of the exposed cells generated mitochondrial ROS, while the only 12%±1% of the control cells produced ROS from this source (***p<0.001). All results are based on 4 independent samples. doi:10.1371/journal.pone.0006446.g001RF-EMR has a negative impact on human spermatozoa overa range of SAR valuesIn light of these results we then extended the range of SAR values over which the consequences of RF-EMR radiation were examined (0.4 W/kg–27.5 W/kg) to include the values covered by conventional mobile phones (0.5 W/kg–1.5 W/kg).High quality spermatozoa selected in discontinuous Percoll gradients displayed a decline in both vitality and motility after exposure to RF-EMR in a dose- dependent manner. The control populations maintained an average vitality of 89%; however, significant reductions in vitality were observed at exposure levels as low as 1.0 W/kg (p<0.01) (Figure 2A). Similarly, the control populations maintained motilities at an average of 86% over the incubation period, however after exposure to RF-EMR at levels of 1.0 W/kg, motility was observed to significantly decrease to 68% (p<0.05) and decreased still further at higher SAR exposures (Figure 2B).Figure 2. RF-EMR exposure reduces motility and vitality of human spermatozoa, in an SAR dependent manner.Percoll-purified spermatozoa (5×106 cells) were suspended in 1 ml BWW in a 35 mm Petri dish and placed within the waveguide while control cells (closed circles) were placed outside the waveguide. Cells in the waveguide were exposed to 1.8 GHz RF-EMR at SAR levels of 0.4, 1.0 2.84.3 10.1 and 27.5 W/kg (open circles) for 16 h at 21°C. Both vitality and motility were reduced ina dose dependent manner. A, Vitality was significantly reduced at a SAR of 1.0 W/kg from 89%±3% to 65%±1% (**p<0.01). B, Motility was also significantly reduced at a SAR of 1.0 W/kg from 86%±2% to 68%±2% (*p<0.05). All results are based on 4 independent samples.doi:10.1371/journal.pone.0006446.g002Reactive Oxygen Species are central to the RF-EMR response Exposure of human spermatozoa to RF-EMR over a range of SAR levels resulted in a dose-dependent activation of ROS generation, as detected by the DHE probe (Figure 3A). In this analysis, a significant increase in ROS positive cells was observed after exposure at 1.0 W/kg (p<0.05); thereafter ROS production rose rapidly with SAR values up to 4.3 W/kg and then began to plateau reaching a peak of 30% at the highest exposure levels assessed (Figure 3A). To determine whether such increases in ROS production might originate from the sperm mitochondria, MSR was employed as a probe. Spermatozoa exposed to increasing levels of RF-EMR, generated a significant, dose-dependent increase in ROS generation by the mitochondria. The response rose rapidly following RF-EMR exposure reaching statistical significance (p<0.001) at an SAR value 2.8 W/kg at which point 16% of the exposed cells were MSR positive. At SAR values above 4.3 W/kg, RF-EMR induced mitochondrial ROS begun to plateau reaching 30% at the maximal SAR values assessed (Figure 3B). By plotting the DHE positive cells against the MSR response for the entire data set (Figure 3D) we observed an extremely strong correlation (R2 = 0.823) between these signals, suggesting that a majority of the ROS production elicited by RF-EMR involved electron leakage from the mitochondrial electron transport chain.Figure 3. RF-EMR induces ROS generation in human spermatozoa, in an SAR-dependent manner unrelated to thermal effects.Percoll-purified spermatozoa (5×106 cells) were suspended in 1 ml BWW in a 35 mm Petri dish and placed within the waveguide while control cells placed outside the waveguide (closed circles). Cells in the waveguide were exposed to 1.8 GHz RF-EMR at SAR levels between 0.4 and 27.5 W/kg (open circles) for 16 h at 21°C. Also, purified sperm cells were subjected to incubation temperatures ranging from 21°C–50°C for 2 h. As the power levels were increased, the cellular generation of ROS increased in a dose-dependent manner. ROS levels were also observed to increase as a result of incubation temperature, but such results were not significant until the temperature exceeded 40°C. A, ROS generation (DHE response) was significantly increased from control levels after exposure to 1.0 W/kg (*p<0.05) and above (***p<0.001). B, RF-EMR induces ROS generation by the sperm mitochondria as monitored by MSR; significant increases were observed at SAR values of 2.8 W/kg (***p<0.001) and above. All results are based on 4 independent samples. C, In order to control for thermal effects, the impact of temperature of cellular ROS generation was monitored; a significant increase in ROS generation was observed as temperatures rose above 40°C (p<0.001). D, Across the entire data set, the total level of ROS generation by human spermatozoa (DHE positive cells) was highly correlated with the level of ROS generation by the mitochondria (MSR positive cells: R2 = 0.823).doi:10.1371/journal.pone.0006446.g003In order to control for bulk thermal effects of RF-EMR exposure, spermatozoa were also incubated at temperatures ranging from 21°C–50°C for 2 h (Figure 3C). This analysis did reveal an effect of heat on free radical generation by human spermatozoa possibly due to the activation of an apoptotic response, however these effects were only significant above 40°C. Thus at the temperature at which these experiments were performed (21°C) the highest observed RF-EMR-induced temperature rise (+0.4°C at 27.5 W/kg), could not of itself account for the increased ROS response observed across the range of SAR settings evaluated in this study.RF-EMR induces oxidative DNA damage (8-OH-dG)In order to determine whether the ROS generation induced on exposure of human spermatozoa to RF-EMR resulted in a state of oxidative stress, we monitored the expression of8-hydroxy-2′-deoxyguanosine (8-OH-dG), a marker for oxidative damage to sperm DNA. As the SAR level was increased, the amount of oxidative DNA damage expressed in the spermatozoa became elevated (Figure 4A). A significant increase in 8-OH-dG expression became apparent at low SAR values (<5.0 W/kg) rising to a maximum of around 20% at the highest levels of exposure (27.5 W/kg). By plotting the 8-OH-dG positive cells against the MSR signal (Figure 4B) it was apparent that a strong positive correlation existed between the two parameters (R2 = 0.727); thehigher the level of mitochondrial ROS generation, the greater the degree of oxidative DNA damage in the spermatozoa.Figure 4. RF-EMR induces oxidative DNA damage in human spermatozoa.Following Percoll fractionation, 5×106 high density, spermatozoa were suspended in 1 ml BWW. The cells were placed in 35 mm Petri dishes and placed inside a waveguide. 5×106 cells in 1 ml BWW were placed outside the waveguide as a control (closed circle). The cells in the waveguide were exposed to 1.8 GHz RF-EMR at SAR levels between 0.4 and 27.5 W/kg (open circles) and all samples were incubated for 16 h at 21°C. Following incubation, Fe2+ and H2O2 was added to cells to act as a positive control, incubated for 1 h, then 100 µl 2 mM DTT/BWW solution was added and incubated for 45 min at 37°C. Cells were fixed and labeled with 100 µl charcoal purified anti-8-OH-dG, FITC tagged antibody at a dilution of 1:50, incubated at 21°C for 1 h, washed and then assessed by flow cytometry. A, As the power levels were increased, the amount of oxidative DNA damage expressed also increased. A significant amount of oxidative DNA damage was observed in cells exposed to 2.8 W/kg (*p<0.05) RF-EMR and above (**p<0.01; ***p<0.001). Results are based on 4 independent samples. B, The levels of 8-OH-dG expression were positively correlated with the levels of ROS generation by the mitochondria (R2 = 0.727).doi:10.1371/journal.pone.0006446.g004RF-EMR induces DNA fragmentation in human spermatozoa To determine whether the oxidative DNA base damage precipitated by RF-EMR-induced ROS generation had any impact on DNA stand breaks in human spermatozoa, the terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay was utilized. As illustrated in Figure 5A, human spermatozoa responded to RF-EMR exposure, with a significant increase in DNA strand breaks atan SAR of 2.8 W/kg (p<0.05) that increased rapidly with rising SAR values and then reached a plateau so that at the highest SAR level assessed (27.5 W/kg), 29% of the cells expressed significant DNA fragmentation. This DNA damage was highly correlated with free radical generation by the sperm mitochondria giving a correlation coefficient of R2 = 0.861 (Figure 5B). Moreover, the level of DNA fragmentation was highly correlated with 8-OH-dG formation (R2 = 0.725; Figure 5C) such that sperm cells exhibiting high levels of oxidative DNA damage, also possessed high levels of DNA fragmentation.Figure 5. RF-EMR induces DNA fragmentation in human spermatozoa.Following Percoll fractionation, 5×106 high density spermatozoa were resuspended in 1 ml BWW, pipetted into 35 mm Petri dishes and placed inside a waveguide. 5×106 cells in 1 ml BWW were placed outside the waveguide as a control (closed circle). The cells in the waveguide were exposed to 1.8 GHz RF-EMR at SAR levels between 0.4 and 27.5 W/kg (open circles) and all samples were incubated for 16 h at 21°C. Following incubation, cells were fixed; DNase-I was used as a positive control. After 1 h incubation at 37°C, 50 µl of label and enzyme master mixes were added to the cells and incubated for 1 h at 37°C. Cells were then washed and assessed by flow cytometry. A, Significant levels of DNA fragmentation was observed in exposed spermatozoa at 2.8 W/kg (*p<0.05) and above (***p<0.001). B, DNA fragmentation was positively correlated with ROS production by the mitochondria as monitored by MSR (R2 = 0.861). C, 8-OH-dG was also positively correlated with DNA fragmentation (R2 = 0.725). Results are based on 4 independent samples.doi:10.1371/journal.pone.0006446.g005DiscussionWhile a high proportion of the male population suffers from infertility associated with defective sperm function [17], the etiology of this condition remains largely unresolved. Notwithstanding the general paucity of information in this area, recent studies have highlighted the interesting finding that male infertility patients are frequently characterized by high levels of DNA damage to their spermatozoa [18]. In light of these data, we have hypothesized that the disruption of sperm fertilizing potential and the concomitant presence of high levels of DNA damage in the sperm nucleus involves a common causative mechanism in the form of oxidative stress [19].Oxidative stress has been known for some time to limit the fertilizing potential of human spermatozoa through the induction of peroxidative damage to the sperm plasma membrane [13], [20]. Oxidative stress is also known to be associated with DNA damage in human spermatozoa [21]. Furthermore, the source of the