Acute and chronic toxicity of endosulfan to the polychaeta, Perinereis aibuhitensis
LPS、肿瘤细胞坏死释放物质对小鼠黑色素瘤B16细胞侵袭、转移能力的影响
![LPS、肿瘤细胞坏死释放物质对小鼠黑色素瘤B16细胞侵袭、转移能力的影响](https://img.taocdn.com/s3/m/68b55e07aeaad1f347933f4a.png)
华中科技大学硕士学位论文LPS、肿瘤细胞坏死释放物质对小鼠黑色素瘤B16细胞侵袭、转移能力姓名:陈欣申请学位级别:硕士专业:生物化学与分子生物学指导教师:张桂梅;冯作化2010-12-27中文摘要目的 本文拟研究LPS,肿瘤细胞坏死释放物质(NTC-Ms)对小鼠黑色素瘤B16细胞侵袭、转移能力的影响,并从分子水平探讨其影响的可能机制,从而为肿瘤治疗提供新的思路。
方法(1)分别用LPS,NTC-Ms,以及Resveratrol联合NTC-Ms作用于B16细胞48h,粘附实验检测三组刺激因素分别对B16细胞粘附能力的影响。
(2)分别用LPS,NTC-Ms,以及Resveratrol联合NTC-Ms作用于B16细胞48h,软琼脂迁移实验检测三组刺激因素分别对B16细胞迁移能力的影响。
(3)分别用LPS,NTC-Ms,以及Resveratrol联合NTC-Ms作用于B16细胞48h,运动实验(Transwell 小室法)检测三组刺激因素分别对B16细胞侵袭和趋化运动能力的影响。
(4)分别用LPS,NTC-Ms,以及Resveratrol联合NTC-Ms作用于B16细胞48h,RT-PCR 法检测B16细胞TLR4,MMP-9 mRNA的表达。
(5)分别用LPS,NTC-Ms,以及Resveratrol联合NTC-Ms作用于B16细胞48h,,明胶酶谱实验检测B16细胞分泌的MMP-9的活性。
结果(1)LPS,NTC-Ms刺激后,B16细胞粘附基底膜的能力均增强;Resveratrol 联合NTC-Ms刺激后,B16细胞对基底膜的粘附能力较单用NTC-Ms减弱。
(2)LPS,NTC-Ms刺激后,迁移至软琼脂底面的B16细胞数均增多;Resveratrol联合NTC-Ms刺激后,迁移至软琼脂底面的B16细胞数减少。
(3)LPS,NTC-Ms 刺激后,B16细胞侵袭和趋化运动能力均增强;Resveratrol联合NTC-Ms刺激后,B16细胞侵袭和趋化运动能力减弱。
lps英文简介(word整理版)
![lps英文简介(word整理版)](https://img.taocdn.com/s3/m/667ad3fe4693daef5ef73dd8.png)
LipopolysaccharideLipo poly saccharides (LPS), also known aslipoglycans and endotoxin, are large moleculesconsisting of a lipid and a polysaccharide(多糖)composed of O-antigen, outer core and inner corejoined by a covalent bond; they are found in theouter membrane of Gram-negative bacteria, andelicit strong immune responses in animals.The term lipo oligo saccharide("LOS") is used torefer to a low molecular weight form of bacteriallipopolysaccharideStructure of a lipopolysaccharide DiscoveryThe toxic activity of LPS was first discovered and termed "endotoxin" (内毒素) by Richard Friedrich Johannes Pfeiffer, who distinguished between exotoxins(外毒素), which he classified as a toxin that is released by bacteria into the environment when bacteria are killed, and endotoxins, which he considered to be a toxin kept "within" the bacterial cell and released only after destruction of the bacterial cell wal l.[1]:84 Subsequent work showed that release of LPS from gram negative microbes(细菌)does not require the necessity of destruction of the bacterial cell wall, but rather, LPS is secreted as part of the normal physiological activity of membrane vesicle(膜囊泡)trafficking in the form of bacterial outer membrane vesicles (OMVs), which may also contain other virulence factors and proteins.[2]Today, the term 'endotoxin' is mostly used synonymously with LPS,[3] although there are a few molecules secreted by other bacteria that are not related to LPS, such as the so-called delta endotoxin proteins secreted by Bacillus thuringiensis.Functions in bacteriaLPS is the major component of the outer membrane of Gram-negative bacteria, contributing greatly to the structural integrity(n.完整)of the bacteria, and protectingthe membrane from certain kinds of chemical attack. LPS also increases the negative charge of the cell membrane and helps stabilize the overall membrane structure. It is of crucial importance to gram-negative bacteria, whose death results if it ismutated or removed. LPS is an endotoxin, and induces a strong response fromnormal animal immune systems. It has also been implicated in non-pathogenic aspects of bacterial ecology, including surface adhesion, bacteriophage(n. [病毒] 噬菌体;抗菌素)sensitivity, and interactions with predators(捕食者)such as amoebae(变形虫).LPS is required for the proper conformation of Omptin activity; however, smoothLPS will sterically hinder omptins.CompositionIt comprises three parts:1.O antigen (or O antigen| Opolysaccharide)2.Core oligosaccharide3.Lipid AO-antigenA repetitive glycan polymer containedwithin an LPS is referred to as the Oantigen, O polysaccharide, or OThe saccharolipid Kdo2-Lipid A.Glucosamine(氨基葡萄糖) residues in blue,Kdo residues in red, acyl chains in black andphosphate groups in green.side-chain of the bacteria. The O antigen is attached to the core oligosaccharide, and comprises the outermost domain of the LPS molecule. The composition of the O chain varies from strain to strain. For example, there are over 160 different O antigen structures produced by different E. coli strains.[4] The presence or absence of O chains determines whether the LPS is considered rough or smooth. Full-length O-chains would render the LPS smooth, whereas the absence or reduction of O-chains would make the LPS rough.[5] Bacteria with rough LPS usually have more penetrable cell membranes to hydrophobic antibiotics, since a rough LPS is more hydrophobic.[6] O antigen is exposed on the very outer surface of the bacterial cell, and, as a consequence, is a target for recognition by host antibodies.CoreMain article: Core oligosaccharideThe Core domain always contains an oligosaccharide component that attaches directly to lipid A and commonly contains sugars such as heptose and3-deoxy-D-mannooctulosonic Acid (also known as KDO, keto-deoxyoctulosonate).[7] The LPS Cores of many bacteria also contain non-carbohydrate components, such as phosphate, amino acids, and ethanolamine substituents.Lipid ALipid A is, in normal circumstances, a phosphorylated glucosamine disaccharide decorated with multiple fatty acids. These hydrophobic fatty acid chains anchor the LPS into the bacterial membrane, and the rest of the LPS projects from the cell surface. The lipid A domain is responsible for much of the toxicity of Gram-negative bacteria. When bacterial cells are lysed by the immune system, fragments of membrane containing lipid A are released into the circulation, causing fever, diarrhea, and possible fatal endotoxic shock (also called septic shock). The Lipid A moiety is a very conserved component of the LPS.[8]Lipo oligo saccharidesLipooligosaccharides (LOS) are glycolipids found in the outer membrane of some types of Gram negative bacteria, such as Neisseria spp. and Haemophilus spp. The term is synonymous with the low molecular weight form of bacterial LPS.[9] LOS plays a central role in maintaining the integrity and functionality of the outer membrane of the Gram negative cell envelope. Lipooligosaccharides play an important role in the pathogenesis of certain bacterial infections because they are capable of acting as immunostimulators and immunomodulators.[9] Furthermore, LOS molecules are responsible for the ability of some bacterial strains to display molecular mimicry and antigenic diversity, aiding in the evasion of host immune defenses and thus contributing to the virulence of these bacterial strains.Chemically, lipooligosaccharides lack O-antigens and possess only the a lipidA-based outer membrane-anchoring moiety, and an oligosaccharide core.[10] In thecase of Neisseria meningitidis, the lipid A portion of the molecule has a symmetricalstructure and the inner core is composed of 3-deoxy-D-manno-2-octulosonic acid(KDO) and heptose (Hep) moieties. The outer core oligosaccharide chain variesdepending on the bacterial strain.[9][10] The term lipooligosaccharide is used to refer tothe low molecular weight form of bacterial lipopolysaccharides, which can becategorized into two forms: the high molecular weight (Mr, or smooth) formpossesses a high molecular weight, repeating polysaccharide O-chain, while the lowmolecular weight (low-Mr or rough) form, lacks the O-chain but possesses a shortoligosaccharide in its place.[9]LPS modifications(修饰)The making of LPS can be modified in order to present a specific sugar structure.Those can be recognised by either other LPS (which enables to inhibit LPS toxins) orglycosyltransferases that use those sugar structure to add more specific sugars. It hasrecently been shown that a specific enzyme in the intestine (alkaline phosphatase) candetoxify LPS by removing the two phosphate groups found on LPS carbohydrates.[11]This may function as an adaptive mechanism to help the host manage potentially toxiceffects of gram-negative bacteria normally found in the small intestine.Biosynthesis and transportLPS Final Assembly:O-antigen subunits are translocatedacross the inner membrane (by Wzx) where they arepolymerized (by Wzy, chain length determined by Wzz) and ligated (by WaaL) on to complete Core-Lipid A molecules (which were translocated by MsbA).[12]LPS Transport: Completed LPS molecules are transported across the periplasm and outer membrane by the proteins LptA, B, C, D, E, F, and G[13]Biological effects on hosts infected withgram-negative bacteriaImmune responseLPS acts as the prototypical(典型的)endotoxin because it binds theCD14/TLR4/MD2receptor complex in many cell types, but especially in monocytes, dendritic cells, macrophages and B cells, which promotes the secretion of pro-inflammatory cytokines, nitric oxide, and eicosanoids(类花生酸).[14]LPS is also an exogenous pyrogen (external fever-inducing substance,外源性致热源)Being of crucial importance to Gram-negative bacteria, these molecules make candidate targets for new antimicrobial(抗菌的)agents.Some researchers doubt reports of generalized toxic effects attributed to(归因于)all lipopolysaccharides, in particular, for cyanobacteria(蓝藻菌).[15]LPS function has been under experimental research for several years due to its role in activating many transcription factors. LPS also produces many types of mediators(介质)involved in septic shock(感染性休克). Humans are much more sensitive to LPS than other animals (e.g., mice). A dose of 1 µg/kg induces shock in humans,but mice will tolerate a dose up to a thousand times higher.[16]This may relate to differences in the level of circulating natural antibodies between the two species.[17][18] Said (上述)et al. showed that LPS causes an IL-10-dependent inhibition of CD4 T-cell expansion and function by up-regulating PD-1(progressed cell death-1,a kind of protein in human)levels on monocytes which leads to IL-10 production by monocytes after binding of PD-1 by PD-L.[19]Endotoxins are in large part responsible for the dramatic clinical manifestations of infections with pathogenic Gram-negative bacteria, such as Neisseria meningitidis,the pathogens that causes meningococcal disease, including meningococcemia, Waterhouse-Friderichsen syndrome, and meningitis.Bruce Beutler was awarded a portion of the 2011 Nobel Prize in Physiology or Medicine for his work demonstrating that TLR4 is the LPS receptor.[20][21]Portions of the LOS from several bacterial strains have been shown to be chemically similar to human host cell surface molecules; the ability of some bacteria to present molecules on their surface which are chemically identical or similar to the surface molecules of some types of host cells is termed molecular mimicry .[22] For example, in Neisseria meningitidis L2,3,5,7,9, the terminal tetrasaccharide portion of the oligosaccharide (lacto-N-neotetraose) is the same tetrasaccharide as that found in paragloboside, a precursor for ABH glycolipid antigens found on human erythrocytes .[9] In another example, the terminal trisaccharide portion (lactotriaose) of the oligosaccharide from pathogenic Neisseria spp. LOS is also found inlactoneoseries glycosphingolipids from human cells.[9] Most meningococci from groups B and C, as well as gonococci , have been shown to have this trisaccharide as part of their LOS structure.[9] The presence of these human cell surface ‘mimics’ may, in addition to acting as a ‘camouflage’ from the immune system, play a role in the abolishment of immune tolerance when infecting hosts with certain human leukocyte antigen (HLA) genotypes, such as HLA-B35.[9]Effect of variability (变异性) on immune responseO-antigens (the outercarbohydrates) are the mostvariable portion of theLPS molecule , impartingthe antigenic specificity . Incontrast, lipid A is the mostconserved part . However,lipid A composition alsomay vary (变异) (e.g., innumber and nature of acyl酰基chains even within orbetween genera 属). Some ofthese variations may impartantagonistic(对立的) properties to these LPS. Forexample Rhodobacter sphaeroides diphosphoryl lipid A (RsDPLA) is a potent antagonist of LPS in human cells, but is an agonist in hamster and equine cells. Toll-like receptors of the innate immune system recognizeLPS and trigger an immune response .It has been speculated that conical Lipid A (e.g., from E. coli) are more agonistic, less conical lipid A like those of Porphyromonas gingivalis may activate a different signal (TLR2 instead of TLR4), and completely cylindrical lipid A like that of Rhodobacter sphaeroides is antagonistic to TLRs.[23][24]LPS gene clusters are highly variable between different strains, subspecies, species of bacterial pathogens of plants and animals.[25][26]Normal human blood serum contains anti-LOS antibodies that are bactericidal and patients that have infections caused by serotypically distinct strains possess anti-LOS antibodies that differ in their specificity compared with normal serum.[27] These differences in humoral immune response to different LOS types can be attributed to the structure of the LOS molecule, primarily within the structure of the oligosaccharide portion of the LOS molecule.[27] In Neisseria gonorrhoeae it has been demonstrated that the antigenicity of LOS molecules can change during an infection due to the ability of these bacteria to synthesize more than one type of LOS,[27] a characteristic known as phase variation. Additionally, Neisseria gonorrhoeae, as well as Neisseria meningitidis and Haemophilus influenzae,[9] are capable of further modifying their LOS in vitro, for example through sialylation (modification with sialic acid residues), and as a result are able to increase their resistance to complement-mediated killing [27] or even down-regulate complement activation[9] or evade the effects of bactericidal antibodies.[9] Sialylation may also contribute to hindered neutrophil attachment and phagocytosis by immune system cells as well as a reduced oxidative burst.[9]Haemophilus somnus, a pathogen of cattle, has also been shown to display LOS phase variation, a characteristic which may help in the evasion of bovine host immune defenses.