The equine antimicrobial peptide eCATH1 is effective against the facultative intracellular pathogen
抗菌肽
抗菌肽(antimicrobial peptide):抗菌肽原指昆虫体内经诱导而产生的一类具有抗菌活性的碱性多肽物质,分子量在2000~7000左右,由20~60个氨基酸残基组成。
这类活性多肽多数具有强碱性、热稳定性以及广谱抗菌等特点。
1分类:结构分为5类:(1)单链无半胱氨酸残基的α-螺旋,或由无规卷曲连接的两段α-螺旋组成的肽;(2)富含某些氨基酸残基但不含半胱氨酸残基的抗菌肽;(3)含1个二硫键的抗菌多肽;(4)有2个或2个以上二硫键、具有β-折叠结构的抗菌肽;(5)由其它已知功能的较大的多肽衍生而来的具有抗菌活性的肽。
来源分类可将其分为6类:(1)昆虫抗菌肽(2)哺乳动物抗菌肽(3)两栖动物抗菌肽(4)鱼类、软体动物、甲壳类动物来源的抗菌肽(5)植物抗菌肽(6)细菌抗菌2效应:抗菌肽具有广谱抗菌活性,对细菌有很强的杀伤作用。
3作用机制:一般认为抗菌肽杀菌机理主要是作用于细菌的细胞膜,破坏其完整性并产生穿孔现象,造成细胞内容物溢出胞外而死亡。
首先由静电吸引而附于细菌膜表面,疏水性的C端插入膜内疏水区并改变膜的构象,多个抗菌肽在膜上形成离子通道而导致某些离子的逸出而死亡。
亦有学者认为抗菌肽作用于膜蛋白引起凝聚、失活及离子通道,引起膜渗透性改变而导致死亡,不同类别的抗菌肽的作用机理可能不一样。
4.抗菌肽对细菌的杀伤作用抗菌肽对革兰氏阴性及阳性细菌均有高效广谱的杀伤作用。
国内外已报道至少有113种以上的不同细菌均能被抗菌肽所杀灭。
5.预防败血症:天然抗菌肽具有选择性免疫激活和调节功能,对败血症有良好的预防和保护作用。
败血症是由细菌感染引起的,伴随有全身性炎症反应综合症状的一种危重疾病。
病原微生物感染诱导促炎症因子大量释放,导致多种重要器官衰竭,具有较高死亡率。
6.总结:抗菌肽要成为药物,还需要解决一些问题。
首先是来源问题。
由于昆虫抗菌肽的天然资源有限,化学合成和基因工程便成为获取抗菌肽的主要手段。
Hp1404, a new antimicrobial peptide
Hp1404,a New Antimicrobial Peptide from the Scorpion Heterometrus petersiiZhongjie Li,Xiaobo Xu,Lanxia Meng,Qian Zhang,Luyang Cao,Wenxin Li,Yingliang Wu,Zhijian Cao *State Key Laboratory of Virology,College of Life Sciences,Wuhan University,Wuhan,PR ChinaAbstractAntimicrobial peptides have attracted much interest as a novel class of antibiotics against a variety of microbes including antibiotics resistant strains.In this study,a new cationic antimicrobial peptide Hp1404was identified from the scorpion Heterometrus petersii ,which is an amphipathic a -helical peptide and has a specific inhibitory activity against gram-positive bacteria including methicillin-resistant Staphylococcus aureus .Hp1404can penetrate the membrane of S.aureus at low concentration,and disrupts the cellular membrane directly at super high concentration.S.aureus does not develop drug resistance after multiple treatments with Hp1404at sub MIC concentration,which is possibly associated with the antibacterial mechanism of the peptide.In addition,Hp1404has low toxicity to both mammalian cells (HC 50=226.6m g/mL and CC 50.100m g/mL)and balb-c mice (Non-toxicity at 80mg/Kg by intraperitoneal injection and LD 50=89.8mg/Kg by intravenous injection).Interestingly,Hp1404can improve the survival rate of the MRSA infected balb-c mice in the peritonitis model.Taken together,Hp1404may have potential applications as an antibacterial agent.Citation:Li Z,Xu X,Meng L,Zhang Q,Cao L,et al.(2014)Hp1404,a New Antimicrobial Peptide from the Scorpion Heterometrus petersii .PLoS ONE 9(5):e97539.doi:10.1371/journal.pone.0097539Editor:Miguel A.R.B.Castanho,Faculdade de Medicine da Universidade de Lisboa,Portugal Received February 25,2014;Accepted April 18,2014;Published May 14,2014Copyright:ß2014Li et al.This is an open-access article distributed under the terms of the Creative Commons Attribution License,which permits unrestricted use,distribution,and reproduction in any medium,provided the original author and source are credited.Funding:This work is supported by the grants from National Key Basic Research Program in China (/English/AreaList.aspx):Nos.2010CB529800,and 2010CB530100.The funders had no role in study design,data collection and analysis,decision to publish,or preparation of the peting Interests:The authors have declared that no competing interests exist.