free radicals responsible for generating such stress appears to be the mitochondria [15]. However, the factors responsible for inducing the mitochondria to leak electrons and propagate the production of ROS have not been elucidated. The research described in this article suggests that one of the key environmental factors involved in the stimulation of sperm mitochondria to produce high levels of ROS, might be excess exposure to RF-EMR from sources such as mobile phones.In a pilot study, human spermatozoa were found to respond to RF-EMR (at 1.8 GHzwith a SAR of 27.5 W/kg) with a range of negative changes including dramatic declines in both sperm vitality and motility. We also observed significant increases in both cytoplasmic ROS levels (DHE) as well as mitochondrial ROS levels (MSR) after RF-EMR exposure. We have previously shown that the chemical induction of mitochondrial ROS production with rotenone can precipitate a state of oxidative stress leading to high levels of lipid peroxidation and a loss of sperm motility [15]. Therefore, these data highlight the particular vulnerability of human spermatozoa to oxidative attack and the potential significance of sperm mitochondria in the generation of free radicals.To assess whether similar effects could be observed at lower power densities, closer to the SAR values associated with mobile phones (0.5–1.5 W/kg) a dose-dependent analysis was conducted. In addition to the conventional assessments of motility and vitality, assays were included to assess the potential for RF-EMR to induce sperm DNA damage and further, whether the DNA damage was oxidative in nature. Confirmation of the detrimental effects of RF-EMR on human sperm was again observed. Over the power density range employed, a significant (P<0.001) dose-dependent response for all sperm parameters was observed, including motility, vitality, ROS generation by the whole cell, ROS generation by the mitochondria, oxidative DNA damage and DNA fragmentation. Furthermore, the profiles of all the observed effects with respect to SAR were intriguingly similar, suggesting a common underlying mechanism.Specifically, all of the responses examined showed an extremely rapid change at low SAR exposures that then reached a plateau at a point where around 30% of the sperm population was affected. This suggests that while we were careful to use only Percoll-purified, high quality spermatozoa in this analysis, there exists within this cell population, a cohort of spermatozoa that are particularly vulnerable to the induction of oxidative stress by RF-EMR. These spermatozoa may have compromised mitochondria, poorly remodeled chromatin or a combination of such factors [15], [22]. Heterogeneity within the sperm population is a feature of the human condition. However, this does not mean that a majority of spermatozoa would not, ultimately, be affected by RF-EMR in vivo; much would depend on the duration of exposure. In vitro, we are limited by the inability of human spermatozoa to survive for more than 24 hours in a simple defined culture medium. In vivo, spermatozoa may take up to a week to move from the seminiferous tubules in the testes to the cauda epididymis and during the whole of this time they would be vulnerable to RF-EMR exposure [23].We recognize that these studies were conducted using spermatozoa suspended in a simple defined culture medium rather than the epididymal plasma in which they would be suspended in vivo. Nevertheless the fact that effects on sperm quality havepreviously been observed in both whole animal radiation experiments [3] and in epidemiological studies of human subjects exposed to various levels of mobile phone radiation [5], [7], [24], emphasizes the biological and clinical relevance of these findings. Moreover, another recent study has found that exposing human spermatozoa to mobile phone radiation for 1 hour leads to significant declines in motility and vitality in concert with an increase in cellular reactive oxygen species generation [25]. The levels of RFEMR exposure were not quantified in this study nor were the sources of ROS identified. Nevertheless, these findings reinforce the general conclusions generated in this paper, particularly with respect to central role played by oxidative stress. The ever-increasing prevalence of mobile communications technology means that humans are now exposed to higher amounts of RF-EMR than ever before. Mobile phones are commonly carried in bags or in pockets in very close proximity to the body. In addition to this, these devices can be stored adjacent to the same part of the body for extended periods of time. In this context, exposure of the male reproductive system to RF-EMR is clearly a significant issue.The particular significance of the present study is that it not only demonstrates a direct effect of RF-EMR on sperm motility, vitality and DNA integrity but also identifies a potential causative mechanism involving electron leakage from the mitochondrial electron transport chain and the induction of oxidative DNA damage. In part, these mechanistic insights have been achieved because the cell type used in these studies, the human spermatozoon, has an extremely simple cellular architecture, lacking significant cytosol and possessing few cellular organelles other than the sperm nucleus, flagellum and mitochondria. One consequence of this structure is that these cells are uniquely vulnerable to oxidative stress. Moreover, such stress is already known to induce the functional and structural lesions observed in this study including both a loss of motility mediated by peroxidative damage to the sperm plasma membrane, as well as the formation of DNA base adducts in the sperm nucleus that ultimately lead to DNA fragmentation [26], [27].Notwithstanding the specialized nature of mammalian spermatozoa, the mechanisms suggested by this study may also apply to RF-EMR-mediated damage in other cell types. The RF-EMR used for communications, including mobile phone networks, is not of high enough power to be classed as ionizing radiation. The latter has sufficient energy to pull away electrons, dramatically altering the properties of affected molecules and typically creating extremely reactive radical species. RF-EMR does not contain sufficient energy for these processes. Nevertheless, this form of radiation may have other effects on larger scale systems such as cells and organelles, which stem from the perturbation of charged molecules and the disruption of electron flow [28], [29]. Mitochondria have one of the largest standing membrane potentials in the body and their energetic functions are entirely dependent on the regulated movement of electrons and protons within the inner mitochondrion membrane.Theoretically, such fluxes might be susceptible to disruptions in local electric fields induced by RF-EMR, offering a potential link between this form of radiation and the non-thermal biological effects observed in this study.This study clearly demonstrates that RF-EMR can damage sperm function via mechanisms that involve the leakage of electrons from the mitochondria and the creation of oxidative stress. These findings have immediate implications for the high rates of male infertility seen in our species, a majority of which is idiopathic. Furthermore, the fact that sperm DNA is damaged by this form of radiation has additional implications for the health and wellbeing of children born to fathers who have experienced high levels of occupational or environmental exposure to RF-EMR around the time of conception. Overall, these finding raise a number of related health policy and patient management issues that deserve our immediate attention. Specifically we recommend that men of reproductive age who engage in high levels of mobile phone use, do not keep their phones in receiving mode below waist level.MethodsEthics StatementThis study was conducted according to the principles expressed in the Declaration of Helsinki. The study was approved by the University of Newcastle (H-712-0799). All patients provided written informed consent for the collection of samples and subsequent analysis.Reagents and SolutionsAll chemicals and reagents used in this research were obtained from Sigma Aldrich (Sigma Chemical Co., St. Louis, MO) unless stated otherwise. All reagents used were of research grade. All fluorescent probes were purchased from Molecular Probes Inc. (Eugene, OR). Biggers, Whitten and Whittingham (BWW) media supplemented with 1 mg/ml polyvinyl alcohol (PVA) was used in all experiments [30]. It was prepared fresh as required and kept at 37°C with an osmolarity in the range of 290–310 mOsm/kg. Human spermatozoaInstitutional and State Government ethical approval was secured for the use of。
锰过氧化物酶的英文Manganese Superoxide Dismutase.Manganese superoxide dismutase (MnSOD), also known as mitochondrial superoxide dismutase, is an antioxidant enzyme that plays a crucial role in protecting cells from oxidative stress. It is a member of the superoxide dismutase (SOD) family of enzymes, which catalyze the conversion of superoxide into hydrogen peroxide and oxygen.Structure and Function.MnSOD is a homotetrameric enzyme, meaning it is composed of four identical subunits. Each subunit contains a manganese ion (Mn2+) bound to a histidine residue, which serves as the catalytic center. The enzyme's active site is located in the protein's interior, where it can access superoxide radicals generated within the mitochondrial matrix.MnSOD catalyzes the dismutation of superoxide into hydrogen peroxide and oxygen according to the following reaction:2 O2+ 2 H+ → H2O2 + O2。
线粒体功能的体外评价方法摘要】线粒体一直被认为是细胞能量生产和代谢工厂,正常的线粒体功能是维持器官的正常功能和细胞稳定的重要因素之一,对于需要高能量代谢的骨骼肌和心肌尤为重要。
线粒体相关疾病的形成及其分子生物学诊断需要临床和实验室的检测。
线粒体疾病的基因双重性,多器官系统特征以及广泛的可识别表型是目前临床诊断所面临的挑战。
为了克服这些临床诊断障碍,实验室对线粒体多方面的评价可以提供相对充足的证据,包括血液学,组织化学分析手段,神经影像分析,刺激实验检测,组织和细胞的酶学分析以及DNA检测(Mancuso M., 2009)。
本文就线粒体的功能性评价方法作以下综述,为临床线粒体相关疾病的诊断提供可行的方法学手段。
【关键词】线粒体呼吸链功能评价【中图分类号】R329 【文献标识码】A 【文章编号】2095-1752(2012)01-0130-021.活性氧族(Reactive Oxygen Species)的产生在细胞内,线粒体是超氧离子(O2-)和其他活性氧族的主要来源,病理情况下,通过异常的氧化反应,线粒体产生约85%的超氧离子(Boveris and Chance, 1973; Dr?ge, 2002). 在线粒体复合体间的电子转运过程中,约2-5%的离子逃逸并释放O2,导致了O2-在复合体I到复合体I I I中的产生,由于线粒体活性增强或呼吸链的抑制作用,氧离子会显著的增加导致了氧化损伤,这可能是脑神经退行性病变发病的机理之一。
活性氧族ROS的生理功能与脑神经元的代谢活性息息相关,过多的ROS会导致线粒体功能异常及神经损伤。
例如,在脑缺血和再灌注过程中,细胞间液中过多的ROS会产生氧化应激,氧化平衡被打破,从而导致细胞性的直接或间接的损伤(L e i e t a l.,1998)。
因此,检测ROS的产生和分布可以从一方面评价线粒体的功能。
目前有很多R O S 的标记物,如广泛应用的二氢氯荧光素dichlorodihydrofluorescein (LeBel et al.,1992),及其各种衍生物如二氢溴乙非啶(Het)(Gallop et al., 1984),和二氢罗丹明dihydrorhodamine(Duganet al., 1995)。
生物技术进展 2023 年 第 13 卷 第 6 期 882 ~ 888Current Biotechnology ISSN 2095‑2341进展评述Reviews线粒体功能障碍在缺氧性肺动脉高压中的作用潘舟 , 胡克 *武汉大学人民医院呼吸与危重症医学科,武汉 430060摘 要:在肺循环中,线粒体除了发挥典型的代谢作用外,还具有调节氧化还原信号、细胞周期、细胞凋亡以及线粒体质量控制等非典型功能。
目前,对线粒体在肺循环中的非典型功能及其响应缺氧的作用机制的研究有限,需要对肺动脉高压的生物学标志物和治疗靶点有更多的了解。