[28] Taken together, these observations suggest that variations in bacterial surface molecules such as LOS can help the pathogen evade both the humoral (antibody and complement-mediated) and the cell-mediated (killing by neutrophils, for example) host immune defenses.Health effectsEndotoxemia(内毒素血症)The presence of endotoxins in the blood is called endotoxemia. It can lead to septic shock, if the immune response is severely pronounced.[29]Moreover, endotoxemia of intestinal origin, especially, at the host-pathogen interface, is considered to be an important factor in the development ofalcoholic hepatitis,[30] which is likely to develop on the basis of the small bowel bacterial overgrowth syndrome and an increased intestinal permeability.[31] Lipid A may cause uncontrolled activation of mammalian immune systems with production of inflammatory mediators that may lead to septic shock.[10] This inflammatory reaction is mediated by Toll-like receptor 4 which is responsible for immune system cell activation.[10] Damage to the endothelial layer of blood vessels caused by these inflammatory mediators can lead to capillary leak syndrome, dilation of blood vessels and a decrease in cardiac function and can lead to septic shock.[32] Pronounced complement activation can also be observed later in the course as the bacteria multiply in the blood.[32] High bacterial proliferation triggering destructive endothelial damage can also lead to disseminated intravascular coagulation (DIC) with loss of function of certain internal organs such as the kidneys, adrenal glands and lungs due to compromised blood supply. The skin can show the effects of vascular damage often coupled with depletion of coagulation factors in the form of petechiae, purpura and ecchymoses. The limbs can also be affected, sometimes with devastating consequences such as the development of gangrene, requiring subsequent amputation.[32] Loss of function of the adrenal glands can cause adrenal insufficiency and additional hemorrhage into the adrenals causes Waterhouse-Friderichsen syndrome, both of which can be life threatening. It has also been reported that gonococcal LOS can cause damage to human fallopian tubes.[27]Auto-immune diseaseThe molecular mimicry of some LOS molecules is thought to cause autoimmune-based host responses, such as flareups of multiple sclerosis.[9][22] Other examples of bacterial mimicry of host structures via LOS are found with the bacteria Helicobacter pylori and Campylobacter jejuni, organisms which cause gastrointestinal disease in humans, and Haemophilus ducreyi which causes chancroid. Certain C. jejuni LPS serotypes (attributed to certain tetra- and pentasaccharide moieties of the core oligosaccharide) have also been implicated with Guillain-Barré syndrome and a variant of Guillain-Barré called Miller-Fisher syndrome.[9]Link to obesityEpidemiological studies have previously shown that increased endotoxin load, which can be a result of increased populations of endotoxin producing bacteria in the intestinal tract, is associated with certain obesity-related patient groups.[33][34][35] Other studies have shown that purified endotoxin from Escherichia coli can induce obesity and insulin-resistance phenotypes when injected into germ-free mouse models.[36] A more recent study has uncovered a potentially contributing role for Enterobacter cloacae B29 toward obesity and insulin resistance in a human patient.[37] The presumed mechanism for the association of endotoxin with obesity is that endotoxin induces aninflammation-mediated pathway accounting for the observed obesity and insulin resistance.[36]Bacterial genera associated with endotoxin-related obesity effects: Escherichia, EnterobacterLaboratory research and biotechnology production systemsLipopolysaccharides are frequent contaminants in plasmid DNA prepared from bacteria or proteins expressed from bacteria, and must be removed from the DNA or protein to avoid contaminating experiments and to avoid toxicity of products manufactured using industrial fermentation.Also, ovalbumin is frequently contaminated with endotoxins. Ovalbumin is one of the extensively studied proteins in animal models and also an established model allergen for airway hyper-responsiveness (AHR). Commercially available ovalbumin that is contaminated with LPS can fully activate endothelial cells in an in-vitro assay of the first step of inflammation, and it falsifies research results, as it does not accurately reflect the effect of sole protein antigen on animal physiology.In pharmaceutical production, it is necessary to remove all traces of endotoxin from drug product containers, as even small amounts of endotoxin will cause illness in humans. A depyrogenation oven is used for this purpose. Temperatures in excess of 300°C are required to break down this substance. A defined endotoxin reduction rate is a correlation between time and temperature. Based on primary packaging material as syringes or vials, a glass temperature of 250°C and a holding time of 30 minutes is typical to achieve a reduction of endotoxin levels by a factor of 1000.The standard assay for detecting presence of endotoxin is the Limulus Amebocyte Lysate (LAL) assay, utilizing blood from the Horseshoe crab.[38] Very low levels of LPS can cause coagulation of the limulus lysate due to a powerful amplification through an enzymatic cascade. However, due to the dwindling population of horseshoe crabs, and the fact that there are factors that interfere with the LAL assay, efforts have been made to develop alternative assays, with the most promising ones being ELISA tests using a recombinant version of a protein in the LAL assay, Factor C.[39]。
姜黄素防治细菌脓毒症小鼠的实验研究
![姜黄素防治细菌脓毒症小鼠的实验研究](https://img.taocdn.com/s3/m/d3ecc9cb7375a417876f8fc5.png)
姜黄素防治细菌脓毒症小鼠的实验研究【摘要】目的探讨姜黄素对大肠埃希菌引发的细菌脓毒症小鼠的爱惜作用。
方式昆明种小鼠90只随机分为对照组、模型组和姜黄素组,每组30只。
姜黄素组腹腔注射姜黄素固体分散剂,天天1次,持续3 d。
第4天,模型组和姜黄素组尾静脉注射041184大肠埃希菌标准菌悬液成立细菌脓毒症模型。
8 h后,每组处死小鼠20只检测血清天门冬氨酸氨基转移酶(AST)、丙氨酸氨基转移酶(ALT)、肿瘤坏死因子α(TNFα)和一氧化氮(NO)含量,观看肝、肾、肺和肠等脏器病理改变,余下小鼠观看24 h生存率。
结果姜黄素组血清AST、ALT、TNF α和NO含量明显低于模型组(P<,病理检查见脏器损害亦明显减轻;24 h存活率90%,明显高于模型组(50%)。
结论姜黄素可减少细菌脓毒症小鼠TNFα、NO和氧自由基的产生,减轻其脏器病理损害。
【关键词】姜黄素休克脓毒大肠杆菌疾病模型动物ABSTRACT: Objective To investigate the effects of curcumin(Cur) on bacterial sepsis induced by Escherichia coli in mice. Methods Sixty mice were divided into 3 groups at random: a control group, an infected group and a curcumin group. Cur group: intraabdominal injection of curcumin solid dispersant(200 mg/kg) was given for three days. The infected group: 078 (5 1010cfu/L, 10 mL/kg) was administrated byintravenous injection to create the bacterial sepsis mice model.8 hours after the injection, the level of AST, ALT, and TNF α and NO of each group was measured and mice were sacrified to observe the pathological change of liver, ridney, lung and intestines. Results Curcumin can remarkably reduce the level of the AST and ALT in the serum of the bacterial sepsis mice induced by (P< as well as that of TNFα(P< and NO(P<. Meanwhile, it can significantly decrease the pathological change of liver. Conclusion Curcumin can play an important role to protect the mice from bacterial sepsis by ways of cytotoxic effects of NO, TNFα and oxygen free radicals.KEY WORDS: curcumin; shock,septic; Escherichia coli; disease models,animal福建医科大学学报 2020年7月第42卷第4期侯君艺等:姜黄素防治细菌脓毒症小鼠的实验研究脓毒症是由严峻细菌感染引发的全身炎症反映综合征的晚期失代偿表现,是严峻烧伤、创伤和感染的常见并发症,进一步进展可致使脓毒症性休克、多器官功能障碍综合症(multiple organ dysfuction syndrome, MODS)。
毒理学基础英文词汇
![毒理学基础英文词汇](https://img.taocdn.com/s3/m/56f3a185e53a580216fcfef6.png)
毒理学基础英文词汇第一章绪论Toxicology(毒理学)environmental toxicology(环境毒理学)clinical toxicology(临床毒理学)forensic toxicology(法医毒理学)regulatory toxicology(管理毒理学)ecotoxicology(生态毒理学)in vivo(体内试验)in vitro(体外试验)in situ(原位)toxicogenomics(毒物基因组学)第二章毒理学基本概念xenobiotic(外源化学物)toxicant或poison(毒物)toxin(毒素)toxicity(毒性)toxic effect(毒作用/毒效应)adverse effect (损害作用)target organ(靶器官)spectrum of toxic effects(毒效应谱)biomarker(生物学标志)graded response(量反应)quantal response(质反应)graded dose-response relationship(剂量-量反应关系/剂量-效应关系)quantal dose-response relationship(剂量-质反应关系/剂量-反应关系)median lethal dose(半数致死剂量,LD50)absolute lethal dose(绝对致死剂量,LD100)minimal lethal dose(最小致死剂量,LD01)median tolerance limit(半数耐受限量)threshold dose(阈剂量)lowest observed adverse effect level (观察到损害作用的最低剂量,LOAEL ) no-observed adverse effect level (未观察到损害作用剂量,NOAEL ),toxic effect zone(毒作用带)acceptable daily intake(每日容许摄入量,ADI)maximum allowable concentration(最高容许浓度)reference dose(参考剂量)第三章外源化学物在体内的生物转运和生物转化biotransportation(生物转运)absorption(吸收)distribution(分布)accumulation(蓄积)excretion(排泄)elimination(消除)toxicodynamics(毒物效应动力学)toxicokinetics(毒物代谢动力学)conjugation(结合)blood-brain barrier(血脑屏障,BBB)enterohepatic circulation(肠肝循环)first pass effect(首过效应)compartment model(房室模型)area under curve(曲线下面积。
英文原版 卡萨特和杜勒斯毒理学第九版和第八版的区别
![英文原版 卡萨特和杜勒斯毒理学第九版和第八版的区别](https://img.taocdn.com/s3/m/7a166b6e7275a417866fb84ae45c3b3567ecdd35.png)
英文原版卡萨特和杜勒斯毒理学第九版和第八版的区别The Difference between the Ninth and Eighth Editions of Casarett and Doull's ToxicologyThe Casarett and Doull's Toxicology textbook is a renowned reference in the field of toxicology, providing comprehensive information on the adverse effects of chemical substances on living organisms. The ninth and eighth editions of this textbook have certain differences that reflect updates and modifications in the field. Here are some key distinctions between the two editions: 1. New Chapters: The ninth edition includes new chapters that were not present in the eighth edition. These chapters cover emerging topics or areas of increasing importance in toxicology. Examples of such chapters in the ninth edition include "Toxicology of Nanoparticles" and "Toxicology of Hormone Disruptors."2. Updated Content: Toxicology is a rapidly evolving field, with new research and discoveries continuously shaping our understanding of chemical toxicity. The ninth edition incorporates the latest advancements and scientific findings in toxicology, ensuring that the content remains up to date. This includes updated information on toxicokinetics, mechanisms of toxicity, and risk assessment.3. Expanded Coverage: Certain topics receive expanded coverage in the ninth edition compared to the eighth edition. These expansions reflect the growing significance of these areas in modern toxicology. For instance, additional information may be provided on the toxicology of specific drug classes like opioids oron the toxic effects of environmental pollutants.4. Revised Organizational Structure: The organization of chapters and sections may be revised in the ninth edition to provide a more logical and coherent flow of information. This ensures easy access to relevant topics and helps readers navigate through complex subject matter efficiently.5. Enhanced Pedagogical Features: The ninth edition may include additional pedagogical features compared to the eighth edition. These features aim to facilitate learning and understanding of the subject matter. Examples of such enhancements may include case studies, visual aids, and self-assessment questions or exercises.It is important to note that the specific differences between the ninth and eighth editions may vary, and it is advisable to compare the tables of content or refer to the publishers' or authors' official statements for a more detailed and accurate understanding of the changes made in each edition.。
Biomarkers of sepsis
![Biomarkers of sepsis](https://img.taocdn.com/s3/m/1616ad1a52ea551810a68718.png)
脓毒症生物标记物摘要脓毒症是一个对于普通感染的异常系统性反应,它可能呈现的是免疫系统对于损伤的反应形式。
当出现多器官功能障碍和病人易收到院内感染的影响时,一个高炎症反应伴随免疫抑制状态。
用生物标志物诊断脓毒症,有利于早期对其进行干预,从而减少死亡的风险。
虽然乳酸当前被认为是公认的用于识别脓毒症的标志物,其他标志物可能有助于增强乳酸用于识别脓毒症的效力。
这些包括脓毒症高炎症反应阶段的标志物,例如:促炎症细胞因子和炎症趋化因子。
对于感染和炎症反应综合产物的蛋白例如C反应蛋白,降钙素。
中性粒细胞和单核细胞活化的生物标志物。
最近,脓毒症免疫抑制阶段的生物标志物,例如:抗炎反应因子,单核细胞和淋巴细胞的细胞表面生物标志物,被检测。
抗炎和促炎标志物的联合作为多重标志物组合可能有助于在多器官功能障碍发生之前识别那些有可能发展为严重脓毒症的患者。
尽管严重脓毒症病人死亡率仍很高,但是生物标志物联合在免疫抑制阶段进行靶向治疗的新方法,有助于减少严重脓毒症相关的死亡率。
英文缩写:APACHE: acute physiology and chronic health evaluation; CARS: compensatory anti-inflammatory response syndrome; CLP: cecal ligation and puncture; CRP: C-reactiveprotein; CTLA-4: cytotoxic T lymphocyte-associated antigen-4; DAMP: damage-associatedmolecular pattern; DIC: disseminated intravascular coagulation; ED: Emergency Department;FDA: US Food and Drug Administration; HBP: heparin-binding protein, azurocidin; HLA: humanleukocyte antigen; HMGB1: high-mobility group box 1; IL: interleukin; IL-1ra: antagonist of theinterleukin-1 receptor; LBP: lipopolysaccharide-binding protein; LPS: lipopolysaccharide;MCP-1: monocyte chemoattractant protein-1; MHC: major histocompatibility complex;MODS: multiple organ dysfunction syndrome; MRP: myeloid related protein; NAD:nicotinamide adenine dinucleotide; NGAL: neutrophil gelatinase-associated lipocalin;PAMP: pathogen-associated molecular pattern; PASS: Procalcitonin and Survival Study; PCR:polymerase chain reaction; PCT: procalcitonin; PD-1: programmed death-1; PMN: polymorphonuclear leukocyte; PTX3: pentraxin 3; RAGE: receptor for advanced glycationend-products;RNA: ribonucleic acid; SIRS: systemic inflammatory response syndrome; SOFA: SequentialOrgan Failure Assessment; TGF: transforming growth factor; TLR:toll-like receptors; TNF: tumornecrosis factor; TREM: triggering receptor expressed on myeloid cells介绍脓毒症是一个不同于普通感染的异常的全身性反应。
理工类专业课复习资料-毒理学基础-名解、大体部分重点总结
![理工类专业课复习资料-毒理学基础-名解、大体部分重点总结](https://img.taocdn.com/s3/m/52e0af1bbc64783e0912a21614791711cc797925.png)
名解、大体部分重点总结1.毒理学(toxicology):的传统定义是研究外源化学物对生物体损害作用的学科,现代毒理学已发展为所有外源因素对生物系统的损害作用,生物学机制,安全性评价与危险性分析的学科。
2.最大耐受剂量(maximal tolerance dose):指化学物质不引起受试对象出现死亡的最高剂量3.自由基(free radical):是独立游离存在的带有不成对电子的分子、原子和离子,它主要由化合物的共价键发生均裂而产生。
4.易感生物学标志(biomarker of susceptibility):是关于个体对外源化学物的生物易感性的指标即反应机体先天具有或后天获得的对暴露外源物质产生反应能力的指标。
5.半减期(half life):外源化学物的血浆浓度下降一半所需要的时间,它是衡量机体消除化学物能力的一个重要参数。
6.癌基因(Oncogene):一类在自然或试验条件下,具有诱发恶性转化的潜在基因。
7.急性毒性(acute toxicity):是指机体(实验动物或人)一次或24小时内接触多次一定剂量外源化合物后在短期内所产生的毒作用及死亡。
包括一般行为、大体形态变化及死亡效应。
8.基准剂量BMD\benchmark dose:是依据动物试验剂量-反应关系的结果,用一定的统计学模式求得的引起一定比例动物出现阳性反应剂量的95%可信限区间的下限值。
9.生物转化(Biotransformation):又称代谢转化,指外源化学物在体内经历酶促反应或非酶促反应而形成的代谢产物的过程。
10.代谢酶遗传多态性:不同种属,不同个体内的同一种代谢酶的基因编码不同,从而导致了其功能的不同,这就是代谢酶遗传多态性11.危险度(risk):又称危险或危险性,指在特定条件下,因接触某种水平的化学毒物而造成机体损伤、发生疾病,甚至死亡的预期概率。