*E-mail:zjcao@IntroductionThe increasing frequency of antibiotic resistance among microorganisms is becoming a more and more serious problem,which has outpaced the development of new antibiotics [1,2,3].It is urgently needed to discover new and more effective antimicro-bial agents.As a potential source of these agents,antimicrobial peptide (AMPs)are ubiquitous in nature,which can be found in microorganisms [4],insects [5],amphibians [6],mammals [7],and plants [8].They are produced as a part of the innate immune system defense,and show potent antimicrobial activity against a broad spectrum of microorganisms including resistant strains [9].Interestingly,the mechanisms of AMPs’s action are different from conventional antibiotics,most of which kill microorganisms rapidly by disrupting the integrity of the cytoplasmic membrane [10,11].Some of them can also interfere with the intracellular processes,such as affecting cell-wall biosynthesis pathway,inhibiting protein biosynthesis,or interacting with nucleic acids [12].These properties make them the attractive candidates for the development of new antimicrobial agents in overcoming microbial resistance.At least 2300different AMPs have been studied (/AP/main.php)during the last three de-cades,and several AMPs have been investigated as therapeutic agents in the past decade [9].As a living fossil,scorpion has survived over 400million years on earth,and developed diverse venom peptides for successful survival during its long-term evolution [13].So far,more and more AMPs have been identified from scorpion venoms,which can be divided into disulphide-bridged and non-disulphide-bridged peptides.Scorpine,a triple disulphide-bridge AMP from the scorpion Pandinus imperator has anti-bacterial and anti-malariaactivities [14].Non-disulphide-bridged AMPs Pandinins (from the scorpion Pandinus imperator )and IsCTs (from the scorpion Opisthacanthus madagascariensis )are a -helical polycationic peptides and have antimicrobial activity against both gram-positive bacteria and gram-negative bacteria [15,16,17].The non-disulphide-bridged AMP Vejovine from the scorpion Vaejovis mexicanus can inhibit the growth of multidrug resistant clinical isolates of gram-negative bacteria [18].These findings make scorpion venom as a potential source for discovering AMPs.We focus our interest on the scorpion species Heterometrus petersii ,which usually inhabits in tropical to subtropical rainforests.Various kinds of bacteria can grow and proliferate in this kind of living environment,which is conducive to the evolution of the scorpion venom to contain more AMPs.In this study,a new AMP named Hp1404was characterized from the venomous gland cDNA library of the scorpion Heterometrus petersii .Hp1404is an amphipathic a -helical peptide.The in vitro antibacterial activities of Hp1404peptide were then investigated using both standard and resistant strains.The mechanism of Hp1404against bacteria was further explored in our work.Finally,we tested the toxicities of Hp1404against mammalian cells and mice and the protective effect of Hp1404against infection to evaluate its potential application as an antibacterial agent.Materials and Methods Ethics statementThe scorpion Heterometrus petersii used in this work was obtained from a scorpion breeding base in Hubei,province of China.Thefemale balb-c(18–21g)mice were obtained from the Animal Facility at Wuhan University Zhong Nan Hospital.The mice were maintained under standard conditions of humidity(5065%), temperature(2562u C)and dark and light cycles(12h each)with free access to food and water.When met certain clinical criteria or at the end of the experiments,the mice were humanely euthanized (anesthetized by intra-peritoneal injection of pentobarbital,and sacrifice by cervical dislocation).All animal experiments were approved