阐述了缺氧诱导的活性氧(reactive oxygen species ,ROS)产生和细胞内钙离子浓度增加,以及缺氧对丙酮酸脱氢酶激酶和丙酮酸激酶M2型表达的影响,总结了线粒体融合和分裂在肺动脉平滑肌细胞增殖、抗凋亡表型等方面的作用,探讨了缺氧对线粒体活性、细胞行为以及线粒体功能障碍对肺动脉高压进展的影响。
深入了解调控线粒体氧化信号、代谢和动态平衡的分子机制对研究和治疗肺动脉高压具有重要意义。
关键词:线粒体功能障碍;肺动脉高压;缺氧;活性氧DOI :10.19586/j.20952341.2023.0072中图分类号:Q591, R363 文献标志码:AThe Role of Mitochondrial Dysfunction in Hypoxic Pulmonary HypertensionPAN Zhou , HU Ke *Department of Respiratory and Critical Care Medicine , Renmin Hospital of Wuhan University , Wuhan 430060, ChinaAbstract :Mitochondria plays a pivotal role in the pulmonary circulation not only in typical metabolic processes but also in regulating oxidative -reduction signaling , cell cycle progression , cell apoptosis , and mitochondrial quality control , among other noncanonical functions. However , current understanding of these noncanonical mitochondrial functions in the pulmonary circulationand their response to hypoxia is limited. Therefore , there is a need for a deeper understanding of the biological markers and therapeutic targets associated with pulmonary arterial hypertension. This review elucidated the effects of hypoxia on mitochondrial activity and cellular behavior , including the induction of reactive oxygen species (ROS ) generation and increased intracellular calcium levels. Furthermore , it highlighted the impact of hypoxia on the expression of pyruvate dehydrogenase kinase and pyruvate kinase M2, as well as the role of mitochondrial fusion and fission in pulmonary artery smooth muscle cell proliferation and resistance to apoptosis. Such investigations contributed to a comprehensive understanding of how hypoxia influences mitochondrial function and how mitochondrial dysfunction affects the progression of PAH. A thorough comprehension of the molecular mechanisms regulating mitochondrial oxidative signaling , metabolism , and dynamic equilibrium holds significant importance in the research and treatment of pulmonary arterial hypertension.Key words :mitochondrial dysfunction ; pulmonary arterial hypertension ; hypoxia ; reactive oxygen species肺动脉高压(pulmonary hypertension ,PH )是一种威胁生命的综合征,肺动脉压力 (pulmonary arterial pressure ,PAP )的升高常导致呼吸困难和运动受限,最终发生右心衰竭。
亚砷酸钠诱导酵母细胞凋亡中SOD1和SOD2基因的作用细胞生物学论文第三篇:亚砷酸钠诱导酵母细胞凋亡中SOD1和SOD2基因的作用摘要:为了探讨SOD1和SOD2基因在亚砷酸钠诱导酵母细胞凋亡中的作用,本实验以酿酒酵母野生株BY4741(WT)及其突变体Δsod1和Δsod2为材料,研究了亚砷酸钠对酵母细胞生长和相对存活率的影响,以及酵母细胞在亚砷酸钠胁迫下活性氧(Reactive oxygen species, ROS)水平、线粒体膜电位和细胞凋亡率的变化。
结果显示,亚砷酸钠可抑制酵母细胞生长,诱导胞内ROS水平和细胞凋亡率升高。
在相同砷处理组中,Δsod1相对存活率、细胞胞内ROS水平和细胞凋亡率显著高于野生株,线粒体膜电位显著低于野生株;Δsod2细胞凋亡率显著高于野生株。
结果表明,亚砷酸钠诱导的酵母细胞凋亡与胞内ROS水平的升高有关,而超氧化物歧化酶基因与亚砷酸钠引起的细胞凋亡密切相关。
关键词:亚砷酸钠; 酵母; 凋亡; 超氧化物歧化酶; 活性氧;Effects of SOD1 and SOD2 Gene Deletions on Arsenic-induced Apoptosis in Yeast CellsWU Lihua YI Huilan CHEN Yanfei QIAO Hongping ZHAO WenjingDepartment of Biology, Taiyuan Normal University School of Life Science, Shanxi UniversityAbstract:To explore the role of SOD1 and SOD2 in sodium arsenite-induced apoptosis in yeast cells, yeast wild-type (BY4741), SOD1 mutant (Δsod1) and SOD2 (Δsod2) mutant strains were used to study the effects of sodium arsenite on the growth and relative survival rate in the yeast cells. In addition, intracellular reactive oxygen species (ROS) level, mitochondrial membrane potential and apoptotic rate of the yeast cells under sodium arsenite-induced stress were determined in this study. The results showed that sodium arsenite induces growth inhibition and cell apoptosis in yeast cells with increased intracellular ROS levels. In the same treatment group, relative growth and clonogenic survival rate were lower, meanwhile, apoptosis rate and intracellular ROS levels were higher significantly in the yeast strain lacking SOD1 (Δsod1) compared to the WT strain, but no significant difference except for apoptosis rate was observed between in Δsod2 and WT strains. These results indicated that sodium arsenite-induced yeast apoptosis was associated with increased intracellular ROS level, and superoxide dismutase is essential in sodium arsenite- induced yeast apoptosis.0 引言凋亡,是一种由基因严格控制的程序性细胞死亡方式,在调控机体生长发育和对外界刺激的反应等生物学过程中起重要作用。
线粒体损伤相关分子模式(Mitochondrial Damage-Associated Molecular Patterns,Mitochondrial DAMPs)是一类细胞内产生的分子,当线粒体受到损伤或功能异常时释放到细胞质内,可能引发免疫系统的免疫应答。
这些分子可以被免疫细胞识别,触发炎症反应,从而参与免疫调节和疾病的发展。
以下是一些与线粒体损伤相关的分子模式:
Mitochondrial DNA(mtDNA):当线粒体受到损伤时,线粒体内的DNA可能被释放到细胞质中,充当一种模式识别受体(PRR),如Toll样受体(TLR)的配体,激活炎症反应。
Cytochrome c:在线粒体损伤时,细胞色素c可能从线粒体中释放到细胞质,与细胞质中的多种蛋白质相互作用,参与细胞凋亡信号通路。
ATP:受损的线粒体释放ATP到细胞质中,ATP作为一种DAMPs,可以激活炎症和免疫反应。
N-formyl peptides:受损的线粒体可能释放N-甲酰化肽段(N-formyl peptides),它们可以作为炎症信号,激活免疫细胞。
ROS(Reactive Oxygen Species):线粒体损伤会导致产生大量的活性氧自由基,ROS的释放可能是一种线粒体与免疫应答之间的连接。
这些线粒体损伤相关分子模式可以激活免疫细胞,引发炎症反应,对于感染、炎症和自身免疫等疾病的发展具有重要作用。
它们的研究有助于深入了解免疫系统与线粒体功能的相互关系,以及在疾病治疗和免疫调控方面的应用。
第 49 卷第 4 期2023年 7 月吉林大学学报(医学版)Journal of Jilin University(Medicine Edition)Vol.49 No.4Jul.2023DOI:10.13481/j.1671‐587X.20230420黄芩素对人舌鳞状细胞癌CAL27细胞增殖的抑制作用及其机制史乃旭1, 郝苗2, 张天夫1, 赵柯林3, 黄子嫣1, 李春艳4, 王晓峰1(1. 吉林大学中日联谊医院口腔科,吉林长春130033;2. 吉林大学中日联谊医院科研中心,吉林长春130033;3. 吉林大学中日联谊医院风湿免疫科,吉林长春130033;4. 新乡医学院第一附属医院口腔颌面外科,河南新乡453100)[摘要]目的目的:探讨黄芩素对人舌鳞状细胞癌(简称舌鳞癌)CAL27细胞增殖的影响,阐明其潜在的作用机制。
方法方法:将对数生长期CAL27细胞分为对照组和不同浓度(12.5、25.0、50.0、100.0和200.0 μmol·L-1)黄芩素组,采用结晶紫染色法观察各组细胞克隆形成情况,CCK-8法检测各组细胞增殖率,2',7'-二氢二氯荧光素二乙酸酯(DCFH-DA)荧光探针检测各组细胞中活性氧(ROS)水平,罗丹明123(Rhodamine123)荧光探针检测各组细胞线粒体膜电位(MMP)水平。
对数生长期CAL27细胞分为对照组和不同浓度(50、100和200 μmol·L-1)黄芩素组,采用流式细胞术检测各组不同细胞周期细胞百分率和细胞凋亡率。
对数生长期CAL27细胞分为对照组、不同浓度(50和100 μmol·L-1)黄芩素组、N-乙酰半胱氨酸(NAC)组、50 μmol·L-1黄芩素+NAC组和100 μmol·L-1黄芩素+NAC组,采用DCFH-DA荧光探针和Rhodamine123荧光探针分别检测黄芩素与NAC联合作用后各组细胞中ROS和MMP水平。
[收稿日期]㊀2020-08-28[修回日期]㊀2020-10-27[基金项目]㊀国家自然科学基金(31670962,81370378);湖南省卫健委临床重大专项(20200011-1003);湖南省大学生创新创业训练计划项目(S201910555137)[作者简介]㊀章舒蕾,硕士研究生,研究方向为动脉粥样硬化病因发病学与防治基础,E-mail 为1029645492@㊂通信作者危当恒,博士,教授,博士研究生导师,研究方向为动脉粥样硬化病因发病学与防治基础,E-mail 为759353094@㊂㊃实验研究㊃[文章编号]㊀1007-3949(2021)29-01-0042-06琥珀酸通过活性氧途径诱导人脐静脉内皮细胞焦亡章舒蕾,梁亚敏,罗涔方,危当恒(南华大学心血管疾病研究所动脉硬化学湖南省重点实验室湖南省动脉硬化性疾病国际科技创新合作基地,湖南省衡阳市421001)[关键词]㊀琥珀酸;㊀人脐静脉内皮细胞;㊀线粒体;㊀活性氧;㊀焦亡;㊀动脉粥样硬化[摘㊀要]㊀目的㊀探讨琥珀酸对人脐静脉内皮细胞(HUVEC )焦亡的影响及其调控机制㊂方法㊀用琥珀酸类似物琥珀酸二乙酯(DS )处理HUVEC 24h ,比色法检测细胞内琥珀酸含量,Western blot 检测细胞焦亡相关蛋白半胱氨酸天冬氨酸特异性蛋白酶1(Caspase-1)㊁白细胞介素1β(IL-1β)㊁IL-18㊁NOD 样受体蛋白3(NLRP3)㊁消皮素D N 端(GSDMD-N )的含量;ATP 测定试剂盒以及活性氧(ROS )荧光探针分别检测琥珀酸对HUVEC 的ATP 以及ROS 生成的影响㊂ROS 清除剂N-乙酰半胱氨酸(NAC )检测ROS 在琥珀酸诱导HUVEC 焦亡中的作用㊂琥珀酸氧化抑制剂丙二酸二甲酯(DMM )检测琥珀酸氧化代谢对ROS 产生的影响㊂结果㊀DS 促HUVEC 内琥珀酸蓄积,上调焦亡相关蛋白Caspase-1㊁IL-1β㊁IL-18㊁GSDMD-N 和NLRP3的表达,抑制ATP 生成并上调ROS 产生㊂NAC 抑制琥珀酸诱导的ROS 生成,并下调上述焦亡相关蛋白的表达㊂DMM 下调琥珀酸诱导的ROS 产生以及HUVEC 的焦亡㊂结论㊀琥珀酸通过氧化代谢上调ROS 生成,进而促进HUVEC 焦亡㊂[中图分类号]㊀R54[文献标识码]㊀ASuccinate induces pyroptosis of human umbilical vein endothelial cells via reactive ox-ygen species pathwayZHANG Shulei,LIANG Yamin,LUO Cenfang,WEI Dangheng(Institute of Cardiovascular Disease &Key Laboratory for Arteriosclerology of Hunan Province &Hunan International Scientif-ic and Technological Cooperation Base of Arteriosclerotic Disease ,Hengyang Medical College ,University of South China ,Hengyang ,Hunan 421001,China )[KEY WORDS ]㊀succinate;㊀human umbilical vein endothelial cell;㊀mitochondria;㊀reactive oxygen species;㊀py-roptosis;㊀atherosclerosis[ABSTRACT ]㊀㊀Aim ㊀To investigate the effect of succinate on pyroptosis of human umbilical vein endothelial cells (HUVEC)and its regulatory mechanism.㊀㊀Methods ㊀HUVECs were treated with succinate analogue diethyl succinate (DS)for 24h,and the content of succinate was detected by colorimetry.㊀Western blot was used to detect the expressions of pyroptosis-related protein cysteinyl aspartate specific proteinase 1(Caspase-1),interleukin-1β(IL-1β),IL-18,NOD-like receptor protein 3(NLRP3),gasdermin D N termine (GSDMD-N).㊀The effects of succinate on ATP and reactive oxygen species (ROS)production of HUVEC were detected by ATP assay kit and ROS fluorescent probe.㊀ROS scavenger N-acetylcysteine (NAC)was used to observe the role of ROS in HUVEC pyroptosis induced by succinate.㊀Dimethyl mal-onate (DMM),a succinate oxidation inhibitor,was used to detect the effect of succinate oxidative metabolism on ROS pro-duction.㊀㊀Results ㊀DS promoted the accumulation of succinate in HUVEC,up-regulated the expressions of pyroptosis-related proteins Caspase-1,IL-1β,IL-18,GSDMD-N and NLRP3,inhibited ATP production and up-regulated ROS pro-duction.㊀NAC inhibited the production of ROS induced by succinate and down-regulated the expressions of above pyropto-sis-related proteins.㊀DMM down-regulated succinate-induced ROS production and HUVEC pyroptosis.㊀㊀Conclusion ㊀Succinate up-regulates ROS production through oxidative metabolism,thus promoting HUVEC pyroptosis.㊀㊀动脉粥样硬化(atherosclerosis,As)为慢性炎症性病理过程,血管内皮细胞炎性活化为As发生发展过程的重要环节[1]㊂近来的研究发现,三羧酸循环中间体及其衍生物通过 非能量代谢途径 调控细胞功能并参与As的发生发展进程[2]㊂琥珀酸为三羧酸循环重要的中间代谢产物,其促进炎症因子的释放以及血管内皮细胞的损伤[3]㊂琥珀酸上调小鼠骨髓来源的树突状细胞白细胞介素1β(interleukin-1β,IL-1β)的表达[4]㊂高糖通过琥珀酸/G蛋白偶联受体91(G-protein coupled receptor 91,GPR91)信号促进血管紧张素Ⅱ释放,损伤人脐静脉内皮细胞(human umbilical vein endothelial cell, HUVEC)[5]㊂Koenis等[6]发现Nur77敲除促As病变,该模型小鼠血清琥珀酸水平明显增加,并且Nur77-/-的巨噬细胞中琥珀酸大量积蓄,但琥珀酸积蓄与血管内皮细胞炎性活化间关系尚不清楚㊂血管内皮细胞焦亡(炎性㊁程序性细胞死亡)发生于As进程,并与As的稳定性密切相关[7]㊂在焦亡过程中,Nod样受体蛋白3(NOD-like receptor pro-tein3,NLRP3)炎性小体被活化,半胱氨酸天冬氨酸特异性蛋白酶1(cysteinyl aspartate specific proteinase1,Caspase-1)前体蛋白被激活,并介导IL-1β和IL-18的加工和成熟㊁活化及裂解消皮素D (gasdermin D,GSDMD)㊂此外,Pro-Caspase-1还能直接裂解GSDMD触发焦亡,参与血管内皮细胞的损伤以及炎性活化[8-9]㊂活性氧(reactive oxygen species,ROS)是细胞焦亡重要的诱导分子,Koenis 等[6]发现琥珀酸大量蓄积的Nur77-/-巨噬细胞伴随有大量的ROS产生㊂因此,本文采用外源性的琥珀酸类似物观察琥珀酸对血管内皮细胞ROS产生以及焦亡的影响,以探讨琥珀酸对血管内皮细胞炎性活化的影响及其机制㊂1㊀材料和方法1.1㊀细胞株与试剂HUVEC购自中国科学院上海生物化学与细胞生物学研究所,琥珀酸二乙酯(diethyl succinate, DS)㊁丙二酸二甲酯(dimethyl malonate,DMM)㊁N-乙酰半胱氨酸(N-acetylcysteine,NAC)购自TCI(上海)化成工业发展有限公司,琥珀酸比色测定试剂盒购自美国Sigma-Aldrich公司,DMEM高糖培养基㊁胎牛血清(fetal bovine serum,FBS)购自美国Gibco公司,ATP测定试剂盒购自中国南京建成生物工程研究所,ROS检测荧光探针二氢乙啶(dihydroethidium,DHE)购自江苏凯基生物技术股份有限公司,BCA蛋白定量试剂盒购自中国上海康为世纪生物科技有限公司,消皮素D N端(GSDMD-N)㊁IL-1β㊁GAPDH㊁Caspase-1㊁NLRP3抗体购自美国Proteintech公司,IL-18抗体购买于美国GeneTex 公司㊂1.2㊀HUVEC培养与处理HUVEC采用含10%FBS的DMEM高糖培养基培养,加入琥珀酸类似物DS(可显著增加胞质和线粒体基质中的琥珀酸),DS的终浓度为10mmol/L;2.5mmol/L NAC预处理3h后加入DS处理24h,观察ROS对DS诱导的血管内皮细胞焦亡的影响; DMM预处理6h后加入DS处理24h,观察琥珀酸氧化代谢途径对血管内皮细胞ROS产生以及焦亡的影响,DMM的浓度10mmol/L㊂1.3㊀BCA蛋白定量法按照说明书处理样品,用酶标仪在562nm波长处测定并记录吸光度,根据标准曲线计算样品中蛋白浓度㊂1.4㊀ATP含量测定将收集好的细胞加入90~100ħ双蒸水,置于热水浴(90~100ħ)中将其匀浆破碎,后将细胞悬液于沸水浴中加热10min,取出细胞悬液用1mL移液枪混匀1min㊂然后按照说明书加试剂,最后混匀,常温静置5min㊂检测波长为636nm,光径为0.5cm,双蒸水调零,测定各管吸光度值,保存并分析结果㊂1.5㊀ROS荧光探针DHE检测从培养箱中取出处理好的细胞,用PBS洗3次㊂加入终浓度为50μmol/L的DHE液,在37ħ水浴箱避光条件下孵育45min,PBS清洗细胞3次,每次6min㊂荧光显微镜拍照,保存并分析结果㊂1.6㊀Western blot检测蛋白的表达收集细胞,使用预冷的PBS洗3次,加入裂解液后4ħ静置30min,12000r/min离心10min,取上清后采用BCA法进行蛋白定量㊂目的蛋白经SDS-PAGE凝胶电泳分离并转移至PVDF膜上,5%脱脂牛奶室温封闭2h,加入单克隆抗体GSDMD (1ʒ1000)㊁IL-1β(1ʒ1000)㊁IL-18(1ʒ1000)㊁Caspase-1(1ʒ1000)㊁NLRP3(1ʒ1000)㊁GAPDH (1ʒ2000)4ħ孵育过夜,TBST洗3次,每次10 min,相应二抗室温孵育2h,ECL发光试剂显色,拍照并保存结果㊂1.7㊀琥珀酸含量测定收集细胞,在冰中快速匀浆,加入100μL低温琥珀酸测定缓冲液,以10000r /min 离心10min ,收集上清液㊂采用琥珀酸比色测定试剂盒,加入各反应物,在37ħ下避光孵育30min ,450nm 波长处测定吸光度㊂1.8㊀统计学方法所有实验数据均用x ʃs 表示,运用Image ProPlus ㊁GraphPad Prism 5统计软件进行数据分析,组间比较采用方差分析及t 检验,P <0.05表示差异具有统计学意义㊂2㊀结㊀果2.1㊀琥珀酸促HUVEC 焦亡㊀㊀为了探讨琥珀酸对血管内皮细胞焦亡的影响,首先观察了琥珀酸的类似物DS 处理24h 后血管内皮细胞内琥珀酸含量,结果显示DS 明显增加血管内皮细胞内琥珀酸含量(图1A)㊂然后Western blot 检测了DS 对血管内皮细胞焦亡相关蛋白NLRP3㊁GSDMD-N㊁Caspase-1㊁IL-18以及IL-1β蛋白表达的影响,结果表明DS 上调NLRP3㊁GSDMD-N㊁Caspase-1㊁IL-18以及IL-1β的表达(图1B)㊂这些结果表明细胞内琥珀酸蓄积促焦亡㊂2.2㊀琥珀酸抑制HUVEC 的ATP 生成线粒体是细胞的能量工厂,ATP 是维持机体正常生理活动的重要物质㊂ATP 含量检测的结果表明DS 抑制ATP 生成(图2)㊂图1.