12.细胞凋亡(apoptosis):是指细胞在一定的生理或病理条件下,受内在遗传机制的控制自动结束生命的过程,是一种自然的生理过程。
OECD 433 Acute Inhalation Toxicity固定浓度法
![OECD 433 Acute Inhalation Toxicity固定浓度法](https://img.taocdn.com/s3/m/54270088daef5ef7ba0d3cea.png)
OECD/OCDE 433Adopted:9 October 2017OECD GUIDELINE FOR TESTING OF CHEMICALSAcute Inhalation Toxicity: Fixed Concentration ProcedureINTRODUCTION1. OECD Guidelines for the Testing of Chemicals are periodically reviewed in the light of scientific progress and animal welfare considerations. The original acute inhalation Guideline 403 was adopted in 1981. Development of an Inhalation Fixed Concentration Procedure (FCP) was considered appropriate, following adoption of the revised oral Fixed Dose Procedure (FDP), OECD Guideline 420 in December 2001 and the deletion of the oral toxicity test, OECD Guideline 401. This FCP guideline will allow the use of a series of fixed concentrations for the determination of acute inhalation toxicity in only one sex.2. Traditional methods for assessing acute toxicity use death/moribundity of animals as the sole endpoint. In 1984, a new approach to acute toxicity testing was suggested by the British Toxicology Society based on the administration of test chemical at a series of fixed concentration levels [1]. This approach avoided using death/ moribundity of animals as either an exclusive or an intended endpoint by incorporating evident clinical signs of toxicity at one of a series of fixed dose levels, as an endpoint on which to base classification of the test chemical. This approach is also taken for this guideline. In agreement with the OECD Guidance Document on Humane Endpoints [2] and definitions contained therein refinements are introduced in order to minimize any suffering and distress by the animals and, to the extent feasible, reduce the number of animals used. Evident toxicity is a general term describing clear signs of toxicity following the administration of a test chemical, such that at the next highest fixed concentration either severe pain and enduring signs of severe distress, moribund condition or probable mortality in most animals can be expected. Guidance on the recognition of evident toxicity has been provided by Sewell et al (2015) [3]: Evident toxicity has been reached if one or more animals display any one of the listed signs (from the day after exposure onwards): tremors, hypoactivity, irregular respiration or bodyweight loss (>10% pre-study value). Should there be lack of clarity regarding the evident toxicity, it can be concluded that the toxicity is not evident, and that further testing should be considered. The statistical properties of the FCP have been evaluated using mathematical modelling [4-6].3. Guidance on the conduct and interpretation of acute inhalation studies can be found in the Guidance Document No. 39 on Acute Inhalation Toxicity Testing [7].1© OECD, (2017)You are free to use this material subject to the terms and conditions available at /termsandconditions/.This Guideline was adopted by the OECD Council by written procedure on 9 October 2017 [C(2017)97].433 OECD/OCDE4. Definitions used in the context of this Guideline can be found in Guidance Document No. 39 on Acute Inhalation Toxicity Testing [7].5. The method provides information on the hazardous properties and allows the test chemical to be ranked and classified according to the United Nations (UN) Globally Harmonized System of Classification and Labelling of Chemicals (GHS) for the classification of chemicals which cause acute toxicity [8].INITIAL CONSIDERATIONS6. All available information on the test chemical should be considered by the testing facility prior to conducting the study. Such information will include the identity and chemical structure of the test chemical; its physico-chemical properties; the results of any other in vitro or in vivo toxicity tests on the test chemical; available (Q)SAR data and toxicological data on structurally related substances; the anticipated use(s) of the test chemical and the potential for human exposure. Some of this information will assist in the selection of an appropriate starting concentration (e..g. through read-across from toxicity of structurally-related chemicals /those of same chemical class, and/or data from predictive software), or will allow further testing to be avoided if available information is sufficient. Before use of the Test Guideline on a mixture for generating data for an intended regulatory purpose, it should be considered whether, and if so why, it may provide adequate results for that purpose. Such considerations are not needed, when there is a regulatory requirement for testing of the mixture.PRINCIPLE OF THE TEST7. It is a principle of the method that only moderately toxic concentrations are used so that ‘eviden t toxicity’ (described in more detail below), rather than death/moribundity is used as an endpoint, and concentrations that are expected to be lethal are avoided. Also, concentrations that are expected to cause marked pain and distress, due to corrosive1or severely irritant actions, should not be administered. When testing an irritating or corrosive chemical refer to GD39 [7] for guidance. Moribund animals, or animals obviously in pain or showing signs of severe and enduring distress shall be humanely killed, and are considered in the interpretation of the test results in the same way as animals that died on test. Criteria for making the decision to kill moribund or severely suffering animals, and guidance on the recognition of predictable or impending death, are the subject of a separate OECD Guidance Document [2].8. Groups of animals of a single sex are exposed for a short period of time to the test chemical in a stepwise procedure using the appropriate fixed concentrations for vapours, dusts/mists (aerosols) or gases as set out in Annex 1. The initial concentration level is selected on the basis of existing information or a sighting study at the concentration expected to produce evident toxicity, clear signs of toxicity without causing severe toxic effects or mortality, that predict exposure to the next highest concentration will cause severe toxicity or death/moribundity in most animals. Analysis by Stallard et al.,(2011) provides the rationale for the sighting study [6]. Guidance on the recognition of evident toxicity is provided in section 40 and has been described by Sewell et al (2015) [3]. It is important to note that ‘evident toxicity’ occurs prior to the clinical signs and conditions associated with pain, suffering, and impending death, that are described in the OECD Guidance Document on humane end-points [2]. This document describes signs and conditions in animals in which evident toxicity has already been exceeded. Further groups of animals 1 Determined using a validated test methods (e.g., TG430 or 431) or an acceptable prediction.2© OECD, (2017)OECD/OCDE 433 may be tested at higher concentrations in the absence of signs of evident toxicity or mortality at lower concentrations.9. This procedure continues until the concentration causing evident toxicity or no more than one death/ moribund animal is identified, or when no effects are seen at the highest concentration or when deaths/ moribundity occur at the lowest concentration. Depending on the outcome of the test (i.e. evident toxicity or mortality/moribundity), testing at one concentration level may be sufficient to allow judgement on the acute toxicity of the test chemical. Evident toxicity is defined as clear signs of toxicity following the administration of a test chemical, such that at the next highest fixed concentration either severe pain and enduring signs of severe distress, moribund condition or probable mortality in most animals can be expected’. Analysis by Sewell et al. (2015) [3] has shown that evident toxicity has been reached if one or more animals display any one of the listed signs (from the day after exposure onwards): tremors, hypoactivity, irregular respiration or bodyweight loss (>10% pre-study value). However, the analysis also includes information on other more rarely observed signs that also have high predictivity. This guidance should be used in conjunction with study director experience and judgement. For further guidance please see GD39 [7].DESCRIPTION OF THE METHODSelection of animal species10. The preferred rodent species is the rat, although on occasion other rodent species may be used. Justification should be provided for the use of other rodent or non-rodent species. Existing information (if available) or a sighting study using one male and one female animal may be performed to select the most sensitive sex for use in the main study [6]. This sighting study is not compulsory. The main purpose of the sighting study is to select an appropriate starting concentration for the main study but it can also inform the choice of sex. Males should be used as a default if there is no apparent difference in sensitivity, and females should only be used if they appear to be more sensitive than males. However, available knowledge of the toxicological or toxicokinetic properties of structurally related chemicals should also be taken in to account and adequate justification of sex selection should be provided.11. Healthy young adult animals of commonly used laboratory strains should be employed. If females are used they should be nulliparous and non-pregnant. Each animal, at the commencement of testing, should be between 8 and 12 weeks old and its weight should fall within an interval of ±20% of the mean body weight of any previously exposed animals.Housing and feeding conditions12. The temperature of the experimental animal room should be 22±3ºC. Although the relative humidity should be at least 30% and preferably not exceed 70% other than during room cleaning the aim should be 45-65%. Lighting should be artificial, the sequence being 12 hours light, 12 hours dark. For feeding, conventional laboratory diets may be used with an unlimited supply of drinking water. Feed should be provided ad libitum where possible. Animals should be group-caged by concentration, but the number of animals per cage should not interfere with clear observations of each animal.3© OECD, (2017)433 OECD/OCDEPreparation of animals13. The animals are acclimatised to the laboratory conditions for at least five days prior to the start of exposure. Animals are randomly selected for use in the study and marked to provide individual identification.Mode of exposure14. Animals are exposed to the test chemical as a gas, vapour, aerosol, or a mixture thereof. The physical state to be tested depends on the physico-chemical properties of the test chemical, the selected concentration, and/or the physical form most likely present during the handling and use of the test chemical. Both head/nose-only and whole-body exposure techniques may be used. The head/nose-only exposure method minimises exposure or uptake by non-inhalation routes and allows testing of individual animals at high concentrations, as required for limit tests, without the need for large quantities of material. Further advantages include; ease of maintenance of a homogenous test atmosphere, less potential for test chemical instability (e.g., reaction with excreta or humidity), and faster equilibration of the chamber atmosphere due to the smaller volume required. The head/nose-only technique does, however, require restraint of the animals throughout the exposure period, which is not necessary for whole-body exposures, and therefore causes more stress than whole-body technique. However, it easier to remove a distressed animal from a nose-only chamber than whole-body chamber. The selected exposure model should be designed to minimise any pain, distress or suffering experienced by the animals, consistent with the scientific objective of the study [2].Head/nose-only exposure technique15. During exposure, the animals are exposed to the test chemical in exposure tubes. The animal restraining tubes should not impose undue stress on the animal, should be constructed in such a way as to avoid hyperthermic stress for the animal and should make it impossible for the animal to avoid inhalation exposure. However, if a negative balance of air volumes supplied and extracted cannot be avoided, a dilution of test atmosphere by bias-airflow (via exposure tubes) should be prevented. The inhalation chamber should be operated in well ventilated chemical hoods. The animals should be tested with inhalation equipment designed to sustain a dynamic air flow which exceeds at least twice the respiration ventilation volume of all animals in the inhalation device. An adequate oxygen content of at least 19% and a carbon dioxide concentration not exceeding 1%, with similar exposure conditions at each exposure port should be ensured. During the sampling of the test atmosphere, a significant disturbance of the airflow dynamics should be avoided. The rate of air flow should be adjusted to ensure that conditions throughout the equipment are essentially the same. The principles of the nose-only exposure technique and its particular advantages and disadvantages are described in GD 39 [7].Whole-body exposure technique16. The animals should be tested using inhalation equipment designed to sustain a dynamic air flow of approximately 12 to 15 air changes per hour. Other air flow rates may be useful to meet specific requirements imposed by the test chemical. However, an adequate oxygen content of at least 19%, a carbon dioxide concentration not exceeding 1%, and an evenly distributed exposure atmosphere should be ensured. As a general rule to ensure the stability of a chamber atmosphere, the total volume of the test animals should not exceed five per cent of the volume of the test chamber. The principles of the whole-body exposure technique and its particular advantages and disadvantages are described in GD 39 [7].4© OECD, (2017)OECD/OCDE 433 Exposure conditions17. A fixed duration of exposure of four hours, excluding equilibration time, is recommended. Other durations may be