by the Institutional Animal Care and Use Committee of Wuhan University.The fresh human red blood cells(hBRCs)were from healthy donor Zhongjie Li,who is the author of this manuscript.The hBRCs-related experiment was approved by the Ethics Committee of the College of Life Sciences of Wuhan University.cDNA library construction and sequencingTwenty specimens of H.petersii were collected and milked for2 days by electrical stimulation.Total RNA was prepared from the glands by using TRIzol reagent(Invitrogen).Poly(A)mRNA was purified by using a Poly(A)Tract mRNA isolation system (Promega).The cDNA library was constructed with the Super-script plasmid system cDNA library construction kit(Gibco/BRL). cDNAs were cloned into the pSPORT1plasmid(Gibco/BRL)and transformed into Escherichia coli DH5a cells.cDNA clones were randomly chosen and sequenced to obtain a reliable representa-tion of the toxin content in the venom gland.Positive clones were identified by using an ABI Prism377XL DNA sequencer with a universal T7promoter primer.Sequence and secondary structure analysisSequence analysis was carried out by using BLASTX(http:// /Blast.cgi),DNAMAN,and GENRUNR. The secondary structure was analyzed by using the online program Heliquest(rs.fr/cgi-bin/ ComputParams.py)and measured by circular dichroism(CD) spectroscopy.The CD assay was performed at room temperature at the UV range of190–250nm at a concentration of0.1mg/mL in:(i)water,(ii)30%TFE/H2O,or(iii)70%TFE/H2O.Spectra were collected from three separate recordings.Peptide synthesis and bacterial strainsThe peptides used in this study were synthesized by GL Biochem(Shanghai,China)with amidated C-terminus,and with a purity of.95%.Staphylococcus aureus AB94004,S.aureus ATCC25923,S.aureus ATCC6538,S.aureus AB208193,S. epidermidis AB208187,S.epidermidis AB208188,Micrococcus luteus AB93113,Bacillus subtilis AB91021, E.coli AB94012, E.coli ATCC25922,Pseudomonas aeruginosa AB93066,Pseudomonas aerugi-nosa ATCC9027and Pseudomonas aeruginosa ATCC27853were purchased from the China Center of Type Culture Collection (CCTCC).MRSA P1381and MRSA P1374were obtained from the302nd military hospital of Beijing,China.Enterococcus faecium, Streptococcus agalactiae were clinical strains from a hospital. Antimicrobial assaysThe antimicrobial activity was determined in the broth microdilution assay from the procedure recommended by the Clinical and Laboratory Standards Institute with some modifica-tions.Briefly,bacteria were cultured in Luria-Bertani(LB)medium to OD600=0.6at37u C,then diluted to105–106cfu/mL in LB medium,and peptide was serially diluted in0.9%saline.160m L of the bacterial suspension and40m L of peptide dilution at varying concentrations were added into96-well plates,and then incubated for16h at37u C with continuous shaking at250rpm.The minimum inhibitory concentration(MIC)was determined as the lowest peptide concentration at which no bacterial growth was observed.Competition assaysA total of1mg/mL of Hp1404peptide solution was mixed with an equal volume of5mg/mL of the lipoteichoic acid(LTA, sigma-aldrich:L2515)or lipopolysaccharide(LPS,sigma-aldrich: L3012)solution.Then,the MICs of Hp1404treated with LTA or LPS against S.aureus AB94004were measured.The experiment was verified by three independent trials.Time-killing kineticsS.aureus AB94004was cultured in LB medium to exponential phase(OD600=0.5),then diluted to ca.107cfu/mL in LB medium,and treated with different concentrations of peptide solutions.Aliquots were taken at defined intervals and washed with 0.9%saline,and then diluted