琥珀酸促HUVEC 焦亡(n =3)A 为DS 促HUVEC 内琥珀酸蓄积;B 为DS 处理HUVEC 24h,Western blot 检测焦亡相关蛋白NLRP3㊁Caspase-1㊁GSDMD-N㊁IL-18以及IL-1β的表达㊂a 为P <0.05,b 为P <0.01,与Control 组比较㊂Figure 1.HUVEC pyroptosis promoted by succinate (n =3)图2.琥珀酸对HUVEC 线粒体ATP 生成的影响(n =3)a 为P <0.01,与Control 组比较㊂Figure 2.Effect of succinate on mitochondrial ATPproduction in HUVEC (n =3)2.3㊀琥珀酸增加HUVEC 的ROS 水平随后,我们采用荧光探针检测琥珀酸对ROS 的影响,结果表明DS 显著增加HUVEC 的ROS 水平(图3)㊂图3.琥珀酸增加HUVEC 的ROS 水平Figure 3.Succinate promoted the generation ofROS in HUVEC2.4㊀ROS 清除剂NAC 抑制琥珀酸诱导的HUVEC 焦亡为了探讨ROS 在琥珀酸诱导血管内皮细胞焦亡中的作用,我们采用ROS 清除剂NAC(2.5mmol /L)预处理血管内皮细胞3h,再DS 孵育HUVEC,结果表明NAC 预处理抑制琥珀酸诱导的ROS 积聚(图4A),并且抑制琥珀酸诱导的焦亡相关蛋白表达(图4B)㊂图4.NAC 抑制琥珀酸诱导的HUVEC 焦亡(n =3)A 为NAC 预处理3h 减少琥珀酸诱导的线粒体ROS 含量;B 为NAC 减少焦亡相关蛋白NLRP3㊁GSDMD-N㊁Caspase-1㊁IL-18和IL-1β的含量㊂a 为P <0.05,与Control 组比较;b 为P <0.05,与DS 组比较㊂Figure 4.NAC inhibited HUVEC pyroptosis induced by succinate (n =3)2.5㊀琥珀酸氧化抑制剂DMM 抑制琥珀酸诱导的HUVEC 焦亡为了进一步探讨琥珀酸促ROS 生成的机制,我们采用琥珀酸氧化抑制剂DMM(10mmol /L)抑制琥珀酸的氧化㊂结果表明DMM 减少血管内皮细胞中ROS 的积聚(图5A),焦亡相关蛋白NLRP3㊁GS-DMD-N㊁Caspase-1㊁IL-18以及IL-1β表达水平降低(图5B),这些结果表明琥珀酸氧化促进ROS 的生成进而促HUVEC 焦亡㊂3㊀讨㊀论琥珀酸是三羧酸循环中间产物,由琥珀酰辅酶A 合成酶催化生成㊂研究发现,琥珀酸通过非能量代谢底物途径参与多种生理和病理过程㊂琥珀酸上调心肌细胞肥大标志物心房利钠多肽和p-Akt /t-Akt 的水平,参与右心室肥厚的形成[10];肿瘤组织琥珀酸/GPR91信号靶向PI3K-HIF-1α轴介导肿瘤相关巨噬细胞极化和肿瘤转移[11]㊂琥珀酸通过HIF-1α/VEGF 轴诱导类风湿关节炎滑膜血管生成[12]㊂我们的研究发现琥珀酸上调血管内皮细胞焦亡标记物的表达,提示细胞内的琥珀酸蓄积促进焦亡㊂焦亡是一种新发现的促炎性㊁程序性细胞死亡方式,在经典的Caspase-1依赖性焦亡通路中,活化的NLRP3促前体Caspase-1成熟,进而剪切IL-18㊁IL-1β和GSDMD,介导细胞焦亡㊂Wu 等[13]发现尼古丁引起血管内皮细胞损伤并诱发焦亡;阿托伐他汀通过lncRNA NEXN-AS1/NEXN 通路抑制血管内皮细胞焦亡,以非降脂途径保护血管内皮细胞功能[14]㊂我们的结果表明,琥珀酸促血管内皮细胞焦亡,提示琥珀酸可能通过焦亡途径引起血管内皮细胞损伤以及炎症活化㊂ROS 是细胞焦亡重要的激活分子,介导氧化低图5.DMM抑制琥珀酸诱导的HUVEC焦亡(n=3)A为DMM抑制琥珀酸诱导的ROS产生;B为DMM减少琥珀酸诱导的焦亡相关蛋白NLRP3㊁GSDMD-N㊁Caspase-1㊁IL-18和IL-1β的含量㊂a为P<0.05,与Control组比较;b为P<0.05,与DS组比较㊂Figure5.DMM inhibited HUVEC pyroptosis induced by succinate(n=3)密度脂蛋白处理的HUVEC焦亡[15];肠道菌群代谢产物氧化三甲胺通过激活ROS-TXNIP-NLRP3炎症小体轴诱导炎症和内皮功能损伤[16]㊂我们的结果表明琥珀酸类似物DS增加了血管内皮细胞ROS含量,ROS清除剂NAC可降低细胞内ROS含量并下调琥珀酸诱导的血管内皮细胞焦亡相关蛋白NLRP3㊁GSDMD-N㊁Caspase-1㊁IL-1β和IL-18的表达,表明琥珀酸通过ROS途径促血管内皮细胞焦亡㊂细胞内ROS来源于呼吸链以及底物的氧化代谢[17]㊂Li等[18]发现DMM抑制腹膜炎小鼠肿瘤坏死因子的分泌和ROS的产生㊂在本研究中我们采用琥珀酸脱氢酶抑制剂DMM观察琥珀酸氧化代谢对ROS生成的影响,结果发现DMM明显减少ROS 的产生及焦亡相关蛋白NLRP3㊁GSDMD-N㊁Caspase-1㊁IL-1β和IL-18的表达,表明细胞内琥珀酸通过氧化途径增加ROS的生成和蓄积并促焦亡发生㊂多位学者研究发现琥珀酸脱氢酶抑制剂DMM通过抑制琥珀酸的氧化代谢改善缺血后再灌注时心㊁脑㊁肾组织损伤[19-21];Mills等[22]发现DMM抑制脂多糖诱导的IL-1β的产生,抑制炎症反应,提示通过抑制琥珀酸的氧化代谢可以抑制血管内皮细胞焦亡并保护血管内皮细胞功能㊂综上所述,琥珀酸通过氧化代谢途径增加ROS 的生成,促血管内皮细胞焦亡,但琥珀酸在As发生㊁发展中的作用有待于进一步的探讨和验证㊂[参考文献][1]李苗,王丽丽,常冰梅.血管内皮细胞功能损伤机制的研究进展[J].中国动脉硬化杂志,2019,27(8): 730-736.[2]Martinez-Reyes I,Chandel NS.Mitochondrial TCA cycle metabolites control physiology and disease[J].Nat Com-mun,2020,11(1):102.[3]Mills E,O N eill LA.Succinate:a metabolic signal in in-flammation[J].Trends Cell Biol,2014,24(5):313-320.[4]Tannahill GM,Curtis AM,Adamik J,et al.Succinate is an inflammatory signal that induces IL-1beta through HIF-1alpha[J].Nature,2013,496(7444):238-242.[5]Peti-Peterdi J.High glucose and renin release:the role of succinate and GPR91[J].Kidney Int,2010,78(12): 1214-1217.[6]Koenis DS,Medzikovic L,van Loenen PB,et al.Nuclear receptor Nur77limits the macrophage inflammatory response through transcriptional reprogramming of mitochondrial me-tabolism[J].Cell Rep,2018,24(8):2127-2140. [7]Xu YJ,Zheng L,Hu YW,et al.Pyroptosis and its rela-tionship to atherosclerosis[J].Clin Chim Acta,2018, 476:28-37.[8]He Y,Hara H,Nunez G.Mechanism and regulation of NLRP3inflammasome activation[J].Trends Biochem Sci, 2016,41(12):1012-1021.[9]Xi H,Zhang Y,Xu Y,et al.Caspase-1inflammasome acti-vation mediates homocysteine-induced pyrop-apoptosis in en-dothelial cells[J].Circ Res,2016,118(10):1525-1539.[10]Yang L,Yu D,Mo R,et al.The succinate receptor GPR91is involved in pressure overload-induced ventricular hypertro-phy[J].PLoS One,2016,11(1):e0147597. [11]Wu JY,Huang TW,Hsieh YT,et al.Cancer-derivedsuccinate promotes macrophage polarization and cancer metastasis via succinate receptor[J].Mol Cell,2020,77(2):213-227.[12]Li Y,Liu Y,Wang C,et al.Succinate induces synovialangiogenesis in rheumatoid arthritis through metabolic re-modeling and HIF-1alpha/VEGF axis[J].Free Radic Biol Med,2018,126:1-14.[13]Wu X,Zhang H,Qi W,et al.Nicotine promotes athero-sclerosis via ROS-NLRP3-mediated endothelial cell pyrop-tosis[J].Cell Death Dis,2018,9(2):171. [14]Wu LM,Wu SG,Chen F,et al.Atorvastatin inhibits py-roptosis through the lncRNA NEXN-AS1/NEXN pathway in human vascular endothelial cells[J].Atherosclerosis,2020,293:26-34.[15]Zeng Z,Chen J,Wu P,et al.Ox-LDL induces vascularendothelial cell pyroptosis through miR-125a-5p/TET2pathway[J].J Cell Physiol,2019,234(5):7475-7491.[16]Wu P,Chen J,Chen J,et al.Trimethylamine N-oxidepromotes ApoE-/-mice atherosclerosis by inducing vascular endothelial cell pyroptosis via the SDHB/ROS pathway [J].J Cell Physiol,2020,235(10):6582-6591. [17]Jardim-Messeder D,Caverzan A,Rauber R,et al.Succi-nate dehydrogenase(mitochondrial complex II)is a source of reactive oxygen species in plants and regulates development and stress responses[J].New Phytol,2015, 208(3):776-789.[18]Li Y,Jia A,Wang Y,et al.Immune effects of glycolysisor oxidative phosphorylation metabolic pathway in protecting against bacterial infection[J].J Cell Physiol,2019,234(11):20298-20309.[19]Beach TE,Prag HA,Pala L,et al.Targeting succinatedehydrogenase with malonate ester prodrugs decreases renal ischemia reperfusion injury[J].Redox Biol,2020, 36:101640.[20]Kula-Alwar D,Prag HA,Krieg T.Targeting succinate me-tabolism in ischemia/reperfusion injury[J].Circulation, 2019,140(24):1968-1970.[21]Zhang J,Wang YT,Miller JH,et al.Accumulation ofsuccinate in cardiac ischemia primarily occurs via canonical Krebs cycle activity[J].Cell Rep,2018,23(9): 2617-2628.[22]Mills EL,Kelly B,Logan A,et al.Succinate dehydro-genase supports metabolic repurposing of mitochondria to drive inflammatory macrophages[J].Cell,2016,167(2):457-470.(此文编辑㊀曾学清)。
生态毒理学报Asian Journal of Ecotoxicology第18卷第4期2023年8月V ol.18,No.4Aug.2023㊀㊀基金项目:湖南省重点研发计划项目(2023NK2029);湖南省本科生/研究生科研创新项目(S202110537040,CX20220694);国家自然科学基金青年项目(21906050)㊀㊀第一作者:汤蕾(2000 ),女,硕士研究生,研究方向为环境毒理学,E -mail:********************㊀㊀*通信作者(Corresponding author ),E -mail:***********************.cn ㊀㊀#共同通信作者(Co -corresponding author ),E -mail:***********************DOI:10.7524/AJE.1673-5897.20220805002汤蕾,李鑫,李昕畅,等.纳米氧化亚铜和氧化铜的肺细胞毒性差异及影响因素[J].生态毒理学报,2023,18(4):478-485Tang L,Li X,Li X C,et al.Different pulmonary cytotoxicity of nano -cuprous oxide and nano -copper oxide and influencing factors [J].Asian Journal of Ecotoxicology,2023,18(4):478-485(in Chinese)纳米氧化亚铜和氧化铜的肺细胞毒性差异及影响因素汤蕾,李鑫,李昕畅,罗琳#,曹林英*湖南农业大学资源环境学院,长沙410128收稿日期:2022-08-05㊀㊀录用日期:2022-11-07摘要:纳米农药的广泛应用将不可避免导致其重要成分(比如纳米氧化铜和纳米氧化亚铜)的环境残留㊂人体可通过灰尘吸入㊁饮水及农产品摄入等方式暴露于纳米氧化铜和纳米氧化亚铜㊂因此,阐明它们的健康危害具有重要意义㊂本研究我们首次对比研究了纳米氧化铜和纳米氧化亚铜的A549肺细胞毒性差异,并从氧化应激和线粒体损伤角度探讨了细胞毒性作用机制㊂实验结果表明纳米氧化铜和纳米氧化亚铜均具有显著的A549细胞毒性,观察到的最低效应浓度分别为20mg ㊃L -1和5mg ㊃L -1㊂纳米氧化亚铜表现出更强的细胞毒性作用,100mg ㊃L -1时细胞活性抑制率达到95%㊂活性氧实验结果表明纳米氧化铜和纳米氧化亚铜均可诱导活性氧生成,呈现时间与浓度依赖关系㊂纳米氧化亚铜的活性氧诱导作用明显强于纳米氧化铜,24h 最大诱导率分别为对照组的3.5倍和1.5倍㊂线粒体膜电位实验结果表明纳米氧化亚铜比纳米氧化铜具有更强的线粒体去极化作用,并呈剂量(1~100mg ㊃L -1)依赖关系,最大线粒体膜电位下降率分别为60%和20%㊂活性氧和线粒体膜电位的结果与细胞毒性的结果基本一致,提示了氧化应激和线粒体损伤可能是导致纳米氧化铜和纳米氧化亚铜细胞毒性的关键分子机制㊂本研究揭示了纳米氧化铜和纳米氧化亚铜的肺细胞毒性差异及潜在分子机制,可为纳米农药的健康风险评估及合理施用提供重要理论依据㊂关键词:纳米氧化铜;纳米氧化亚铜;肺细胞;细胞毒性;氧化应激;线粒体损伤文章编号:1673-5897(2023)4-478-08㊀㊀中图分类号:X171.5㊀㊀文献标识码:ADifferent Pulmonary Cytotoxicity of Nano-cuprous Oxide and Nano-copper Oxide and Influencing FactorsTang Lei,Li Xin,Li Xinchang,Luo Lin #,Cao Linying *College of Resources and Environment,Hunan Agricultural University,Changsha 410128,ChinaReceived 5August 2022㊀㊀accepted 7November 2022Abstract :The wide application of nano -pesticides leads to residues of its main components (such as nano -copper oxide and nano -cuprous oxide)in the environment inevitably.The human body can expose to nano -copper oxide and nano -cuprous oxide through inhaling dust,drinking water and ingesting agricultural products.Therefore,it is of great significance to elucidate their health hazards.In the present study,we firstly investigated the cytotoxicity vari -ation of nano -copper oxide and nano -cuprous oxide on A549cells as well as explored the cytotoxic mechanisms第4期汤蕾等:纳米氧化亚铜和氧化铜的肺细胞毒性差异及影响因素479㊀from the perspectives of oxidative stress and mitochondrial damage.Results showed that both nano-copper oxide and nano-cuprous oxide had significant cytotoxic effects on A549cells,with the lowest observed effective concen-trations of20mg㊃L-1and5mg㊃L-1,respectively.Nano-cuprous oxide exhibited stronger cytotoxic effects than nano-copper oxide with inhibition rate of cell viability reaching95%at100mg㊃L-1.The experimental results of reactive oxygen species showed that both nano-copper oxide and nano-cuprous oxide could induce the generation of reactive oxygen species in a time-dependent and concentration-dependent manner.The induction effect of nano-cuprous oxide on reactive oxygen species was significantly stronger than that of nano-copper oxide,with the maxi-mum induction rate of3.5-fold and1.5-fold compared to the ctrl group at24h.The experimental results of mito-chondrial membrane potential showed that nano-cuprous oxide had stronger activity on mitochondrial depolariza-tion than nano-copper oxide in a dose-dependent manner(1~100mg㊃L-1),with the maximum reduction rates of mitochondrial membrane potential reaching60%and20%,respectively.The results of reactive oxygen species and mitochondrial membrane potential were consistent with the results of cytotoxicity,indicating that oxidative stress and mitochondrial damage might be the key molecular mechanisms of cytotoxicity of nano-copper oxide and nano-cuprous oxide.This study revealed the different pulmonary cytotoxicity of nano-copper oxide and nano-cuprous oxide as well as the potential molecular mechanisms,which can provide important theoretical basis for the health risk assessment and rational application of nano-pesticides.Keywords:nano-copper oxide;nano-cuprous oxide;lung cell;cytotoxicity;oxidative stress;mitochondria damage㊀㊀目前,我国的农药利用率低㊁用量大,造成了农药残留量大及环境污染等问题㊂纳米技术在农业的可持续发展中显示出巨大的应用潜力,其中纳米农药可增加农药活性成分的稳定性,延长有效持续时间,有效改善农药利用率,从而减少农药的环境负荷,其在农业生产中的使用已备受关注㊂但是随着纳米农药的广泛使用,将不可避免地导致了它们在环境中的残留,甚至进入人体产生危害,因此,阐明它们的生态风险及环境健康危害具有重要意义[1-2]㊂纳米农药主要分为两大类:纳米材料作为载体包裹有机小分子农药活性成分;无机纳米农药㊂纳米氧化铜(CuO NPs)和纳米氧化亚铜(Cu2O NPs)是无机纳米农药的2种典型成分[1,3]㊂由于CuO NPs和Cu2O NPs具有很好的抗菌活性,对多种菌类有效,被列为推荐药剂在农业领域得到广泛应用[4-6],并且一些研究表明,Cu2O NPs的抗菌活性较CuO NPs更强[7-8]㊂纳米颗粒(nano-particles,NPs)进入人体最常见的途径是通过呼吸系统吸入,深入肺部,引起肺部氧化应激和炎症反应[9-10]㊂同时,NPs也可以通过肺泡或其他方式(如饮水㊁皮肤接触等)进入血液并影响其他器官[11]㊂与其他NPs类似,除了通过呼吸㊁饮水及皮肤暴露等方式以外,由于CuO NPs和Cu2O NPs在农业系统的广泛使用,通过农产品的摄入也是它们进入人体的重要途径㊂NPs对生物体的毒性机制主要是诱导氧化应激㊁炎症反应及DNA损伤等[9,12-13]㊂目前已有较多研究显示了CuO NPs 具有较强的细胞毒性作用㊂例如孙婷婷和蒋澄宇[14]通过比较CuO㊁Fe2O3㊁TiO2㊁SiO2等金属NPs对小鼠的肺部毒性作用,发现CuO NPs具有较强的毒性,可导致小鼠急性肺损伤,而其他几种金属NPs的作用较小;Fahmy和Cormier[15]对SiO2NPs㊁Fe2O3 