needed to meet specific requirements.18. To establish suitable exposure concentrations, a technical trial test without animals is mandatory. It is technically difficult to generate test atmospheres to accurately meet specified fixed exposure concentrations. Therefore, to prevent unnecessary repeat testing, individual chamber concentration samples should not deviate from the mean chamber concentration by more than ±10% for gases and vapours, andby no more than ±20% for liquid or solid aerosols.In the case of potentially explosive test chemicals, care should be taken to avoid favourable conditions for explosions. Further guidance can be found in GD39 [7]. Particle size19. As it is difficult to predict the most responsive region of the respiratory tract or the most harmful particle size, the particle size distribution of dusts and aerosols should be such that exposure of all regions of the tract can be achieved. An aerosol with a mass median aerodynamic diameter (MMAD) ≤4 µ m and a geometric standard deviation (GSD) in the range of 1.5 to 3.0 is recommended to ensure that comprehensive respiratory tract exposure occurs [10]. In case a laboratory deviates from the recommended MMAD, an explanation and justification should be given. For example, metal fumes may be smaller than this standard, and charged particles, fibres, and hygroscopic materials (which increase in size in the moist environment of the respiratory tract) may exceed this standard.Generation of test atmospheres20. Where necessary, a suitable vehicle may be added to the test chemical to help generate an appropriate concentration and respirability of the test chemical in the atmosphere. Where a vehicle is used to help generate an appropriate concentration of the substance in the atmosphere, the acute inhalation toxicity of the vehicle should be known. A concurrent vehicle (or any other) control group is not considered necessary for well characterised control vehicles. Particulate material may be subjected to mechanical processes to achieve the required particle size distribution, however, care should be taken not to decompose or alter the test chemical, which should be analytically verified, as rigorous mechanical processes could change particle size distribution from that of pristine materials. Adequate care should be taken not to contaminate the test chemical. It is not necessary to test non-friable granular materials which are purposefully formulated to be un-inhalable. An attrition test should be used to demonstrate that respirable particles are not produced when the granular material is handled. If an attrition test produces respirable articles, an inhalation toxicity test should be performed.MONITORING OF EXPOSURE CONDITIONSChamber airflow21. The flow of air through the exposure chambers should be monitored continuously and recorded at least three times during each exposure.Chamber temperature22. The air temperature in the animal’s breathing zo ne should be monitored continuously and recorded at least three times during each exposure. Ideally the temperature should remain within the range5© OECD, (2017)433 OECD/OCDE22±3°C. Deviations from this range should be commented upon with an assessment of the effect, if any, on the outcome of the exposure.Relative humidity23. The relative humidity in the animal’s breathing zone, for both the head/nose only and the whole body exposures, should be monitored continuously and recorded three times during each exposure where possible. The RH should ideally be maintained in the range of 30 to 70% but it is recognised that under certain circumstances this may either be unattainable (e.g., when testing water based formulations) or may not be measurable due to interference by the test chemical with the test method.Concentration of test chemical24. Actual concentrations of the test chemical should be measured in the breathing zone of the rats in both the head/nose only and the whole body exposures. During the exposure period, the actual concentrations of the test chemical shall be held as constant as practicable (see paragraph 17) and monitored continuously or intermittently depending on the method of analysis. If intermittent sampling is used at least five samples should be taken at approximately hourly intervals. If not feasible due to limited air flow rates or low concentrations, fewer samples may be collected, with a minimum of one sample may be collected over the entire exposure period. For single component solid aerosols and liquids that are of extremely low volatility, gravimetric analysis is acceptable. When performing gravimetric sampling at the higher exposure concentrations used in these studies, care should be taken to calibrate the flow meter (or dry gas meter) used to determine sampled volume as a function of the pressure drop across the filter (based upon the relationship pressure x volume = constant). A calibration volume curve should be generated for each flow meter or dry gas meter used.25. For aerosols of liquid formulations that can be evaporated to a constant weight, gravimetric analysis of the dried residue may be used. Appropriate extrapolation to calculate the weight of formulation should be applied to the gravimetric data. It is not necessary to analyse inert ingredients provided the mixture at the animal’s breathing zone is analogous to the formulation; the grounds for this conclus ion should be provided in the study report. If there is some difficulty in measuring chamber analytical concentration due to precipitation, non-homogenous mixtures, volatile components, or other factors, additional analyses of inert components may be necessary.26. Where gravimetric analysis is unsuitable and the test atmosphere contains more than one component, chemical analysis of the major active ingredient followed by extrapolation to the concentration of formulation may be acceptable but should be justified.27. Whenever the test chemical is a formulation, the analytical concentration should be reported for the total formulation and not just for the active ingredient.Particle size distribution28. The particle size distribution of the test aerosol should be determined at least twice during each 4- hour exposure. A range of sampling devices is suitable but the device selected should allow calculation of the MMAD (See paragraph 18). Geometric Mean Diameter (GMD) may be determined by using a Scanning Mobility Particle Sizer (SMPS). In the case of multi-component aerosols the principles given above for determination of concentration should be applied. Adequate information should be available6© OECD, (2017)OECD/OCDE 433 within the testing facility to demonstrate that such samplers collect an atmospheric sample that is representative of the atmosphere to which the animals are exposed.Nominal concentration29. The nominal exposure chamber concentration should be determined by recording the amount of test chemical disseminated into the exposure chamber/tube during the generation period and dividing this by the total airflow through the chamber/tube during the same period.PROCEDURESighting study30. The purpose of the sighting study is to allow selection of the most sensitive sex and an appropriate starting concentration for the main study. However, a sighting study may not be warranted if prior information is available to guide the choice of a starting concentration and/or the most sensitive sex. Therefore, this study should not be considered compulsory. The test chemical is administered to two animals (one male and one female) simultaneously at a chosen starting concentration for a period of at least four hours. Testing continues in a sequential manner depending on the outcome following the flow charts in Annex 1 [6]. If both animals demonstrate the same response of death, non-fatal evident toxicity or no effects, the sighting study either stops and leads to a main study conducted in males or continues to test two animals (one male and one female) at the next concentration. If, at any concentration, a sex difference is indicated, the main study will be conducted using the sex that is shown to be the more sensitive, and the sighting study continues with that sex alone in such a way as to determine an appropriate main study starting concentration. The sighting study is completed when a decision on the starting concentration for the main study can be made, based on signs of evident toxicity or if a death/ moribundity is seen at the lowest fixed concentration. Males should be used for the main study if there is no apparent difference in sensitivity between sexes.31. The starting concentration for the sighting study is selected from the fixed concentration levels found in Annex 1 as a concentration expected to produce evident toxicity based, when possible, on evidence from existing data on the same chemical and/or structurally related chemicals. In the absence of such information, the starting concentration will be 10 mg/L, 1 mg/L or 2500 ppm for vapours, dusts/mists (aerosols) and gases, respectively.32. A period of at least 24 hours will be allowed between the testing of each pair of animals. All animals should normally be observed for at least one week.33. In cases where an animal tested at the lowest fixed concentration level in the sighting study dies or exhibits clear clinical signs of toxicity, the normal procedure is to terminate the study and assign the test chemical to GHS [8] Category 1 without proceeding to main study testing (as shown in Annex 1). However, if further confirmation of the classification is required (i.e. if death/moribundity of only one sex occurs), an optional supplementary procedure may be conducted, as follows. An additional animal of the most sensitive sex is tested at the lowest fixed concentration. If this animal dies, then GHS Category 1 will be confirmed and the study will be immediately terminated. If the animal survives, then a maximum of three additional animals will be tested at this concentration. Because there will be a high risk of mortality, these animals should be tested in a sequential manner to protect animal welfare. The time interval between exposure of each animal should be sufficient to establish that the previous animal is likely to survive. If an additional death/moribundity occurs, the testing sequence will be immediately terminated and no further7© OECD, (2017)433 OECD/OCDEanimals will be tested. The classification will be as shown in Annex 1: Category 1 if there are two or more deaths/moribundities (outcome A), or Category 2 if there is one death/moribundity (outcome B).MAIN STUDYNumbers of animals and concentration levels34. The action to be taken following testing at the starting concentration level is indicated by the flow charts in Annex 1. Depending on the outcome of each study, one of three actions will be required; either stop testing and assign the appropriate hazard classification class, test at a higher fixed concentration or test at a lower fixed concentration. However, a concentration level, which caused death/moribundity in the sighting study, will not be revisited in the main study, to protect animal from unnecessary suffering (see Annex 1). Experience has shown that the most likely outcome at the starting concentration level will be that the test chemical can be classified and no further testing will be necessary. When testing a descending series and 2-3 deaths are observed (within the scope of outcome A), then in the interests of animal welfare the test should be halted and the test chemical classified according to outcome C of the next concentration in the series. Guidance on the recognition of evident toxicity (within the scope of outcome B) is provided in section 40 and described by Sewell et al. (2015) [3]35. A total of five animals of one sex (the most sensitive sex as indicated in the sighting study, or males only) will normally be used for each concentration level investigated, in addition to the pairs of animals used in the sighting study.36. The time interval between exposures at each level is determined by the onset, duration and severity of toxic signs. Exposure of animals at the next concentration should be delayed until there is confidence in the survival of the previously tested animals. A period of three or four days between the exposures at each concentration level is recommended to allow for the observation of delayed toxicity. The time interval may be adjusted as appropriate, e.g., in case of inconclusive response.Limit Test37. The limit test is primarily used in situations where the study director has information indicating that the test chemical is likely to be non-toxic, i.e., having toxicity only above regulatory limit doses. Information about the toxicity of the test chemical can be gained from knowledge about similar tested compounds or similar tested mixtures or products, taking into consideration the identity and percentage of components known to be of toxicological significance. In those situations where there is little or no information about its toxicity, or in which the test chemical is expected to be toxic, the main test should be performed.38. Using the normal procedure, a main study starting concentration of 20 mg/l, 5mg/l or 20,000 ppm for vapours, dusts/mists (aerosols) and gases, respectively, followed by exposure of a further five animals at this level serves as a limit test for this guideline, if achievable. When testing aerosols, the primary goal should be to achieve a respirable particle size (i.e. an MMAD of ≤4 μm). This is possible with most test chemicals at a concentration of 2 mg/L. Aerosol testing at greater than 2 mg/L should only be attempted if a respirable particle size can be achieved. In some cases, as required by some regulatory authorities, testing up to the limit of GHS class 5 may be conducted. However, testing in animals in Category 5 ranges is discouraged and should only be considered when there is a strong likelihood that the results of such testing would have a direct relevance to the protection of human health [8].8© OECD, (2017)。
砒霜纳米乳治疗小鼠Lewis肺癌的效果初步评价.
![砒霜纳米乳治疗小鼠Lewis肺癌的效果初步评价.](https://img.taocdn.com/s3/m/645ed4c7d15abe23482f4df8.png)
砒霜纳米乳治疗小鼠Lewis肺癌的效果初步评价[ 09-09-07 16:12:00 ] 编辑:studa20作者:许严伟,杜钢军,林海红,宋紫辉,张硕,王梅【摘要】目的评价砒霜纳米乳治疗小鼠Lewis肺癌的效果及对免疫系统的毒性。
方法采用MTT法及流式细胞仪分别检测A549细胞的增殖和凋亡;采用小鼠Lewis肺癌皮下移植肿瘤模型观察砒霜的抗肿瘤作用,通过小鼠碳粒廓清和迟发性变态反应考察砒霜对免疫系统的毒性。
结果砒霜纳米乳可诱导A549细胞的凋亡,流式结果出现DNA亚二倍体凋亡峰;砒霜纳米乳与其水溶液连续灌胃给药8 d对小鼠Lewis肺癌皮下肿瘤的抑瘤率分别为42.0%和33.2%,砒霜水溶液对小鼠碳粒廓清能力及迟发性变态反应均有降低作用,但其纳米乳对两者均无明显影响。
结论砒霜纳米乳对小鼠Lewis肺癌皮下肿瘤的效果优于其等剂量水溶液,且能降低对机体免疫系统的损害。
【关键词】砒霜纳米乳 Lewis肺癌免疫Abstract:Objective To evaluate the therapeutic action of arsenic nano-emulsion on Lewis lung cancer and its toxicity toimmunity .MethodsThe proliferation and cell cycle of A549 cells were determined by MTT assay and flow cytometry respectively. The antitumor of arsenic was tested by tumor injected subcutaneously in mice and its toxicity to immunity was examined by clearance rate of charcoal particles and delayed type hypersensitivity.ResultsArsenic nano-emulsion and its water solution taken orally for eight days had respectively 42.0% and 33.2% inhibition on lewis lung cancer. The clearance rate of charcoal particles and delayed typehypersensitivity were depressed by arsenic water solution but not by arsenic nano-emulsion in mice. ConclusionArsenic nano-emulsion has better therapeutic action on Lewis lung cancer than arsenic water solution does at the same dose, and it also reduces toxicity to immunity.Key words:Arsenic nanoemulsion; Lewis lung cancer;Immunity肿瘤在人类死亡的主要疾病中居第2位,近年来发病率呈逐年上升趋势[1]。
地塞米松对脓毒症大鼠急性肾功能衰竭核因子 和炎症细胞因子的影响
![地塞米松对脓毒症大鼠急性肾功能衰竭核因子 和炎症细胞因子的影响](https://img.taocdn.com/s3/m/ae0d6b0aba1aa8114431d9da.png)
4 参考文献 [1] Gerecke A C, Giger W, Hartmann P C, et al. Anaerobic
degradation of brominated flame retardants in sewage sludge [J] . Chemosphere ,2006 ,64 (2 ) :311-317. [2] van der Ven L T, van de Kuil T, Leonards P E, et al . A 28-day oral dose toxicity study in Wistar rats enhanced to detect endocrine
实 用 医 学 杂 志 2009 年 第 25 卷 第 12 期
图 1 A 组 6 h 肾脏病理切片 (HE 染 色 × 1 000);图 2 B 组 6 h 肾脏病理切片 (HE 染色 × 1 000),箭 头 示 PMN 浸 润 ;图 3
C 组 6 h 肾 脏 病 理 切 片 (HE 染 色 41
effects of decabromodiphenyl ether (decaBDE) [J]. Toxicology Letters, 2008,179(1):6-14. [3] 丁淑瑾,王志新,陈敦金. 母源性十溴联苯醚胃灌后对出生子鼠 数 量 及 体 重 的 影 响 [J]. 实 用 医 学 杂 志 ,2008,24 (12):2056- 2057. [4] 陈 敦 金 ,余 琳 , 廖 秦 平 , 等 . 母 源 性 BDE-209 胃 灌 后 对 子 鼠 学 习 记 忆 能 力 的 影 响 以 及 血 清 BDE-209 浓 度 的 测 定 [J]. 中 华 围 产 医 学 杂 志 , 2006,9( 6):412- 415. [5] Tseng L H, Lee C W, Pan M H, et al. Postnatal exposure of the male mouseto2,2_,3,3_,4,4_,5,5_,6,6_-decabrominated diphenyl ether: Decreased epididymal sperm functions without alterations in DNA content and histology in testis[J]. Toxicology,2006,224(1-2): 33-43. [6] 周俊,陈敦金,廖秦平,等. 孕期、哺乳期暴露十溴联苯醚对子代 大 鼠 免 疫 功 能 的 影 响 [J]. 南 方 医 科 大 学 学 报 ,2006,26 ( 6): 738-741. [7] Gilchrist R B, Ritter L J, Armstrong D T. Oocyt-somatic cell interactions during follicle devel opment in mammals [J]. Anim Reprod Sci, 2004, 82-83: 431-446.
药物毒理学之名词解释
![药物毒理学之名词解释](https://img.taocdn.com/s3/m/68cffd82fad6195f312ba6dd.png)
毒理学(toxicology )是一门研究在特定条件下,外源物(化学、生物、物理)对生物体有害作用的综合性学科。
毒性(toxicity):药物在机体中可能产生的有毒作用暴露(exposure):机体以不同途径和方式对药物的接触。
靶部位(target site):药物对机体产生毒性作用并造成损害的部位靶组织(target tissue):药物对机体产生毒性作用并造成损害的组织靶器官(target organ):药物对机体产生毒性作用并造成损害的器官。
剂量(dose):机体暴露于药物的量(外剂量、内剂量)效应、反应(effect, response):机体暴露于药物后出现的生物学改变量反应(graded response ):毒性反应强弱呈连续增减的量变。
质反应(quantal response ):毒性反应只能用全或无、阴性或阳性表示剂量-反应关系(dose-response-relationship):药物作用于机体的剂量与所引起的生物学效应强度或发生率间的关系。
未观察到损害作用的剂量(No-Observed Adverse Effect Level,NOAEL):用最敏感方法未能检出外源物毒性效应的最大剂量最大耐受量(maximal tolerance dose, MTD):机体能耐受的最大剂量。
半数致死量(median lethal dose,LD 50 ):能引起半数实验动物死亡的浓度或剂量最小中毒量(minimum toxic dose ):诱发机体产生毒性效应的最低剂量最小致死剂量(minimal Lethal Dose,LD 01):引起实验动物出现死亡的最低剂量毒性反应(toxic reaction ):剂量过大或药物在体内蓄积过多时对机体的脏器或组织发生的危害性反应。
过敏反应(allergic reaction):非肽类药物作为半抗原与机体蛋白结合后,经过敏感化过程而发生的反应。
脓毒症定义和诊断标准的演进
![脓毒症定义和诊断标准的演进](https://img.taocdn.com/s3/m/e3fd62fc6394dd88d0d233d4b14e852458fb3917.png)
脓毒症定义和诊断标准的演进陈晓洁;董天皞;张桂萍;刘斯;王起运;贺星;董凯【摘要】脓毒症作为威胁生命的综合征,其发病机制复杂,是重症患者死亡的主要原因之一.自20世纪90年代初"脓毒症"概念提出以来,经历了"脓毒症-1"到"脓毒症-3"定义的演进,但是前两次定义的基本核心并无变化.2016年,第45届美国重症医学年会发布了"脓毒症-3"新定义及相应的临床诊断标准,将脓毒症定义为针对感染的宿主反应失调引起的致命性器官功能障碍.脓毒症-3是基于学者们对脓毒症本质有了更加深刻的理解而提出的,体现了人类对脓毒症发病机制的深入认识,满足了诊断标准合理化、临床诊断准确化和便捷化的需要.%Sepsis,a life-threatening syndrome with a complex pathogenesis,is one of the leading causes of death among severe cases. Since sepsis defined in 1990s,the evolution of sepsis definition has experienced from sepsis-1 to sepsis-3,but there was no change in basic cores of sepsis-1 and sepsis-2. In 45th Critical Care Congress of the Society of Critical Care Medicine′s(SCCM)in 2016,sepsis-3 was defined as life-threatening organ dysfunction caused by a dysregulated host responseto infection. Sepsis-3 not only reflected the deeper understanding of the pathogenesis of sepsis,but also met needs of rationalized diagnostic criteria,accurate and convenient clinical diagnosis.【期刊名称】《医学综述》【年(卷),期】2017(023)016【总页数】6页(P3230-3235)【关键词】脓毒症;脓毒症-1;脓毒症-2;脓毒症-3;诊断标准;感染【作者】陈晓洁;董天皞;张桂萍;刘斯;王起运;贺星;董凯【作者单位】天津红日药业股份有限公司,天津301700;天津红日药业股份有限公司,天津301700;天津红日药业股份有限公司,天津301700;天津红日药业股份有限公司,天津301700;天津红日药业股份有限公司,天津301700;天津红日药业股份有限公司,天津301700;天津红日药业股份有限公司,天津301700【正文语种】中文【中图分类】R631脓毒症作为一种由感染诱发的一系列病理、生理以及生化异常的综合征,是重症医学面临的主要问题,该病患者最终多因器官功能障碍死亡[1-2]。
毒理学名词解释
![毒理学名词解释](https://img.taocdn.com/s3/m/acee82dee109581b6bd97f19227916888486b98f.png)
毒理学名词解释第一篇:毒理学名词解释名词解释1、毒理学(T oxicology):研究外源性化学物质对生物机体的损害作用的学科(传统定义)。
2、现代毒理学(modern Toxicology):研究所有外源因素(如化学、物理和生物因素)对生物系统的损害作用、生物学机制、安全性评价与危险性分析的科学。
1、外源化学物(Xenobiotics):是在人类生活的外界环境中存在、可能与机体接触并进入机体,在体内呈现一定的生物学作用的化学物质,又称为“外源生物活性物质”。
2、毒性(toxicity):化学物引起有害作用的固有能力,毒性是一种内在的、不变的性质,取决于物质的化学结构。
3、毒物(poison,toxicant):在较低的剂量下可导致机体损伤的物质称为毒物。
4、损害作用(adverseeffect):(毒效应)指影响机体行为的生物化学改变,功能紊乱或病理损害,或者降低对外界环境应激的反应能力。
5、靶器官(target organ):外源化学物直接发挥毒作用的器官。
6、生物学标志(biomarker):外源化学物通过生物学屏障并进入组织或体液后,对该外源化学物或其生物学后果的测定指标。
通常把生物学标志分为暴露标志、效应标志和易感性标志。
7、毒物兴奋效应(Hormesis):指毒物在低剂量时有刺激作用,而在高剂量时有抑制作用。
其基本形式是U型,双相剂量-反应曲线。
8、半数致死剂量/浓度(median lethal dose or concentration,LD50/LC50):引起半数动物死亡所需的剂量。
通过统计处理计算得到,常用以表示急性毒性的大小,最敏感。
化学物质的急性毒性越大,其LD50的数值越小。
9、阈值(threshold):一种物质使机体(人或实验动物)开始发生效应的剂量或浓度,即低于阈值时效应不发生,而达到阈值时效应将发生。
10、急性毒作用带(acute toxic effect zone,Zac):半数致死剂量与急性阈剂量的比值,表示为:Zac=LD50/Limac。
脓毒症大鼠生物喋呤及铁代谢的相关性研究
![脓毒症大鼠生物喋呤及铁代谢的相关性研究](https://img.taocdn.com/s3/m/95bc9c3cfab069dc5122019d.png)
脓毒症大鼠生物喋呤及铁代谢的相关性研究摘要】目的探讨腹腔感染致脓毒症时重要器官生物喋呤、铁代谢的病理生理意义。
方法腹腔感染致脓毒症模型采用盲肠结扎穿孔法(CLP),用反相高效液相分析法测定24只大鼠肝、肺、肾等组织生物喋呤含量,用分光光度法测定其铁含量。
结果脓毒症大鼠2h时肝、肺、肾组织生物喋呤含量显著增多,而铁含量变化不显著。
结论生物喋呤参与了腹腔感染所致脓毒症的发生、发展过程,而与铁代谢有待于进一步研究。
【关键词】脓毒症生物喋呤铁代谢脓毒症及常常并发的多器官功能不全综合征(MODS)是内、外科危重病人死亡的重要原因之一[1],而生物喋呤(BH4)是脓毒症主要的指标。
本研究利用脓毒症大鼠模型,在不同时相观察不同组织中生物喋呤与铁代谢的指标,籍以为MODS的防治寻求可能的途径。
1 材料和方法1.1动物分组雄性Wister大鼠24只,体重220~300g,实验动物动物饲养1周以上,实验前夜禁食,自由饮水。
采用盲肠结扎穿孔(CLP)致脓毒症模型:2%戊巴比妥钠(80mg/kg体重)腹腔麻醉后固定、铺无菌洞巾,沿腹正中线作2cm切口,暴露盲肠,丝线结扎盲肠根部,避免肠梗阻,用18号针穿通盲肠3次,并留置1条宽2mm、两头贯通盲肠的橡皮片以防止针孔闭合。
其后,将盲肠放回腹腔,逐层缝合腹部切口。
动物皮下注射林格氏液抗休克。
动物随机分为以下两组:(1)正常对照(n=6):麻醉后活杀,留取肝、肺、肾组织标本待测;(2)脓毒症组(n=18):行CLP后,分别于2、8、16h活杀,留取肝、肺、肾组织标本待测。
1.2观察指标和方法1.2.1组织BH4含量测定:采用反相高效液相分析法[2]1.2.2组织中铁含量测定:采用南京建成生物工程研究所研制的分光光度法铁测定试剂盒1.3统计学处理各指标以㏑X±S表示,实验资料应用SAS统计软件包来进行处理,包括T检验方差及相关分析2 结果2.1组织生物喋呤含量变化正常对照组大鼠肝、肺、肾组织均有一定量的BH4,脓毒症大鼠各组织中BH4含量于2h即显著增多,于8h达高峰。
内毒素血症と血液凝固线溶系 マウス系统と内毒素に対する感受性.