NPs和CuO NPs进行比较研究,发现CuO NPs导致气管上皮Hep-2细胞活力出现显著的剂量依赖性下降,而其他NPs基本无细胞毒性作用,并且CuO NPs的细胞毒性作用与氧化损伤直接相关;Fu[16]的研究证明CuO NPs可以抑制HepG2细胞增殖,对细胞产生氧化损伤作用,其机制与ROS诱导线粒体介导的细胞凋亡途径有关㊂此外,也有研究表明CuO NPs对植物[17-18]㊁水生动物[19]及微生物[20-21]的毒性作用,并且与氧化应激有关㊂目前已有较多研究表明了CuO NPs的毒性作用,但对Cu2O NPs的研究非常缺乏㊂虽然有研究表明Cu2O NPs具有更强的抗菌活性,但其是否具有更强的非靶标毒性作用进而产生更严重的健康危害,需要进一步的毒理学评估㊂因此,本研究以A549人源肺细胞作为模型,对比研究了CuO NPs和Cu2O NPs的细胞毒性差异,并探讨了影响其毒性差异的潜在因素,有望为其环境健康风险评估及安全使用提供理论参考㊂480㊀生态毒理学报第18卷1㊀材料与方法(Materials and methods)1.1㊀实验材料与试剂人肺腺癌细胞A549㊁细胞培养基DMEM㊁PBS缓冲溶液购于武汉普诺赛生命科技有限公司;活性氧检测探针(DCFH-DA)㊁线粒体膜电位检测探针(Mito-Tracker Red CMXRos)购自碧云天生物技术有限公司;二甲基亚砜(DMSO,纯度为99%)和噻唑蓝(MTT,纯度>99%)购自上海麦克林生化科技有限公司;纳米氧化铜(CuO NPs,纯度>99.9%)和纳米氧化亚铜(Cu2O NPs,纯度>99.9%)购自北京中科科优科技有限公司,CuO NPs和Cu2O NPs暴露前采用细胞培养基配制,现配现用㊂1.2㊀仪器设备BB150-2TCS-L CO2培养箱(赛默飞世尔科技有限公司,美国);TECAN Spark20M酶标仪(帝肯集团有限公司,瑞士);FA004分析天平(上海舜宇恒平仪器有限公司,中国);ZEISS Sigma300型扫射电子显微镜(scanning electron microscope,SEM;蔡司,德国);FEI Tecnai F20透射电子显微镜(transmission electron microscope,TEM;FEI,美国);Malvern Zeta-sizer Nano ZS90型动态光散射仪(dynamic light scattering,DLS;马尔文,英国);AA-6880原子吸收仪(岛津,日本)㊂1.3㊀材料的表征将CuO NPs和Cu2O NPs分散在去离子水中,超声后得到悬浮液㊂采用德国ZEISS Sigma300 SEM和美国FEI Tecnai F20TEM拍摄NPs形貌㊂用纳米粒度Zeta电位仪测定Zeta电位,同时以动态光散射法测定水合粒径㊂1.4㊀细胞毒性检测A549细胞采用加入了100μg㊃mL-1链霉素㊁100U㊃mL-1青霉素和10%(VʒV)胎牛血清的完全培养基(普诺塞,武汉)在恒温恒湿培养箱(37ħ和5%(VʒV)CO2)中进行培养㊂在96孔板中每孔接种A549细胞(5ˑ104个),贴壁培养24h㊂暴露不同浓度(0.1~100mg㊃L-1)的CuO NPs和Cu2O NPs,继续培养24h㊂吸出染毒液,用PBS对细胞进行清洗2次㊂加入MTT溶液孵育3h,吸去探针,加入100μL DMSO,以溶解MTT的深紫色产物(甲瓒)㊂轻轻摇晃,用酶标仪,以690nm为参考波长,在490 nm的吸收度测定细胞活性㊂1.5㊀活性氧检测将A549细胞悬浮液(5ˑ104个,100μL)接种在96孔板中孵育24h㊂吸出培养基,加入10μmol∙L-1的DCFH-DA探针(100μL)孵化20min后吸出,用100μL PBS轻轻洗涤2次㊂将细胞暴露于不同浓度(1~100mg㊃L-1)的CuO NPs和Cu2O NPs㊂采用酶标仪检测荧光强度(激发波长485nm,发射波长535nm)㊂1.6㊀线粒体膜电位检测将A549细胞(5ˑ104个,100μL)接种在96孔板中孵育24h㊂吸出培养基,将细胞分别暴露于不同浓度(1~100mg㊃L-1)的CuO NPs和Cu2O NPs12 h㊂然后将含污染物的溶液吸出,分别加入100nmol ㊃L-1的线粒体膜电位探针,孵化20min,再将探针去除后,用PBS将细胞洗3次,最后加入100μL的PBS溶液用酶标仪检测荧光强度,设置激发波长为565nm,发射波长610nm㊂1.7㊀培养基中铜离子浓度检测将配好的20㊁40㊁50㊁100mg㊃L-1的CuO NPs和Cu2O NPs悬浊液加入到12孔板中,与细胞在相同条件下处理24h后,离心30min(12000r㊃min-1),取上清液㊂用火焰原子吸收光谱仪检测上清液中铜离子含量㊂1.8㊀统计学方法采用GraphPad Prism8和Excel进行实验数据处理和分析㊂所有的实验都进行3次重复,每个实验组都设置至少3个平行,用平均值ʃ标准差来表示试验的结果㊂*P<0.05表示处理组与对照组具有显著性差异㊂2㊀结果(Results)2.1㊀NPs的表征SEM和TEM图像显示CuO NPs和Cu2O NPs 呈不规则形状的颗粒(图1(a)~(d)),CuO NPs呈棒状,而Cu2O NPs呈球状㊂Zeta电位测定结果表明,CuO NPs的Zeta电位为(19.97ʃ5.98)mV,Cu2O NPs 的Zeta电位为(25.5ʃ6.55)mV㊂DLS结果显示CuONPs的水合粒径为403nm,Cu2O NPs的水合粒径为432nm(图1(e)和(f)),在水溶液中分散比较均匀,有部分团聚㊂2.2㊀CuO NPs和Cu2O NPs对A549细胞活性的影响如图2所示,无酚红DMEM配制的CuO NPs和Cu2O NPs呈均匀分散溶液,其中CuO NPs呈灰黑色,而Cu2O NPs呈浅黄色㊂如图3(a)所示,CuONPs和Cu2O NPs均对A549细胞产生了明显的毒第4期汤蕾等:纳米氧化亚铜和氧化铜的肺细胞毒性差异及影响因素481㊀图1㊀纳米氧化铜(CuO NPs )和纳米氧化亚铜(Cu 2O NPs )的形态及粒径表征注:(a)CuO NPs 的扫描电镜图;(b)Cu 2O NPs 的扫描电镜图;(c)CuO NPs 的透射电镜图;(d)Cu 2O NPs 的透射电镜图;(e)动态光散射测定CuO NPs 的水合半径;(f)动态光散射测定Cu 2O NPs 的水合半径㊂Fig.1㊀Morphology and particle size characterization of nano -copper oxide (CuO NPs)and nano -cuprous oxide (Cu 2O NPs)Note:(a)Scanning electron microscopy photograph of CuO NPs;(b)Scanning electron microscopy photograph of Cu 2O NPs;(c)Transmiss i on electron microscopy photograph of CuO NPs;(d)Transmission electron microscopy photograph of Cu 2O NPs;(e)Hydrated radius of CuO NPs determined by dynamic light scattering;(f)Hydrated radius of Cu 2O NPs determined by dynamic lightscattering.图2㊀纳米氧化铜(CuO NPs )和纳米氧化亚铜(Cu 2O NPs )的溶液Fig.2㊀Solution of nano -copper oxide (CuO NPs)and nano -cuprous oxide (Cu 2O NPs)482㊀生态毒理学报第18卷图3㊀纳米颗粒(NPs)(a)及铜离子(Cu2+)(b)对A549细胞的活性影响注:Ctrl组表示等量稀释纳米材料的培养基处理,*表示与对照组相比差异显著(P<0.05)㊂Fig.3㊀Effects of nanoparticles(NPs)(a)and copper ion(Cu2+)(b)on A549cell viability Note:Ctrl group represents treatment with equal volume medium used to dilute nanomaterials;*represents significant difference compared with the control(P<0.05).性,并且呈剂量依赖效应,观察到的最低效应浓度分别为20mg㊃L-1和5mg㊃L-1㊂并且不同浓度的细胞毒性对比也表明Cu2O NPs的A549细胞毒性效应明显强于CuO NPs㊂同时,考察了铜离子的细胞毒性以便作为对比㊂如图3(b)所示,A549细胞暴露在低浓度Cu2+溶液中时,活细胞数量与对照组相比有所增加,说明低浓度的Cu2+可以促进细胞增殖,这可能与Cu是人体生长的必需元素有关㊂当Cu2+浓度达到50mg㊃L-1时表现出细胞毒性作用,但是与CuO NPs和Cu2O NPs相比,Cu2+细胞毒性相对较小㊂2.3㊀CuO NPs和Cu2O NPs对A549细胞胞内活性氧生成的影响㊀㊀前面我们发现了CuO NPs和Cu2O NPs的肺细胞毒性差异,为了进一步探讨细胞毒性差异的分子机理,对两者诱导活性氧生成进行了测定㊂NPs在细胞中与细胞器的相互作用促进活性氧的产生是其产生细胞毒性作用的典型分子机制之一㊂由图4可知,CuO NPs和Cu2O NPs暴露A549肺细胞3㊁6㊁12及24h均可导致ROS过量生成,随着暴露时间增加明显出现了ROS的积累,并呈现剂量依赖关系㊂此外,Cu2O NPs诱导ROS生成效应明显强于CuO NPs㊂这一现象与它们的细胞毒性是一致的㊂因此,我们推断CuO NPs和Cu2O NPs的细胞毒性差异与两者诱导氧化应激效应有关㊂2.4㊀CuO NPs和Cu2O NPs对线粒体膜电位的影响NPs可以侵入细胞并粘附在线粒体膜上,从而破坏线粒体膜并导致其去极化㊂如图5所示,不同浓度CuO NPs㊁Cu2O NPs暴露A549细胞12h后,均可在ȡ40mg㊃L-1浓度下导致线粒体膜电位降低,导致线粒体去极化作用㊂同时,发现Cu2O NPs对肺细胞A549线粒体去极化作用明显大于CuO NPs,这与两者细胞毒性实验结果吻合㊂因此,CuONPs㊁Cu2O NPs的不同细胞毒性可能与两者对线粒体损伤程度不同也有一定关系㊂2.5㊀培养基中CuO NPs和Cu2O NPs释放的铜离子浓度㊀㊀金属离子的溶出是金属NPs产生细胞毒性的重要来源㊂CuO NPs和Cu2O NPs产生的细胞毒性差异也可能与它们溶出的铜离子浓度不同有关㊂因此,进一步对培养基中溶出的铜离子浓度进行了考察㊂但是由图6可知,CuO NPs和Cu2O NPs在培养基中释放的铜离子浓度没有明显差别,所释放的最大铜离子浓度均<16mg㊃L-1,说明两者的毒性差异可能并不是由于其在培养基中所释放的铜离子总量不同所导致㊂3㊀讨论(Discussion)随着农业纳米技术研发支出的迅速增加,中国有可能成为全球最大的纳米农药生产国和消费国㊂因此,纳米农药的调控和科学评价迫在眉睫㊂纳米材料通过呼吸进入肺部是其产生毒害作用的主要途径之一㊂本研究选用人源肺细胞A549作为细胞模型,以CuO NPs和Cu2O NPs为研究对象,首次对比第4期汤蕾等:纳米氧化亚铜和氧化铜的肺细胞毒性差异及影响因素483㊀图4㊀纳米氧化铜(CuO NPs )和纳米氧化亚铜(Cu 2O NPs )对A549细胞活性氧(ROS )生成的影响(不同暴露时间)注:(a)3h ;(b)6h ;(c)12h ;(d)24h ;Ctrl 组表示等量稀释纳米材料的培养基处理㊂Fig.4㊀Effects of nano -copper oxide (CuO NPs)and nano -cuprous oxide (Cu 2O NPs)on reactive oxygen species (ROS)generation in A549cells (different exposure time)Note:(a)3h;(b)6h;(c)12h;(d)24h;Ctrl group represents treatment with equal volume medium used to dilutenanomaterials.图5㊀纳米氧化铜(CuO NPs )和纳米氧化亚铜(Cu 2O NPs )对A549细胞线粒体膜电位(MMP )的影响注:Ctrl 组表示等量稀释纳米材料的培养基处理,*表示与对照组相比差异显著(P <0.05)㊂Fig.5㊀Effects of nano -copper oxide (CuO NPs)and nano -cuprous oxide (Cu 2O NPs)on mitochondrialmembrane potential (MMP)of A549cellsNote:Ctrl group represents treatment with equal volume medium used to dilute nanomaterials;*represents significantdifference compared with the control (P <0.05).图6㊀纳米氧化铜(CuO NPs )和纳米氧化亚铜(Cu 2O NPs )在细胞培养基中铜离子的释放Fig.6㊀Copper ions release from nano -copperoxide (CuO NPs)and nano -cuprous oxide (Cu 2O NPs)in cell culture medium研究两者的肺细胞毒性差异,并从氧化应激和线粒体损伤等角度探索两者导致细胞毒性差异的潜在分子机制,并对比了游离态铜离子的影响㊂484㊀生态毒理学报第18卷本研究发现纳米农药中2种主要成分CuO NPs 和Cu 2O NPs 具有明显的肺细胞毒性作用㊂尤其是高浓度的CuO NPs 和Cu 2O NPs(ȡ50mg ㊃L -1)表现出极高的毒性,细胞存活率急剧下降㊂Cu 2O NPs 和CuO NPs 短期染毒造成A549细胞毒性的临界浓度约为5mg ㊃L -1和20mg ㊃L -1㊂Cu 2O NPs 可在比CuO NPs 浓度低得多的情况下对细胞产生毒性,说明Cu 2O NPs 比CuO NPs 毒性更大㊂其中CuO NPs 的研究结果与文献报道的细胞毒性效应结果基本一致[14-16]㊂同时,我们首次发现Cu 2O NPs 的细胞毒性作用较CuO NPs 明显更强,其毒性效应值得更多关注㊂氧化损伤是NPs 产生细胞毒性的公认分子机制之一[9,12-13]㊂因此,首先探讨了CuO NPs 和Cu 2ONPs 的细胞毒性差异是否与氧化应激诱导能力不同有关㊂通过ROS 检测,发现CuO NPs 和Cu 2O NPs 均能诱导ROS 的过量生成,并呈剂量依赖关系㊂但是Cu 2O NPs 的ROS 诱导效应明显强于CuO NPs ㊂由此,我们推断Cu 2O NPs 具有更强的细胞毒性作用很可能与其氧化损伤能力更强有关㊂ROS 的大量积累使得细胞处于氧化应激状态,可导致DNA 等生物大分子氧化损伤,进而导致细胞损伤和死亡㊂线粒体是ROS 生成的主要场所,而NPs 导致线粒体等细胞器的损伤也是其产生细胞毒性效应的主要分子机制[22]㊂因此,进一步探讨了CuO NPs 和Cu 2O NPs 对线粒体的损伤效应㊂CuO NPs 和Cu 2O NPs 均能以剂量依赖的方式导致A549细胞线粒体膜电位的降低㊂CuO NPs 和Cu 2O NPs 可通过诱导线粒体去极化作用,进而导致细胞凋亡,这也是两者产生细胞毒性的原因之一㊂同时,发现Cu 2O NPs 诱导A549细胞线粒体去极化的作用明显强于CuO NPs ,这很可能也是Cu 2O NPs 具有更强细胞毒性作用的重要原因㊂影响金属NPs 毒性的主要因素有尺寸㊁形貌㊁组分㊁氧化态以及释放的重金属含量等[22-23]㊂从SEM 可以看到Cu 2O NPs 更接近球形,而CuO NPs呈棒状,两者形貌差异可能进一步影响它们进入细胞的量㊂一般来说,NPs 粒径越小,细胞毒性越大㊂从水合粒径来看,Cu 2O NPs 与CuO NPs 没有很大差别,这可能不是导致毒性差异的主要原因㊂金属NPs 毒性与游离态铜离子的关系并无确切结论,仍存在一定争议㊂本研究的结果显示Cu 2O NPs 与CuO NPs 在细胞培养基中所释放的游离态铜离子浓度并无显著差别,并且均低于16mg ㊃L -1,在该浓度范围,铜离子并无显著细胞毒性㊂但是纳米颗粒进入细胞后颗粒表面的金属离子以及胞内溶解释放的铜离子的性质和毒性如何尚不清楚㊂此外,水和粒径检测表明我们所采用的纳米颗粒在水溶液中的水合半径达到400nm ,可能通过胞噬作用摄取进入细胞然后再次溶解产生毒性作用[24]㊂因此,后续工作需重点考察纳米颗粒进入细胞后的状态以及胞内游离态铜离子的浓度及价态,对于阐明两者毒性差异的影响因素非常关键㊂综上所述,所研究的Cu 2O NPs 对A549细胞毒性作用远大于CuO NPs ,并且与氧化应激和线粒体功能损伤有关,但是具体是何种因素导致两者毒性差异仍需进一步深入探究㊂通信作者简介:曹林英(1988 ),女,博士,副教授,主要研究方向为环境毒理学㊂共同通信作者简介:罗琳(1969 ),男,博士,教授,主要研究方向为污染处理处置与生态学㊂参考文献(References ):[1]㊀Li L,Xu Z L,Kah M,et al.Nanopesticides:A compre -hensive assessment of environmental risk is needed before widespread agricultural application [J ].Environmental Science &Technology,2019,53(14):7923-7924[2]㊀Usman M,Farooq M,Wakeel A,et al.Nanotechnology inagriculture:Current status,challenges and future opportu -nities [J].The Science of the Total Environment,2020,721:137778[3]㊀Kumar S,Nehra M,Dilbaghi N,et al.Nano -based smartpesticide formulations:Emerging opportunities for agri -culture [J].Journal of Controlled Release:Official Journal of the Controlled Release Society,2019,294:131-153[4]㊀谢婷,谭辉.无机铜氧化物在农药领域的应用专利技术综述[J].科技创新与应用,2019(19):23-24[5]㊀陈朗,姜辉,周艳明,等.纳米农药的环境安全性浅析[J].农药科学与管理,2018,39(5):30-38Chen L,Jiang H,Zhou Y M,et al.A brief analysis on the environmental safety of nano -enabled pesticides [J].Pesti -cide Science and Administration,2018,39(5):30-38(in Chinese)[6]㊀范文宏,刘通,史志伟,等.立方体和八面体微/纳米氧化亚铜对大型水蚤(Daphnia magna )的氧化胁迫和生理损伤[J].生态毒理学报,2016,11(5):40-48Fan W H,Liu T,Shi Z W,et al.Oxidative stress and第4期汤蕾等:纳米氧化亚铜和氧化铜的肺细胞毒性差异及影响因素485㊀physiological damage of cubic and octahedral Cu2O mi-cro/nanocrystals to Daphnia magna[J].Asian Journal ofEcotoxicology,2016,11(5):40-48(in Chinese)[7]㊀Gurianov Y,Nakonechny F,Albo Y,et al.Antibacterialcomposites of cuprous oxide nanoparticles and polyethy-lene[J].International Journal of Molecular Sciences,2019,20(2):439[8]㊀Rodríguez-Llamazares S,Mondaca M A,Badilla C,et al.PVC/copper oxide composites and their effect on bacterialadherence[J].Journal of the Chilean Chemical Society,2012,57(2):1163-1165[9]㊀Oberdörster G,Oberdörster E,Oberdörster J.Nanotoxicol-ogy:An emerging discipline evolving from studies of ul-trafine particles[J].Environmental Health Perspectives,2005,113(7):823-839[10]㊀Weichenthal S,Dufresne A,Infante-Rivard C.Indoor ul-trafine particles and childhood asthma:Exploring a poten-tial public health concern[J].Indoor Air,2007,17(2):81-91[11]㊀Jain A,Ranjan S,Dasgupta N,et al.Nanomaterials infood and agriculture:An overview on their safety con-cerns and regulatory issues[J].Critical Reviews in FoodScience and Nutrition,2018,58(2):297-317[12]㊀Manke A,Wang L Y,Rojanasakul Y.Mechanisms of nan-oparticle-induced oxidative stress and toxicity[J].BioMedResearch International,2013,2013:942916[13]㊀Bhabra G,Sood A,Fisher B,et al.Nanoparticles cancause DNA damage across a cellular barrier[J].NatureNanotechnology,2009,4(12):876-883[14]㊀孙婷婷,蒋澄宇.纳米氧化铜导致小鼠急性肺损伤[J].基础医学与临床,2012,32(4):386-389Sun T T,Jiang C Y.Copper oxide nanoparticles induce a-cute pulmonary injury in mice[J].Basic&Clinical Medi-cine,2012,32(4):386-389(in Chinese)[15]㊀Fahmy B,Cormier S A.Copper oxide nanoparticles in-duce oxidative stress and cytotoxicity in airway epithelialcells[J].Toxicology in Vitro,2009,23(7):1365-1371 [16]㊀Fu X.Oxidative stress induced by CuO nanoparticles(CuO NPs)to human hepatocarcinoma(HepG2)cells[J].Journal of Cancer Therapy,2015,6(10):889-895 [17]㊀Zhao L J,Ortiz C,Adeleye A S,et al.Metabolomics todetect response of lettuce(Lactuca sativa)to Cu(OH)2 nanopesticides:Oxidative stress response and detoxifica-tion mechanisms[J].Environmental Science&Technolo-gy,2016,50(17):9697-9707[18]㊀王淑玲,张玉喜,刘汉柱,等.氧化铜纳米颗粒对水稻幼苗根系代谢毒性的研究[J].环境科学,2014,35(5):1968-1973Wang S L,Zhang Y X,Liu H Z,et al.Phytotoxicity ofcopper oxide nanoparticles to metabolic activity in theroots of rice[J].Environmental Science,2014,35(5):1968-1973(in Chinese)[19]㊀Vignardi C P,Muller E B,Tran K,et al.Conventional andnano-copper pesticides are equally toxic to the estuarineamphipod Leptocheirus plumulosus[J].Aquatic Toxicolo-gy,2020,224:105481[20]㊀Zhang X X,Xu Z L,Qian X T,et al.Assessing the im-pacts of Cu(OH)2nanopesticide and ionic copper on the soil enzyme activity and bacterial community[J].Journalof Agricultural and Food Chemistry,2020,68(11):3372-3381[21]㊀Carley L N,Panchagavi R,Song X,et al.Long-termeffects of copper nanopesticides on soil and sedimentcommunity diversity in two outdoor mesocosm experi-ments[J].Environmental Science&Technology,2020,54(14):8878-8889[22]㊀Sengul A B,Asmatulu E.Toxicity of metal and metal ox-ide nanoparticles:A review[J].Environmental ChemistryLetters,2020,18(5):1659-1683[23]㊀García-Torra V,Cano A,Espina M,et al.State of the arton toxicological mechanisms of metal and metal oxidenanoparticles and strategies to reduce toxicological risks[J].Toxics,2021,9(8):195[24]㊀Manzanares D,Ceña V.Endocytosis:The nanoparticleand submicron nanocompounds gateway into the cell[J].Pharmaceutics,2020,12(4):371Ң。