![内毒素血症と血液凝固线溶系 マウス系统と内毒素に対する感受性.](https://img.taocdn.com/s3/m/eb40534da26925c52dc5bf35.png)
*Collected 18 flours after in injection of 50 ug of endotoxin (S, typhimurium).
** Collected 4 hours after i, v, injection of 100 ug of endotoxin (E, col i)
CYtotoxiclty
(%)
Two reactions were tested 18 hours after i, p, injection of endotoxin,
( ) No. of mice. * p <0,001 compared
with control.
Fig. 3 Bone marrow reactions after endo-
each other.
endotoxin induced strainand that these parameters
Fig. 1 Disseminated intravascular coagulation (DIC) in endotoxemia
Table 1 Sedimentation of the procoagulant activity
Bone marrow cells from 10 mice were pooled and 14, 500 X g sediment was prepared. Post-LPS-cells were collected 4 hours after i. v. injection of 100 ;ig of S. typhimurium endotoxin. Table 3 Procoagulant activity of cell populations of mouse bone marrow after
毒理学题目
![毒理学题目](https://img.taocdn.com/s3/m/c568aef232d4b14e852458fb770bf78a64293a51.png)
毒理学题目2008级预防医学毒理学期末复习资料一、名解1、Toxicology(毒理学):是研究化学物对生物体有害效应的一门科学。
2、In vivo studies(体内试验):是指使用整体的高等动物(主要是哺乳动物)模型体系的试验。
3、In vitro test systems(体外试验):是指不采用整体高等动物作为预测人类效应模型的那些试验体系。
4、T oxicogenomics(毒理基因组学):将功能基因组学技术用于毒理学,研究外源化学物有害效应和基因组功能与活性的关系的。
5、poisoning(中毒):是生物体受到毒物作用而引起功能性或器质性改变后出现的疾病状态。
根据病变发生的快慢,中毒可分为急性中毒和慢性中毒。
6、first-pass effect(首过效应):有时化学物在经胃肠道吸收进入体循环前可先在胃肠壁或肝脏代谢,这种化学物在进入体循环之前被机体清除的现象叫做首过效应。
7、biomarker(生物学标志):是指外源化学物通过生物学屏障进入组织或体液后,对该外源化合物或其生物学后果的测定指标,可分为暴露标志效应标志易感性标志。
8、CL(清除率):表示化学物质由各种途径(如经肾脏和肠道排泄,在肝脏进行生物转化,或经肺呼出)从机体清除的速率,即单位时间内清除的含有化学物质的体液量来确定。
9、distribution(分布):指外源化学物吸收进入血流或淋巴液后,随体循环分散到全身组织器官的过程。
10、free radicals(自由基):is a molecule or molecular fragment that contains one or more unpaired electrons in its outer orbital.是指独立游离存在的带有不成对电子的分子、原子或离子11、ultimate toxicant(终毒物):is the chemical species that reacts with the endogenous target molecule or critically alters the biological environment,initiating structural and/or functional alterations that result is toxicity.是指直接与内源性靶分子反应或严重地改变生物学微环境,启动结构性和功能性改变的化学物种类12、detoxication(解毒):Biotransformations that eliminate the ultimate toxicant or prevent its formation are called detoxications.是指排除终毒物或阻止其形成的生物学转化过程13、synergistic effect:多种化学物同时存在时的毒效应超过各单个化学物分别作用时毒物效应的总和。
药物毒理学之名词解释
![药物毒理学之名词解释](https://img.taocdn.com/s3/m/4a470eef9fc3d5bbfd0a79563c1ec5da50e2d63d.png)
药物毒理学之名词解释毒理学(toxicology )是一门研究在特定条件下,外源物(化学、生物、物理)对生物体有害作用的综合性学科。
毒性(toxicity):药物在机体中可能产生的有毒作用暴露(exposure):机体以不同途径和方式对药物的接触。
靶部位(target site):药物对机体产生毒性作用并造成损害的部位靶组织(target tissue):药物对机体产生毒性作用并造成损害的组织靶器官(target organ):药物对机体产生毒性作用并造成损害的器官。
剂量(dose):机体暴露于药物的量(外剂量、内剂量)效应、反应(effect, response):机体暴露于药物后出现的生物学改变量反应(graded response ):毒性反应强弱呈连续增减的量变。
质反应(quantal response ):毒性反应只能用全或无、阴性或阳性表示剂量-反应关系(dose-response-relationship):药物作用于机体的剂量与所引起的生物学效应强度或发生率间的关系。
未观察到损害作用的剂量(No-Observed Adverse Effect Level,NOAEL):用最敏感方法未能检出外源物毒性效应的最大剂量最大耐受量(maximal tolerance dose, MTD):机体能耐受的最大剂量。
半数致死量(median lethal dose,LD 50 ):能引起半数实验动物死亡的浓度或剂量最小中毒量(minimum toxic dose ):诱发机体产生毒性效应的最低剂量最小致死剂量(minimal Lethal Dose,LD 01):引起实验动物出现死亡的最低剂量毒性反应(toxic reaction ):剂量过大或药物在体内蓄积过多时对机体的脏器或组织发生的危害性反应。
过敏反应(allergic reaction):非肽类药物作为半抗原与机体蛋白结合后,经过敏感化过程而发生的反应。
毒理学基本概念-药理学
![毒理学基本概念-药理学](https://img.taocdn.com/s3/m/8804f2d0eefdc8d377ee3203.png)
Zch = Limac / Limch
精选2021版课件
22毒作用范围ຫໍສະໝຸດ • 用NOAEL表示阈剂量,毒作用带相当于毒作用范围 (margin of toxic effect, MOT) MOTac = LD50 / NOAELac MOTch = NOAELac / NOAELch
精选2021版课件
11
9. 效应与反应
• 效应(effect):也叫量反应(gradual response),指外源化 学物与机体接触后产生的生物学改变,可以用计量单位来表示, 如mg、mmHg、U等,属于计量资料
• 反应(response):也叫质反应(quantal response),指外源 化学物与机体接触后产生某种效应的个体在群体中所占的比率, 一般用百分率或比值表示,属于计数资料
• 毒物与非毒物之间没有绝对界限,取决于接触的剂量和接 触的条件
精选2021版课件
5
4. 毒效应
• 毒效应/作用(toxic effect):外源化学物本身或其代谢产 物在作用部位达到一定数量并停留一定时间,与生物大分 子成分相互作用,引起机体有害的生物学改变,又叫有害 作用(adverse effect)
精选2021版课件
10
8. 剂量
• 剂量(dose):是决定外源化学物对机体损害作用的重要因素 (“The dose makes the poison”,Paracelsus)。包括:
• 外剂量:给药剂量(administered dose),应用剂量(applied dose), 暴露剂量(exposure dose)
PFOS对斑马鱼的急性毒性及安全浓度评价
![PFOS对斑马鱼的急性毒性及安全浓度评价](https://img.taocdn.com/s3/m/122c531a17fc700abb68a98271fe910ef12dae7e.png)
PFOS对斑马鱼的急性毒性及安全浓度评价郭晋姝;王金有;武佳琪;曹谨玲【摘要】[Objective] To investigate the ecological toxicity effects of perfluorooctane sulfonates (PFOS) on Danio rerio.[Method] The acute toxicity effects of different concentrations of PFOS on D.rerio were studied by using static exposure method.[Result] The toxic symptoms of D.rerio appeared under the exposure of different concentrations of PFOS such as body roll, loss of equilibrium, declined swimming ability and respiratory ability.The death rate of D.rerio enhanced with increase of PFOS concentration and prolonging of exposure time, and there were obvious dose-response and time-response correlations.LC50 of PFOS to D.rerio at 24 , 48 , 72 , 96 h were 26.09, 9.08, 3.91, 2.58 mg/L, respectively.The safe concentration of PFOS to D.rerio was 0.258 mg/L.According to the grading standard of acute toxicity, the toxicity of PFOS to D.rerio belonged to high toxicity.[Conclusion] The research results can provide scientific basis for studying the ecological toxicity of PFOS and evaluating its ecological risk and hazards.%[目的]探讨全氟辛烷磺酸(PFOS)对斑马鱼的生态毒性效应.[方法]采用静态染毒法研究不同浓度的PFOS暴露对斑马鱼的急性毒性效应.[结果]不同浓度PFOS暴露条件下斑马鱼出现鱼体侧翻、失去平衡、游泳能力和和呼吸能力减弱等中毒现象,随着PFOS暴露浓度的增加和暴露时间的延长,斑马鱼的死亡率也相应增加,存在明显的剂量效应和时间效应关系.PFOS对斑马鱼24、48、72、96h的半致死浓度(LC50)分别为26.09、9.08、3.91和2.58 mg/L,安全浓度为0.258mg/L.根据急性毒性分级标准,判断PFOS对斑马鱼的毒性为高毒.[结论]该研究结果可为掌握PFOS的生态毒性以及评价其生态风险和危害提供科学依据.【期刊名称】《安徽农业科学》【年(卷),期】2017(045)007【总页数】4页(P92-95)【关键词】PFOS;斑马鱼;急性毒性;安全浓度;LC50【作者】郭晋姝;王金有;武佳琪;曹谨玲【作者单位】山西农业大学动物科技学院,山西太谷 030801;山西农业大学动物科技学院,山西太谷 030801;山西农业大学动物科技学院,山西太谷 030801;山西农业大学动物科技学院,山西太谷 030801【正文语种】中文【中图分类】S.94;X.174全氟辛烷磺酸(Perfluorooctane sulfonate,PFOS)是全氟化合物中最具有代表性的一种物质。
- 1、下载文档前请自行甄别文档内容的完整性,平台不提供额外的编辑、内容补充、找答案等附加服务。
- 2、"仅部分预览"的文档,不可在线预览部分如存在完整性等问题,可反馈申请退款(可完整预览的文档不适用该条件!)。
- 3、如文档侵犯您的权益,请联系客服反馈,我们会尽快为您处理(人工客服工作时间:9:00-18:30)。
Acute and chronic toxicity of endosulfan to the polychaeta, Perinereis1aibuhitensis in the indoor culture2345ABSTRACT67The polychaeta, Perinereis aibuhitensis is considered to be a key species in estuarine 8ecosystems, and it has been shown that it can improve the condition of sediment. The 9acute toxicity of endosulfan in polychaeta and its decomposition were studied. The 10series of endosulfan test concentrations 0, 1.25, 2.5, 5, 10, 15, 20mgL-1was set to 11determine toxicity to Perinereis aibuhitensis. The 24h median lethal concentration 12(24h LC50) was 55.57mgL-1, while 48h median lethal concentration (48h LC50) was 1315.56mgL-1. The safe concentration was found to be about 1.556mgL-1. In a long-term 14exposure experiment of 30 days, the endosulfan concentration in the seawater was 15initially set at 3mgL-1. During experiment, the specimen effectively decomposed 16endosulfan and a major endosulfan metabolite endosulfan sulfate. This shows their 17potential to significantly improve the aquatic sediment of an environment polluted by 18endosulfan and other toxic organic pollutants.1920Keywords:Endosulfan, endosulfan sulfate, environment restoration, LC50, Perinereis 21aibuhitensis.2223INTRODUCTION2425The organochlorine pesticide Endosulfan is used globally to control insects and 26mites. Endosulfan(6, 7, 8, 9, 10, 10 -hexachloro-1, 5, 5a, 6, 9, 9a-hexa-hydro-6, 279-methano-2, 4, 3-benzodioxathiepin-3-oxide) is applied as a mixture that typically 28contains two diastereoisomers, known as α-endosulfan and β-endosulfan,the form 2995% of the endosulfan mixture in ratios ranging from 2:1 to 7:3. [1] Endosulfan enters 30the aquatic environments by off-field movements, through runoff of contaminated 31surface soil due to rain events, accidental releases of irrigation tail water, or through 32the drift and overspray from aerial applications.[2]Once entering the environment, 33endosulfan changes to the diol form in water and the sulfate form in soil or sediment, 34and is further broken down to the ether, hydroxyl ether lactone, and alcohol forms.[3-5] 35Endosulfan sulfate is the sole metabolite which is considered to be toxic to the 36organism and have a similar toxicity with endosulfan.[6] The endosulfan and 37endosulfan sulfate are persistent when absorbed to soil and sediment; the half-lives 38range from weeks to years.[7,8]3940Both endosulfan and endosulfan sulfate are highly toxic to non-target aquatic 41organisms. Endosulfan was previously reported to be a fish toxicant in the 1950s.[9] It 42is also well documented that endosulfan was toxicity to crabs and other aquatic 43animals.[10] In South Korea, endosulfan has been sold and is still widely used because 44of it’s high efficiency and low cost. Furthermore, human dietary intakes of endosulfan 45in South Korea significantly increased, from 38.68 (1.3) ng kg bw−1 d−1in 1994 to 4692.17 (4.4) ng kg bw−1 d−1in 2011. [11] As seafood is popular with Korean people, it 47is therefor important to reduce the pollution of endosulfan and its metabolite in the 48seawater and aquatic sediments.4950The class Polychaeta, to which Perinereis aibuhitensis belongs to are a 51free-swimming group of annelids. As a deposit-feeding species, Perinereis 52aibuhitensis is often used to improve the coastal sediment. It has been shown to have 53potential of improving sediment conditions where scallop cultures exist. [12]5455There are parameters that can be measured to determine whether the aquatic 56environmental quality has been improved. In seawater, total organic carbon (TOC) 57and total nitrogen (TN) are the fundamental factors for the eutrophication of seawater.58In order to determine the rate at which endosulfan is removed from the sediment, the 59concentrations of endosulfan, endosulfan sulfate, and specemin density are also 60required. In addition, to detect the acute toxicity of endosulfan, LC50(Lethal 61Concentration, 50%) is also an important parameter. LC50 value is the concentration of62a material in water that will kill 50% of the test subjects when administered as a single63exposure over a specific time period.6465While most of the studies related to endosulfan toxicity and the decomposition in the 66respects of bioremediation were conducted in soil environment using microorganisms 67and earthworm, [13-16] there has been no works dealing with benthic species in coastal 68regions and their application to endosulfan degradation. Considering its widespread 69use in agriculture, the long range transport and endosulfan’s persistence in nature, it is 70important to assess the impact it has on the coastal benthos and it’s decay in the 71coastal sediment.7273In the present study, we evaluate the toxicity of endosulfan to the specimen and the 74endosulfans affect on the polychaeta’s ability to improve sediment. The aim of the 75study is to determine whether polychaeta, Perinereis aibuhitensis, could be introduced 76into endosulfan polluted environments for restoration purposes.7778MATERIALS AND METHOD7980Animal, sediment and seawater for indoor experiment8182The test specimens,P. aibuhitensis, were collected in Mokpo, Jeonnam, Korea.83Following collection, the specimens were immediately placed in an insulated box and 84transported to the laboratory. The specimens were then acclimated to test condition for 8596h before exposure to endosulfan. The sediments were purchased from the 86biotechnology company, without infaunal organisms. The sediment has a pH of 7, a 87particle diameter was less than 1mm, and was made up of 10% sand, 70% silt and 8820% clay. The specimens burrowed into the sediment, and survived without showing 89any abnormal appearance or behavior.9091Acute toxicity and degradation experiment9293The endosulfan used was purchased from Fluka. And stock solutions of endosulfan 94were prepared by dissolving the crystalline solids into 15 mL acetonitrile with 95concentrations dependent upon the experimental conditions. The primary tests for 96acute toxicity detection were conducted using the concentration of 5, 10, 20 and 40 97mgL-1 for a duration of 1 day. None of the animals in the 5 mgL-1 group died however 98all the animals in the 40 mgL-1 group had. According to the result, the acute toxicity 99bioassays, a series of endosulfan concentrations of 0, 1.25, 2.5, 5, 10, 15, 20mgL-1 100were obtained by diluting the stock solution with 30L of seawater. Then according to 101the LC50, the final concentration of endosulfan in the seawater for long-term exposed 102experiment was 3mgL-1. Two control experiments were conducted, one with only 103seawater and endosulfan with aconcentration of 3mgL-1, and the other containing only 104specimens in the tank. The experiment group contained both polychaeta and seawater 105with 3mgL-1 endosulfan.106107The experiments were conducted in a 10L tank with 5cm of sediment on the bottom, 108with 30L of circulating seawater. Three replications were prepared for each group. 30 109polychaetas (body length 16.0 ± 1.3 mm; body weight 0.4 ± 0.1 g) were introduced to 110each tank. Average water quality conditions during the experiment were a temperature 111of 22 ℃, a pH of 7.5, and a salinity of 30.6 gL-1. Water and sediment oxygenation and 112turbulence were produced with air pumps and air stones. In the acute toxicity test, the 113polychaetes were examined every 24h and the dead ones were removed. Over the 11430-day long-term exposure experiment, seawater, sediment and polychaeta samples 115were collected at days 0, 5, 10, 15, 20, 25, 30 and stored at -20℃ for further analysis. 116Nothing was supplied to the polychaeta for the duration of the experiments, as the 117animals could attain nutrition from the sediment.118119Chemical Analysis120For endosulfan and endosulfan sulfate, we determined their sediment concentrations 121in the control group, and both sediment and polychaeta concentrations in experiment 122group, as shown control sediment, experiment sediment and experiment worm in the 123Figure.1 and Figure.2.The endosulfan and endosulfan sulfate concentration were 124detected in the sediment and specimen at the sample times, 0, 5, 15, 25 and 30 days. 125The samples were analyzed using gas chromatography (Shimadzu 2010plus). 50g of 126sediment was extracted for 30 min with 100 mL CH3CN in a 300rpm mixer and then 127shook for additional 30min after adding 20-30g NaCl. The mixture was then 128centrifuged at 3000rpm for 5min and 10mL of the extract was decanted. Solvent was 129removed and the residue was extracted with 1mL of acetone/hexane (2/8 V/V). The 130sample solution was loaded into a solid phase extraction catridge (Florisil) and 131extracted with 5mL of acetone/hexane (2/8 V/V). After removal of the solvent, the 132residue was dissolved in 1mL acetone. Endosulfan was quantitatively analyzed by 133Shimadzu 2010plus equipped with an ECD and a 30 mm x 0.32 mm x 0.25 mm 134fused-silica DB-5 capillary column (Restok, RTx-5). The oven temperature 135programme was as follows: 180°C for 1 min, 3°C min -1to 250°C, hold for 1 min, 13620°C min -1to 300°C, hold for 3 min. Endosulfan and endosulfan sulfate were 137identified using a standard curve138139For polychaeta samples, 1-3 g of tissue sample was added to 100 mL of CH3CN, the 140mixture was grounded and centrifuged in 5000rpm for 3min. The next procedure was 141carried out as described above for sediment sample.142143The seawater samples were analyzed by the TOC and TN at the time 0, 5, 10 and 15 144days. TOC and TN of the seawater samples were analyzed by TOC/TN analyzer 145(Shimazu, TOC-V/TNM-1).146147Statistical analysis148149Statistical comparisons of mean cumulative mortalities were examined with ANOV A 150test at α=0.05. Lethal concentration of endosulfan at which 50% mortality of the test 151organism occurred (LC50) and its 95% confidence limit were estimated for 24h and 15248h by Probit analysis.[17] An application factor of 0.1 is recommended to be 153multiplied with the LC50 value to estimate the safe concentration.[18] So the safe 154concentration could be calculated by LC50×0.1.155156A one-way ANOV A followed by Duncan’s multiple comparison procedure was used 157to determine the significant difference of TOC, TN in seawater; endosulfan and 158endosulfan sulfate concentration with different elapsed time.159160RESULTS AND DISCUSSION161162Acute toxicity bioassays163164From the results (Table 1), none of the specimens had died in low concentrations 165(1.25, 2.5 and 5 mgL-1) over the course of the experiment, and at 24h, the mortalities 166at the concentrations of 10, 15, 20mgL-1were still low. There were no significant 167differences (P>0.05) among all concentrations after 24h. But after 48h, the mortalities 168at the concentrations of 10, 15, 20 mgL-1increased to 26.7%, 48.3% and 63.3% 169respectively. The moralities in the higher concentrations (10, 15 and 20 mgL-1) were 170found to be significantly different (P<0.05) from the lower concentrations (0, 1.25, 2.5 171and 5 mgL-1).172173Probit analysis in SPSS was carried out between the concentrations and mortalities. 174The probity of mortality was linearly dependent on the Log concentration (Tab le 2). 175Compared the R2between the 24h and 48h, the probity of mortality and Log 176concentration at 48h was more linearly dependent.177178According to the regression equation, the 24h median lethal concentration (24h LC50) 179of polychaeta by endosulfan was 55.57mgL-1 (Table 3) and its 95% confidence limits 180were 31.74-794.48. The 48h LC50 of polychaeta by endosulfan was 15.56mgL-1 and its 18195% confidence limits were 13.73-18.09. The concentration deemed safe was 1821.56mgL-1.183184Endosulfan, like other cyclodiene insecticides, has been proposed to cause 185neurotoxicity through GABA-gated chloride channel inhibition.[19,20]The endosulfan 186causes decreases in adenylate energy charge, oxygen consumption, hemolymph amino 187acids, succinate ehydrogenase, heart-rate (mussel) and altered osmoregulation.[19]. The 188present results indicated that the endosulfan is slightly toxic (10-100mgL-1) to the 189specimens, according to toxicity category. [21] The LC50 of endosulfan in 48h for fish 190has been reported as below 0.1mgL-1, [11, 22] and below 1mgL-1for crustaceans. [22, 23] 191Compared to other aquatic organisms, the polychaeta can endure high concentrations 192of endosulfan.193194Endosulfan and endosulfan sulfate for long-term exposed experiment195196Endosulfan and endosulfan sulfate in seawater samples were not detected by HPLC. It 197can be attributed to the hydrophobic nature of endosulfan (1.32mgL-1) it therefore 198tends to be adsorbed to the soil particles. [2, 14] Endosulfan sulfate was not detected, as 199it is not released outside the polychaeta specimens, it was trace detected in sediment. 200201For endosulfan (Fig.1), the accumulated amount of endosulfan by the polychaeta was 202the highest at the time 0. This means that the accumulation fate of endosulfan is very 203high. The endosulfan can be concentrated by the polychaeta because the endofulfan 204concentration in the polychaeta is much higher than it is in the sediment. In the 205control sediment with only endosulfan, the concentration increased in the first 5 days. 206Pablo and Hyne[2] have documented that endosulfan has the tendency to move 207preferentially towards or remain bound to solid gravel and sediment substrate. This 208may be the main reason for the increase of endosulfan in the first 5 days. After 5 days 209the endulsulfan concentration slowly declined with the time and almost absent after 21030 days. In the experiment group, endosulfan concentration in the sediment (t=0, 4.5 211mgL-1) was lower than 6.7 mgL-1of the control sediment indicating consumption and 212decomposition of endosulfan began immediately. Furthermore, endosulfan 213concentration in sediment of the experiment group decreased continuously to 0.11 214mgL-1after 30 days. The concentration of endosulfan in polychaeta reached the 215maximum at the time 0 and decreased with time. It is reported that freshwater animals 216accumulate endosulfan to some extent, but they lose the compound rapidly during 217depuration. [25]218219Endosulfan sulfate was also detected, (Fig.2) it accumulated in the specimens 220reaching a maximum concentration at the time of 15 days and then decayed 221completely. The formation of endosulfan sulfate took place simultaneously with the 222decomposition of endosulfan. In the sediment for both control and experiment group, 223little endofulfan sulfate was detected. The fact that endosulfan sulfate concentration in 224polychaeta eventually decreased and only trace amounts were detected in the sediment 225of both control and experiment groups, tells us that endosulfan sulfate was not 226extruded from the worm but decomposed further inside the worm. The other possible 227metabolites, such as endosulfan diol, endosulfan ether, endosulfan hydroxyl ether and 228endosulfan lactone were not detected at any time. Therefore polychaetas should be 229considered an excellent candidate for the remediation of endosulfan contaminated 230coastal environment, as they do not produce the harmful daughter pollutants.231232Seawater improvement for long-term exposed experiment233234For seawater (Fig.3 and Fig.4), the TOC and TN increased significantly (P<0.05) once 235the endosulfan dissolved in acetonitrile was introduced to the experiment at the time 0. 