世界最新医学信息文摘 2021年第21卷第23期101投稿邮箱:sjzxyx88@·综述·线粒体氧化应激与肿瘤的研究进展韩晓丹1,黄国锦1,2(通信作者*)(1.桂林医学院附属医院呼吸疾病实验室,广西 桂林 541000;2. 桂林医学院广西肝损伤与修复分子医学重点实验室,广西 桂林 541000;)摘要:线粒体在肿瘤生物能学中起关键作用,被认为是抗肿瘤治疗的潜在靶标。
线粒体产生的活性氧(reactive oxygen species, ROS )在生理浓度时,是维持生命活动的主要介质,但在病理状态下,其浓度升高后可打破抗氧化保护机制的平衡,导致氧化应激的发生。
氧化应激可以引起细胞的损伤,从而参与包括肿瘤在内的一些与细胞损伤和死亡相关的疾病。
本文将从线粒体氧化应激通过调控细胞信号传导通路、与腺苷酸活化蛋白激酶(AMP-activated protein kinase, AMPK )相互作用及调节自噬等方面,系统讨论线粒体氧化应激在肿瘤发生发展中的作用,进一步讨论线粒体氧化应激与肿瘤的治疗之间的关系并作一综述。
关键词:线粒体;氧化应激;活性氧;肿瘤中图分类号:R73 文献标识码:A DOI :10.3969/j.issn.1671-3141.2021.23.035本文引用格式:韩晓丹,黄国锦.线粒体氧化应激与肿瘤的研究进展[J].世界最新医学信息文摘,2021,21(23):101-103.Research Progress of Mitochondrial Oxidative Stress and TumorHAN Xiao-dan 1,HUANG Guo-jin 1,2*(1. Laboratory of Respiratory Diseases, the Affiliated Hospital of Guilin Medical University, Guilin Guangxi 541000; 2. GuangxiKey Laboratory of Molecular Medicine in Liver Injury and Repair, Guilin Medical University, Guilin Guangxi 541000)ABSTRACT: Mitochondria play a key role in tumor bioenergetics and are considered as potential targets for anti-tumor therapy. Mitochondrial metabolites, especially reactive oxygen species (ROS), are the main mediators for maintaining life activities at physiological concentrations. However, under pathological conditions, an increase in the concentration of reactive oxygen species breaks the balance of antioxidant protection mechanisms, which will cause oxidative stress. Oxidative stress can cause cell damage and participate in some diseases related to cell injury and death, including tumors. This article will systematically discuss the role of mitochondrial oxidative stress in tumorigenesis and development from the aspects of regulating cell signal transduction pathway, interacting with adenylate activated protein kinase (AMPK) and regulating autophagy, and further discuss the relationship between mitochondrial oxidative stress and tumor treatment.KEY WORDS: mitochondria; oxidative stress; reactive oxygen species; tumor0 引言在正常的生理情况下,细胞主要依靠葡萄糖进入细胞后通过有氧氧化和无氧酵解产生ATP 来供能[1,2]。
· 论著·下调GSK3β通过抑制ITPR1-GRP75-VDAC1复合体功能减轻衰老肾小管上皮细胞缺氧/复氧损伤倪海强 顾世琦 彭宣 宫念樵【摘要】 目的 探讨糖原合成酶激酶3β(GSK3β)对衰老小鼠原代肾小管上皮细胞(RTEC )缺氧/复氧(H/R )损伤的影响及其调控机制。
方法 将RTEC 分成为Young 组即正常生长的年轻RTEC 、Old 组即使用Etoposide 诱导的衰老RTEC 、Old+Ad-shNC+H/R 组即使用Etoposide 诱导衰老再转染腺病毒阴性对照(Ad-shNC )后进行H/R 处理,Old+Ad-shGSK3β+H/R 组即使用Etoposide 诱导衰老后再转染靶向沉默GSK3β的短发夹RNA 腺病毒(Ad-shGSK3β)后进行H/R 处理。
采用流式细胞术检测各组细胞凋亡水平和线粒体活性氧水平,采用免疫荧光染色法检测各组钙离子水平,采用蛋白质印迹法检测各组GSK3β、线粒体相关的内质网膜(MAM )相关蛋白肌醇1,4,5-三磷酸受体1(ITPR1)、电压依赖性阴离子通道1(VDAC1)、葡萄糖调节蛋白75(GRP75)表达及磷酸化水平,采用免疫共沉淀分析GSK3β与MAM 相关蛋白的相互作用。
结果 与Young 组比较,Old 组细胞凋亡水平、线粒体活性氧水平及线粒体钙离子水平均较高;与Old 组比较,Old+Ad-shNC+H/R 组细胞凋亡水平、线粒体活性氧水平及线粒体钙离子水平均较高;与Old+Ad-shNC+H/R 组比较,Old+Ad-shGSK3β+H/R 组细胞凋亡水平、线粒体活性氧水平及线粒体钙离子水平均较低,差异均有统计学意义DOI: 10.3969/j.issn.1674-7445.2024018基金项目:国家自然科学基金(82170772、82370759);湖北陈孝平科技发展基金会青年科学专项基金(CXPJJH122001-2210)作者单位: 430030 武汉,华中科技大学同济医学院附属同济医院器官移植研究所 器官移植教育部重点实验室 国家卫生健康委员会器官移植重点实验室 中国医学科学院器官移植重点实验室作者简介:倪海强(ORCID 0000-0002-6010-603X ),博士研究生,研究方向为肾脏缺血-再灌注损伤,Email :***************通信作者:宫念樵(ORCID 0000-0001-7634-1440),博士,主任医师,研究方向为器官移植、移植免疫、干细胞治疗和器官老化,Email :***************第 15 卷 第 3 期器官移植Vol. 15 No.3 2024 年 5 月Organ Transplantation May 2024 第 3 期倪海强等.下调GSK3β通过抑制ITPR1-GRP75-VDAC1复合体功能减轻衰老肾小管上皮细胞缺氧/复氧损伤·407·(均为P<0.05)。
Update on Mitochondrial Reactive Oxygen SpeciesMitochondrial Reactive Oxygen Species.Contribution to Oxidative Stress and Interorganellar SignalingDavid M.Rhoads*,Ann L.Umbach,Chalivendra C.Subbaiah,and James N.SiedowSchool of Life Sciences,Arizona State University,Tempe,Arizona85287–4501(D.M.R.,C.C.S.);and Developmental,Cell and Molecular Biology Group and Biology Department,Duke University, Durham,North Carolina27708–1000(A.L.U.,J.N.S.)The inner membrane of a plant mitochondrion con-tains the mitochondrial electron transport chain (mtETC),consisting of protein complexes that use an energy source-derived reductant to form a proton gradient across the membrane.This proton gradient drives ATP synthesis,a primary mitochondrial func-tion.Ultimately,electrons in the mtETC are transferred through one of two pathways from ubiquinone to a terminal oxidase,where oxygen is reduced to water. Specifically,electrons pass through either the‘‘stan-dard’’cytochrome pathway,which includes two sites of proton pumping downstream of the ubiquinone pool, to cytochrome c oxidase(COX),or the alternative pathway,which consists of the cyanide-insensitive alternative oxidase(AOX)and has no proton-pumping sites after the ubiquinone pool(Finnegan et al.,2004). Because the mtETC harbors electrons with sufficient free energy to directly reduce molecular oxygen,it is considered the unavoidable primary source of mito-chondrial reactive oxygen species(mtROS)production, a necessary accompaniment to aerobic respiration. Formation of mtROS takes place under normal res-piratory conditions but can be enhanced in response to a range of abnormal conditions,including exposure to biotic and abiotic stresses.The marked reactivity of ROS toward biological molecules,including lipids, proteins,and nucleic acids,requires multiple mecha-nisms for keeping mtROS levels under control,includ-ing pathways that attenuate mtROS formation in response to imposed stresses and protective,antioxi-dant enzyme systems.However,when mtROS forma-tion exceeds normal levels despite the operation of these protective mechanisms,there are downstream consequences for the cell,including altered gene ex-pression and even programmed cell death(PCD).Not simply agents of damage,mtROS also play roles in the signaling required to bring about these changes.The cell ultimately must strike a balance between the level of ROS required to elicit an appropriate response to a changing condition while at the same time keeping ROS levels sufficiently low to prevent large-scale cellular damage.On the other hand,cells must be able to determine a mtROS-initiated,extreme measure at the cellular level is appropriate for the good of the plant as a whole(e.g.the hypersensitive response to pathogen attack).This article provides an overview of our cur-rent understanding of plant mtROS.While much re-mains to be established,mtROS clearly play important roles in the responses of plants to the variety of envi-ronmental conditions they experience on a regular basis,as well as more extreme environmental stresses.GENERATION OF mtROSThe known sites of mtROS production in the mtETC are complexes I and III,where superoxide anion(O22)is formed and in turn is reduced by dismutation to H2O2 (Raha and Robinson,2000;Møller,2001;Sweetlove and Foyer,2004).H2O2,a compound of relatively low tox-icity,can react with reduced Fe21and Cu1to produce highly toxic hydroxyl radicals and,being uncharged, can also penetrate membranes and leave the mitochon-drion(Grene,2002;Sweetlove and Foyer,2004).The ubisemiquinone intermediate formed at complexes I and III is the principal electron donor to oxygen,al-though other complex I sites are also potential donors (Raha and Robinson,2000;Sweetlove and Foyer,2004). Thus,the overall reduction level of the mitochondrial ubiquinone pool will be the primary determinate of mtROS output(Sweetlove and Foyer,2004).The amount of ROS produced by mitochondria and the fraction of total cellular ROS that come from mito-chondria are difficult to determine,in part because ROS levels in general are difficult to measure accu-rately(Veljovic-Jovanovic et al.,2002;Halliwell and Whiteman,2004).For isolated mitochondria and sub-mitochondrial particles,ROS evolution varies with conditions but falls within the wide range of0.2to 30.0nmol min21mg protein21for either H2O2or super-oxide(studies compiled in Møller,2001;Popov et al., 2003).While we are unaware of any quantitative mea-surements of ROS production by mitochondria in vivo, estimates(Foyer and Noctor,2003)indicate that the in situ level of mtROS evolution normally will be con-siderably less than that of chloroplasts or peroxisomes in the light due to the operation of photosynthesis and*Corresponding author;e-mail drhoads@;fax480–965–6899.The author responsible for distribution of materials integral to the findings presented in this article in accordance with the policy described in the Instructions for Authors()is: David M.Rhoads(drhoads@)./cgi/doi/10.1104/pp.106.079129.photorespiration.However,in the dark or in non-green tissues,mitochondria will be a major source of ROS(e.g.Puntarulo et al.,1988).Production of mtROS will increase if the rate of electrons leaving the mtETC through the terminal oxidases is slowed and/or the rate of electron input increases in excess of the ability of the two respiratory pathways to process the elec-trons,leading to an overreduced ubiquinone pool. This principle has been demonstrated in isolated mi-tochondria,where rates of H2O2and superoxide gen-eration show a substrate-dependent increase upon the addition of specific inhibitors of either the cytochrome pathway or AOX(Popov et al.,1997;Møller,2001). Mitochondria in situ show the same effect.Studies using whole cells have shown increased ROS produc-tion specifically from mitochondria in the presence of the cytochrome pathway inhibitor antimycin A(AA; Maxwell et al.,1999;Yao et al.,2002).Of primary interest in mitochondrial research is the likelihood that naturally occurring physiological and environmental conditions encountered by plants can give rise to an overreduced ubiquinone pool and con-comitant increased mtROS production.For exam-ple,the endogenous signaling molecule nitric oxide (NO)is an inhibitor of COX,but not AOX(Millar and Day,1996),at concentrations achieved in vivo during normal seedling development(Caro and Puntarulo, 1999).Therefore,NO production could lead to in-creased mtROS formation.Many stresses cause oxida-tive damage in plant tissues,and increases in mtROS resulting from mtETC perturbations have been impli-cated as at least partly responsible for