236The reason for the elevated TOC and TN at the beginning of the experiment was due 237to the CH3CN solvent, which was used to dissolve the endosulfan. 5mL of CH3CN 238used in each experiment and is equivalent to 3.93g. The theoretical TOC and TN 239caused by CH3CN in 30L seawater will be 76.44mgL-1 and 44.67mgL-1. The TOC and 240TN then decreased with time, which can be largely attributed to ventilation of the 241solvent by the aeration of seawater through the circulating system, the sorption of the 242sediment [8] and the possible decomposition by polychaeta. However, it should be 243noted that the decrease of TOC and TN in experiment group was more dramatic than 244the control group, indicating that the the polychaeta influenced the reduction.245246The movement of benthic invertebrates through sediment can accelerate the solvent 247ventilation, making favorable conditions for their aerobic decomposition. For example, 248the presence of various benthic marine organisms aids the acceleration of the 249biodegradation processes of DDT to DDE. [26] This finding and interpretation can be 250applied to removal of other volatile organic pollutants, which contaminate the coastal 251sediments and need to be cleaned by a environmentally benign and ecologically 252sustainable method.253254CONCLUSION2552561. According to the acute toxicity bioassay, the 48h LC50to P. aibuhitensis was 25715.56mgL-1, and the safe concentration is 1.56mgL-1in our experiment mesocosm. 258Compared to other aquatic species, P. aibuhitensis could endure a high concentration 259of endosulfan, which makes it possible to use the species to improve environment in 260endoulfan polluted areas.2612622. During the long-term exposure experiment, the endosulfan and endosulfan sulfate 263could be degraded without producing the harmful daughter pollutants by P. 264aibuhitensis effectively. In addition, the decrease of TOC and TN in experiment group 265is more profound than the control group due to the presence of P. aibuhitensis. So P. 266aibuhitensis can be considered a good candidate for the remediation of endosulfan 267contaminated coastal environment.268269ACKNOWLEDGEMENT270271The research was supported by Technology Development Program for Fisheries, 272Ministry for Food, Agriculture, Forestry and Fisheries, Republic of Korea.273274REFERENCE2751. Herrmann,M. Preliminary risk profile of endosulfan. Berlin Germany: 276Umweltbundesamt, 2002; 8.2772. Pablo, F.; Hyne, R. Endosulfan Application to a Stream Mesocosm: Studies on Fate, 278Uptake into Passive Samplers and Caged Toxicity Test with the Fish. Arch Environ 279Con Tox 2009, 56(3), 525-535.2803. Martens, R. Degradation of [8, 9,-14C] endosulfan by soil microorganisms. Appl 281Environ Microbiol 1976, 31(6), 853-8.2824. Miles, J.R.W.; Moy, P. Degradation of endosulfan and its metabolites by a mixed 283culture of soil microorganisms. Bulletin of Environmental Contamination and 284Toxicology 1979, 23(1), 13-19.2855. Stanley, K.A.; Curtis, L.R.; Simonich, S.L.; Tanguay, R.L. Endosulfan I and 286endosulfan sulfate disrupts zebrafish embryonic development. Aquat Toxicol 2009, 28795(4), 355-361.2886. Berntssen, M.H.; Glover, C.N.; Robb, D.H.; Jakobsen J.V.; Petri D. Accumulation 289and elimination kinetics of dietary endosulfan in Atlantic salmon (Salmo salar). 290Aquat Toxicol 2008, 86(1), 104-111.2917. Peterson, S.M.; Batley, G.E. The fate of endosulfan in aquatic ecosystems. 292Environmental Pollution 1993, 82(2), 143-152.2938. Leonard, A.W.; Hyne, R.V.; Lim, R.P.; Leigh, K.A.; Le, J.; Beckett, R. Fate and 294toxicity of endosulfan in Namoi River water and bottom sediment. J Environ Qual 2952001, 30(3), 750-759.2969. Maier-Bode, H. Properties, effects, residues and analytics of insecticide endosulfan, 297Residue Rev. 1968, 22, 1-44.29810. Rajeswari, K.; Kalarani, V.; Reddy, D.C.; Ramamurthi, R. Acute toxicity of 299endosulfan to crabs: Effect on hydromineral balance. Bulletin of Environmental 300Contamination and Toxicology 1988, 40, 212-218.30111. Desalegn, B.; Takasuga, T.; Harada, K.H.; Hitomi, T.; Fujii, Y.; Y ang, H.R.; Wang, 302P., Senevirathna, S.; Koizumi, A. Historical trends in human dietary intakes of 303endosulfan and toxaphene in China, Korea and Japan. Chemosphere 2011,83, 3041398-1405.30512. Kang, K.H.; Zhang, Z.F.; Bao, Z.M.; Zhou, B.; Kim, Y.H.; Webster, C.D. Effects of 306clamworm, Perinereis aibuhiteusis, density on acid volatile sulfide, chemical oxygen 307demand, and loss of ignition in benthic sediment, and on survival of scallop, 308Chlamys farreri, spat. Journal of the World Aquaculture Society 2010, 41,131-138. 30913. Kumar, M.; Philip L. Bioremediation of endosulfan contaminated soil and water 310optimization of operating conditions in laboratory scale reactors. J Hazard. Mater 3112006, 136(2), 354-36431214.Bhalerao, T.S.; Puranik, P.R. Biodegradation of organochlorine pesticide, 313endosulfan, by a fungal soil isolate, Aspergillus nige r, International Biodeterioration 314and Biodegradation 2007, 59, 315–319.31515. Goswami, S.; Vig, K.; Singh, D.K. Biodegradation of αand βendosulfan by 316Aspergillus sydoni. Chemosphere 2009, 75, 883-888.31716. Liu, W.; Zhu, L.S; Wang, J.; Wang, J.H.; Xie, H.; Song, Y. Assessment of the 318genotoxicity of endosulfan in earthworm and white clover plants using the comet 319assay. Arch Environ Contam Toxicol. 2009, 56(4), 742-746.32017. Finney, D.J. Probit analysis (3rd edition), Cambridge University Press, Cambridge, 321UK, 1971; 318.32218.Akinbulumo, M.O.; Fagbenro, O.A.; Fasakin, E.A. Acute toxicity of ethanolic 323extract of Derris elliptica roots to African catfish (Clarias gariepinus, Burchell, 324fingerlings. Biological and Environmental Sciences Journal of the Tropics (BEST) 3252005, 2, 126-131.32619. Naqvi, S.M.; V aishnavi, C. Bioaccumulative potential and toxicity of endosulfan 327insecticide to non-target animals. Comp Biochem Physiol C 1993, 105(3), 347-361. 32820. Jia, Z.; Misra, H.P. Developmental exposure to pesticides zineb and/or endosulfan 329renders the nigrostriatal dopamine system more susceptible to these environmental 330chemicals later in life. Neurotoxicology 2007, 28(4), 727-735.33121. Kamrin, M.A. Pesticide Profiles: Toxicity, Environmental Impact, and Fate. Lewis 332Publishers, 1997; 8.33322. Gill, T.S.; Pande, J.; Tewari, H. Individual and combined toxicity of common 334pesticides to teleost Puntius conchonius Hamilton. Indian J Exp Biol 1991, 29(2), 335145-148.33623. Office of Pesticide Programs. Pesticide Ecotoxicity Database (Formerly: 337Environmental Effects Database) Environmental Fate and Effects Division. U.S. 338EPA, Washington, D.C, 2000.33924.Wan, M.T.; Kuo, J.N.; Buday, C.; Schroeder, G.; V an Aggelen, G., Pasternak, J. 340Toxicity of α-, β-, (α+β)-endosulfan and their formulated and degradation products 341to Daphnia magna, Hyalella azteca,Oncophynchus mykiss, Oncophynchus kisutch, 342and biological implications in streams. Environmental Toxicology and Chemistry 3432005, 24(5), 1146-1154.34425. Naqvi, S.M.; Newton, D.J. Bioaccumulation of endosulfan (Thiodan insecticide) in 345the tissues of Louisiana crayfish, Procambarus clarkia,.J Environ Sci Health B 1990, 34625(4),511-526.34726.Pandit, G.G.; Sahu, S.K.; Sharma, S.; Puranik, V.D. Distribution and fate of 348persistent organochlorine pesticides in coastal marine environment of Mumbai. 349Environment International 2006, 32, 240-243.350FIGURE CAPTIONS351352Figure 1. The concentration of endosulfan, in the sediment and in P.aibuhitensis 353atdifferent elapsed times.354355Figure 2. The concentration of endosulfan sulfate, in the sediment and in 356P.aibuhitensis at different elapsed times. * Indicates significantly different from the 357time 0 (P<0.05).358359Figure 3.The TOC of seawater at different elapsed times.* indicates significantly 360different from initial TOC (P<0.05). Initial TOC means the TOC of the sediment in 361the natural condition.362363Figure 4. The TN of seawater at different elapsed times.* indicates significantly 364different from initial TN (P<0.05). Initial TN means the TN of the seawater in the 365natural condition.366367Fig1. The concentration of endosulfan, in the sediment and in P.aibuhitensis368at different elapsed times.369370Fig.2. The concentration of endosulfan sulfate, in the sediment and in P.aibuhitensis at 371different elapsed times. * indicates significantly different from the time 0 (P<0.05). 372373374Fig.3. The TOC of seawater at different elapsed times.* indicates significantly 375different from initial TOC (P<0.05). Initial TOC means the TOC of the sediment in 376the natural condition.377378Fig.4. The TN of seawater at different elapsed times.* indicates significantly different 379from initial TN (P<0.05). Initial TN means the TN of the seawater in the natural 380condition.381382Table 1. The acute toxicity of different concentrations of endosulfan to polychaeta 383Concentration (mgL-1)Cumulative Mortality(%)24h 48h0 0c0c1.25 0c0c2.5 0c0c5 0c0c10 5.0bc26.7b15 6.7bc48.3a20 13.3bc63.3aDifferent letters indicate significant difference (P<0.05).384385Table 2. Regression equation at different times386Time R2 Regression equation24h 0.877 Y=-4.341+2.488X48h 0.999 Y=-4.414+3.476X 387Table 3. The LC50 and safe concentration of endosulfan for the polychaeta388LC50(mgL-1) 95% confidence limit Safe concentration (mgL-1) 24h 55.57 31.74-794.48 5.5648h 15.56 13.73-18.09 1.56389390审稿意见(2011年10月4日)3911.392。