the damage and plant responses observed in several of these cases, including chilling(Prasad et al.,1994a,1994b;Purvis et al.,1995),salt stress(Herna´ndez et al.,1993;Mittova et al.,2003),and phosphate deficiency(Juszczuk et al., 2001;Parsons et al.,1999;Malusa`et al.,2002).The preceding examples together with other studies strongly suggest that mtROS are involved in the re-sponses of plants to stresses,as well as other plant processes.This means that plants are able to detect changes in mtROS output against a background of ROS production from other sources.Because mtROS output is predicted to be relatively constant during the course of a day/night cycle(Foyer and Noctor,2003; Sweetlove and Foyer,2004),changes in mtROS levels, even if low in magnitude,could be distinguished and registered by local detection mechanisms.The concept of local detection is consistent with accumulating evi-dence that the specific source of ROS is important in determining appropriate cellular responses(Dutilleul et al.,2003;Laloi et al.,2004;Clifton et al.,2005).A change in mtROS level is likely to have a consequence, including direct damage if levels increase(in which case damaged molecules could be part of signaling) or/and as early participants themselves in a signaling pathway(s).Therefore,as discussed below,mitochon-dria must be capable of controlling their ROS levels under normal conditions,detoxifying excess ROS, repairing oxidative damage arising from ROS forma-tion,and modulating mtROS production appropri-ately for signaling.CONSEQUENCES OF mtROS IN MITOCHONDRIA Oxidative Damage to Mitochondrial Lipids Peroxidation of mitochondrial membrane polyun-saturated fatty acids is initiated by the abstraction of a hydrogen atom by ROS,especially by hydroxyl radicals. This leads to the formation of cytotoxic lipid aldehydes, alkenals,and hydroxyalkenals(HAEs),such as the much-studied4-hydroxy-2-nonenal(HNE)and malon-dialdehyde.Inhibition of the mtETC with AA can generate mitochondrial HAEs to levels similar to those generated by general oxidative stress through chemical treatments such as H2O2or menadione(a compound that causes superoxide production;Sweetlove et al., 2002;Winger et al.,2005).Once formed,lipid perox-idation products can cause cellular damage by reacting with proteins,other lipids,and nucleic acids.Key oxylipins and smaller,lipid-derived reactive electro-phile species may also be produced from lipid perox-idation(Alme´ras et al.,2003),but,to our knowledge, there is no direct evidence of these compounds being produced in plant mitochondria from oxidative stress.Effects of ROS on Mitochondrial ProteinsProteins can be damaged and/or inhibited by oxi-dative conditions in several ways,including:(1)direct oxidation of amino acids by ROS,such as the oxidation of Cys residues to form disulfide bonds,oxidation of Met residues to form Met sulfoxide,and oxidation of Arg,Lys,Pro,and Thr residues,which creates car-bonyl groups in the side chains(Berlett and Stadtman, 1997;Dean et al.,1997);(2)oxidation that breaks the peptide backbone(Dean et al.,1997);(3)reactions with lipid peroxidation products(such as HNE);(4)reac-tions with reactive nitrogen species that are formed by reaction of NO with ROS(Sakamoto et al.,2003);and (5)direct ROS interaction with metal cofactors,illus-trated by the TCA cycle iron-sulfur enzyme aconitase, which is sensitive to H2O2(Verniquet et al.,1991)and superoxide(Flint et al.,1993).Proteomics approaches have been undertaken to de-termine the damaging effects of oxidative stress on mi-tochondrial proteins(Sweetlove et al.,2002;Kristensen et al.,2004;Taylor et al.,2005).These studies primarily used chemical or environmental stresses to impose general,non-mitochondria-specific oxidative condi-tions,though some studies also employed AA(see below),which causes mtROS production specifically. Proteins that accumulate but that are degraded(as determined by identification of fragments)as a result of high ROS levels imposed by addition of H2O2or menadione were identified(Sweetlove et al.,2002). Oxidatively damaged mitochondrial proteins include subunits of the pyruvate decarboxylase complex,sub-units of ATP synthase,and enzymes of the TCA cycle.Rhoads et al.Further,mitochondrial proteins oxidized by treatment with H2O2were‘‘tagged’’with dinitrophenylhydrazine, which forms covalent bonds with carbonyl groups resulting from oxidation of amino acids(Kristensen et al.,2004).Thirty-eight labeled,oxidized mitochon-drial proteins were identified.Several of these proteins were among those previously observed to be damaged by oxidative stresses(Sweetlove et al.,2002;Taylor et al.,2002,2005).Although such proteomics experi-ments do not survey the full mitochondrial proteome, it is clear that several important mitochondrial pro-teins are damaged by general oxidative stresses and therefore could be damaged by mtROS.Most signifi-cantly for this discussion,many proteins degraded during more general oxidative treatments were also degraded following AA treatment(Sweetlove et al., 2002),demonstrating that mtROS specifically can dam-age key mitochondrial proteins.Treatment of mitochondria with HNE or paraquat (which causes superoxide formation in chloroplasts and mitochondria)or cold or drought treatment of plants leads to formation of a covalent HNE-derived adduct of the lipoic acid moiety of several mitochon-drial enzymes,including Gly decarboxylase(an enzyme in the photorespiratory pathway),2-oxoglutarate de-hydrogenase(a TCA cycle enzyme),and pyruvate decarboxylase(Millar and Leaver,2000;Taylor et al., 2002,2005).Although it has not been proven that the damage is from mtROS specifically or mitochondrial lipid peroxidation products produced during these stresses,inhibition of the mtETC does result in elevated malondialdehyde levels(Taylor et al.,2005;Winger et al.,2005).These results indicate that oxidative con-ditions due to production of mtROS could cause dam-age to proteins through formation of HAEs that react with the lipoic acid moiety.HNE can also form adducts with Cys,His,and Lys residues,causing altered en-zyme function(Schaur,2003).One of these reactions is likely the cause of HNE inhibition of AOX(Winger et al., 2005).While this seems counterintuitive since one role of AOX is to help prevent mtROS formation,it may allow cells to sense an extreme stress that dramatically enhances the accumulation of mtROS and initiation of a more extreme response,such as PCD(see below). While oxidative damage to proteins occurs under stressful conditions,it has also been shown to be a normal part of Arabidopsis(Arabidopsis thaliana)leaf maturation(Johansson et al.,2004).In addition,selec-tive protein oxidation has been suggested to have a role in controlling the course of metabolic activity dur-ing seed germination where the mitochondrial ATP synthase was one of the targeted proteins(Job et al., 2005).If this is true,then mtROS can also impact cell physiology through this oxidative mechanism.Oxidative Damage to mtDNAHydroxyl radicals are highly reactive and can dam-age nuclear and mitochondrial DNA,which cells try to repair(Roldan-Arjona et al.,2000;Doudican et al.,2005).Accumulated damage to mtDNA,which can be caused by mtROS over the course of the lives of animals, causes decreased mitochondrial function and can con-tribute to aging and diseases(Allen,1996;Raha and Robinson,2000;Trifunovic et al.,2004;Wallace,2005). For mtDNA,damage from oxidative stress(including mtETC inhibition by AA)and effects on mitochondrial function have been studied in other organisms(Raha and Robinson,2000;Doudican et al.,2005;Wallace, 2005).However,there is a paucity of such studies using plant models.In the long term,plants could avoid the consequences of DNA damage by replacing organs that contain damaged genomic or mtDNA.On the other hand,there could be an accumulation of mutations in meristematic tissues that are maintained in cells of organs that develop from these tissues, though we are not aware of any studies addressing this issue.One factor helping prevent the adverse effects of mtDNA damage in plant cells is the redun-dancy of genomic sequences in each mitochondrion (e.g.Mackenzie et al.,1994;Backert et al.,1997).The copy number of individual genomic regions can help determine levels of gene expression(Muise and Hauswirth,1995;Hedtke et al.,1999)and,therefore, could help dictate the extent of the effects of mutation caused by ROS.In addition,the number of mitochondria in plant cells ranges from hundreds to,perhaps,thou-sands(Douce,1985).Heteroplasmy and recombina-tion help to clear deleterious mutations from damaged mitochondrial genomes and maintain their functional integrity(Barr et al.,2005).Although a low rate of recombination in mitochondria seems to cause an accumulation of nonsynonymous mutations in mito-chondrial genomes at an accelerated rate relative to nuclear genomes,the rate of accumulation is slow enough that severefitness losses can only occur on a time scale of tens of millions of years(Lynch and Blanchard,1998).Nevertheless,these‘‘long-term’’strat-egies would not alleviate consequences of potential mtDNA damage in cells by severe oxidative stress.ANTIOXIDANT COMPOUNDS AND PROTECTIVE ENZYMES IN MITOCHONDRIAPlant mitochondria can modulate superoxide pro-duction from the mtETC by two mechanisms that act to keep the ubiquinone pool reduction level low.The first,AOX,is not inhibited by the proton gradient across the inner membrane and can function when the cytochrome pathway is impaired(Finnegan et al., 2004).That AOX might act to maintain a basal ubi-quinone pool reduction state was initially proposed by Purvis and Shewfelt(1993)and is supported by stud-ies with roots treated with a cytochrome pathway inhibitor(Millenaar et al.,1998).Further,operation of AOX diminishes mtROS production(Purvis,1997, Popov et al.,1997;Maxwell et al.,1999).Increased ex-pression of alternative mitochondrial NAD(P)H de-hydrogenases,which do not translocate protons,oftenMitochondrial Reactive Oxygen Speciesaccompanies increased AOX expression(Clifton et al., 2005).Use of these enzymes serves as an alternative to complex I and could further help decrease mtROS production.The second,uncoupling protein(UCP), also is found in the inner mitochondrial membrane. UCP uncouples by facilitating a proton leak across the membrane and consequently removes inhibition of the mtETC(Sluse and Jarmuszkiewicz,2004;Hourton-Cabassa et al.,2004).Like AOX,UCP function in isolated mitochondria decreases ROS formation (Kowaltowski et al.,1998;Casolo et al.,2000).On the other hand,ROS are actually required for UCP activity (Pastore et al.,2000;Considine et al.,2003),a direct activator being HNE(Smith et al.,2004).This ROS requirement by UCP is in contrast to the effect of ROS on AOX.Exposure of AOX to experimental oxidative stress can drive the protein into the inactive,disulfide-linked form(Vanlerberghe et al.,1999;A.L.Umbach, unpublished data),and HNE inhibits AOX(Winger et al.,2005).Therefore,while both AOX and UCP may act to forestall mtROS production,only UCP may be able to operate when ROS levels become increased. Once superoxide has been generated from the mtETC,thefirst step in detoxification,dismutating superoxide to H2O2,is catalyzed by mitochondrial manganese superoxide dismutase(MnSOD;Kliebenstein et al.,1998;Møller,2001).The presence of catalase, though reported in maize(Zea mays)mitochondria, does not appear to be typical(Sweetlove and Foyer, 2004).Consequently,H2O2is removed by other mech-anisms in plant mitochondria.One of these is the mitochondrial glutathione-ascorbate cycle(Jime´nez et al.,1997;Chew et al.,2003).Ascorbate peroxidase reduces H2O2to H2O using ascorbate,which is re-reduced by the sequential action of monodehydroas-corbate reductase and dehydroascorbate reductase, which uses reduced glutathione in thefinal reduction stly,glutathione reductase regenerates re-duced glutathione.Peroxiredoxins also could reduce mitochondrial H2O2.These enzymes use reduced thi-oredoxin or glutathione sequentially with glutathione as reductant sources,which in turn are reduced by thioredoxin reductase and glutathione reductase (Sweetlove and Foyer,2004).An Arabidopsis mito-chondrial type II peroxiredoxin(PrxII F)has been iden-tified by proteomics and characterized(Heazlewood et al.,2004;Finkemeier et al.,2005),and plant mito-chondria contain thioredoxins(Sweetlove and Foyer, 2004).Ultimately,the electrons for all these systems are derived from mitochondrial NADPH,and a high level of employment of these systems could affect overall mitochondrial and even cellular redox status.In addition to directly detoxifying ROS,these and other enzyme systems,together with their electron donors,may be involved in repairing lipid peroxidation damage and some forms of protein oxidation.For ex-ample,glutathione S-transferases(GSTs)can inactivate HNE by converting it to the glutathione conjugate GS-HNE,and there is evidence for at least one GST in plant mitochondria(Heazlewood et al.,2004).The type II peroxiredoxin/thioredoxin system mentioned above could also reduce lipid peroxides(Rouhier et al., 2004).Also,although most oxidative damage to pro-teins is irreversible and results in protein degradation, Arabidopsis mitochondria contain a protein disulfide isomerase that,functioning together with thioredoxin and NADPH,can reduce disulfides resulting from protein oxidation(Sweetlove et al.,2002).It remains to be determined whether these mechanisms function in plant mitochondria to decrease oxidative damage. Mitochondria can control their oxidative state indi-rectly through metabolism as well.One example is the g-aminobutyrate shunt for succinate synthesis,which bypasses two enzymes of the TCA cycle that are known to become inactivated by oxidative stress.Plants that lack this bypass show increased tissue damage and H2O2 levels,perhaps because less NAD(P)H is generated in the mitochondria than is needed to maintain reducing conditions(Bouche´et al.,2003;Bouche´and Fromm, 2004).Another example is the biosynthesis of ascorbate, the last step of which involves the mtETC(Siendones et al.,1999;Bartoli et al.,2000;Millar et al.,2003).Stresses on the mtETC could perturb ascorbate synthesis with drastic effects on mitochondrial and cellular redox bal-ance(Millar et al.,2003),as exemplified by ascorbate-deficient Arabidopsis mutants(Pavet et al.,2005).The antioxidant systems of plant mitochondria could connect mtROS to other cellular processes through their effects on the overall availability and reduction state of glutathione,ascorbate,thioredoxin, and NADPH in the cell.For example,glutathione and ascorbate induce expression of plant defense genes and are proposed signals of cellular redox status and PCD(Foyer et al.,1997;Noctor and Foyer,1998;de Pinto et al.,2002).Glutathione also is a potential com-ponent of cold stress signaling(Foyer et al.,1997),and thioredoxin is involved in redox activation of numer-ous enzyme systems(Gelhaye et al.,2005).Mitochon-drial events can also modify rates of ROS evolution elsewhere in the cell.The mtETC is recognized as needed for processing excess reductant produced by the light reactions of photosynthesis.Should reductant levels originating from the chloroplast exceed the processing ability of the mtETC,not only would mtROS production likely increase,but chloroplast ROS pro-duction could also increase due to an overly reduced photosynthetic reaction system(Fernie et al.,2004; Finnegan et al.,2004).Therefore,mitochondrial anti-oxidant systems are positioned to affect ROS and re-dox status and modulate many cellular functions(Foyer et al.,1997;Noctor and Foyer,1998).mtROS AS SIGNALS AFFECTING NUCLEAR GENE EXPRESSION AND CELL FATEAltered Nuclear Gene ExpressionAltered mitochondrial function can cause altered nu-clear gene expression through mitochondria-to-nucleusRhoads et al.signaling,which is referred to as mitochondrial retro-grade regulation(MRR).This phenomenon has been studied in yeast,animal cell cultures,and plants(for review,see Butow and Avadhani,2004;Rhoads and Vanlerberghe,2004).There are several interesting ex-amples in which nuclear gene expression was altered in plants by mitochondrial dysfunction caused by nuclear mutation or transgenic expression of mito-chondrial proteins(Kushnir et al.,2001;Go´mez-Casati et al.,2002;Rhoads et al.,2005).Although not directly demonstrated in these examples,there is evidence for the importance of mtROS in plant MRR.Inhibition of the cytochrome pathway by AA results in production of mtROS(Maxwell et al.,1999;Yao et al.,2002)and altered nuclear gene expression(Saisho et al.,1997;Yu et al.,2001;Karpova et al.,2002;Maxwell et al.,2002). The direct involvement of mtROS in MRR in plants is strongly suggested by the dramatic reduction of AA-initiated gene induction observed when antioxidants are added prior to AA(Maxwell et al.,1999). Although inhibition by antioxidants of gene induc-tion from cytochrome pathway disruption shows the importance of ROS in this process,it does not address whether mtROS leave the mitochondria and initiate gene expression changes.One of the two ubisemi-quinone binding sites of complex III is close to the mitochondrial intermembrane space,providing a mechanism for superoxide production accessible to the cytoplasm(Møller,2001).Observations of H2O2 apparently escaping from mitochondria at specific sites(Yao et al.,2002)in response to AA treatment also suggest that direct effects of mtROS outside the mitochondria,such as initiation of gene expression changes,are possible and that release may be through specific channels or pores.Although clearly not defin-itive in plants,related observations support the hy-pothesis that plant mtROS are released at specific sites. Gene induction from AA treatment of suspension-cultured tobacco(Nicotiana tabacum)cells can be in-hibited by inhibitors of the permeability transition pore(Maxwell et al.,2002),and superoxide has been measured directly leaving respiring isolated rat mito-chondria by an apparently channel-mediated mecha-nism(Han et al.,2003).However,changes in the mitochondria initiated by increased mtROS may pro-duce secondary signals from local(in or close to mitochondria)ROS detection mechanisms that are transmitted to the nucleus.As has been discussed elsewhere(Rhoads and Vanlerberghe,2004),any of the known signaling pathways or pathways yet to be discovered could be involved in transmitting a signal to the nucleus.Attractive candidates are those signal-ing components that are likely involved in oxidative stress sensing and/or redox signaling.Several poten-tial signaling proteins were localized to mitochondria (Heazlewood et al.,2004)and could be involved as local detection mechanisms for increased mtROS and initiation of signaling to the nucleus.Among the common themes in plant MRR following mtETC inhibition are induction of genes encoding(1)proteins involved in mitochondrial respiration via reactions other than those in the cytochrome pathway, including AOX and alternative NAD(P)H dehydro-genases;(2)antioxidant enzymes,such as GSTs and a monodehydroascorbate reductase;and(3)proteins for plant defenses against biotic and abiotic stresses(Saisho et al.,1997;Yu et al.,2001;Maxwell et al.,2002;Clifton et al.,2005).At least for the Arabidopsis AOX1a gene, induction is most likely the result of increased tran-scription controlled at a promoter region(Dojcinovic et al.,2005;Zarkovic et al.,2005).In plants that lack a fully functioning mtETC due to mutations in the mitochondrial genome,a new cellular homeostasis must be attained.Altered nuclear gene expression is part of this new homeostasis.Mutants exhibit altered expression of genes encoding AOXs, heat shock proteins,and antioxidant enzymes(Karpova et al.,2002;Dutilleul et al.,2003;Kuzmin et al.,2004). Thus,permanent alteration of mtETC function results in constitutively altered nuclear gene expression.In-terestingly,tobacco plants impaired in complex I func-tion exhibit increased respiration(Gutierres et al., 1997)but lower cellular H2O2levels,likely due to the observed increased antioxidant enzyme expression and activity.These plants also exhibit increased levels of NAD,NADH,and NADPH(Dutilleul et al.,2005), but no change in redox components glutathione and ascorbate(Dutilleul et al.,2003)Overall,they have greatly altered metabolism to adjust to severely altered mitochondrial function but are still dramatically al-tered phenotypically(Gutierres et al.,1997;Dutilleul et al.,2005).Because these plants have already attained an interesting new homeostasis,it is not clear what signaling from the mitochondria is responsible for changes in nuclear gene expression.Nevertheless,it is possible that increased mtROS production and/or shifted redox status directed by mitochondria contrib-ute to signaling(Dutilleul et al.,2003,2005). Finally,mtROS could diffuse from mitochondria and contribute to other forms of signaling by increas-ing ROS at other locations in the cell(such as the chloroplast;see e.g.op den Camp et al.,2003,and refs. therein),which could result in altered nuclear gene expression.PCD and Response to PathogensPCD is an important part of certain plant responses to stresses and includes the hypersensitive response to pathogens.An early cellular signal for this process is frequently an increase in tissue ROS production due to plasma membrane NADPH oxidase activity(Overmyer et al.,2003).Plant mitochondrial responses to PCD signals are similar to those of animal mitochondria and include undergoing a permeability transition,re-lease of cytochrome c,and a decrease in ATP production (Xie and Chen,2000;Arpagaus et al.,2002;Tiwari et al., 2002;Krause and Durner,2004).Increases in mtROS production are coincident with these processes,probablyMitochondrial Reactive Oxygen Species。