appropriately in the saline and plated on LB agar.The plates were incubated at37u C for24h, and the colony forming units(CFU)were counted.Confocal laser-scanning microscopyThe assay was measured according to the method[19]with some modifications.S.aureus AB94004was cultured to mid logarithmic phase(ca.107cfu/mL),harvested by centrifugation, and washed and resuspended with15mM sodium phosphates buffer(pH7.4).After incubated with12.5m g/mL fluorescein isothiocyanate(FITC)-labeled peptide at37u C for20min,the cells were washed.The glass slide was immersed in the suspension for10min to let the cells immobilized,and then examined by an Olympus IX70confocal laser-scanning microscope. Transmission electron microscopy(TEM)The exponential phase bacteria S.aureus AB94004(ca.107cfu/ mL)was incubated with Hp1404at the final concentration of26 MIC or56MIC or without Hp1404for20min at37u C. Samples were harvested by centrifugation,and washed with0.9% salt solution.The bacteria were fixed with2%glutaraldehyde in 0.1M PBS for1h,and with1%osmium tetroxide at4u C for another1h.After fixation,the bacteria were stained with1% uranyl acetate and then dehydrated by using a series of graded ethyl alcohols.After this,the samples were embedded in epoxy resins and stained with1%uranyl acetate and lead citrate.The samples were then semi-thin sectioned and examined by using a HITACHIH-8100transmission electron microscope. Fluorescence measurementsS.aureus AB94004was cultured to exponential phase(ca. 107cfu/mL),then the fluorescence dye SYTOX green(Invitro-gen)was added at a final concentration of5m M.After incubated for10min,the peptides were added at a final concentration of16 MIC,26MIC.Fluorescence was measured at the excitation and emission wavelengths of488and525nm,respectively.Drug resistant assaysThe initial MIC of Hp1404and control antibiotics kanamycin against S.aureus AB94004was obtained as described above. Bacteria from duplicate wells at the concentration of1/2-MIC were then diluted to105,106cfu/mL in LB medium for the new MIC assay.The experiment was repeated each day for15 passages.Checkerboard assayThe checkerboard assay is widely used for evaluation of properties of different antibacterial combinations [20].Kanamycin was serially diluted in 0.9%saline in the presence of a constant amount of peptide,equal to one-quarter of the peptide MIC.Then the combination MIC was measured.The fractional inhibitory concentration index (FICI)was used to determine the synergy between antimicrobial agents.Synergy was defined as an FICI #0.5.Indifference or absence of interaction was defined as 0.5,FICI ,4.Antagonism was defined as FICI .4.The FICI wascalculated as follows:FICI ~cMIC A MIC A alone z cMIC BMIC B alone,cMIC:MIC in combination.Hemolytic activityFresh hRBCs from healthy donors were washed 3times in 0.9%saline,and resuspended in the same salt solution to a concentra-tion of 2%(v/v).100m L of peptide solution and 100m L of RBC suspension were added to the wells of 96-well plate,which made the final concentration of hBRCs was 1%(v/v).After incubated for 1h at 37u C with gently shaking,samples were centrifuged at 1000g for 10min.The supernatant (100m L)from each well was transferred to a new 96-well plate and the absorbance wasmeasured at 490nm.0.9%saline and 1%Triton X-100was served as negative and positive controls,respectively.Hemolysis %~H sample {H negativeH positive {H negative|100%.H :absorbance at490nm.Cell culture and MTT assay The HFF,HEK293T and A375cell lines were cultured inDulbecco’s modified Eagle medium supplemented with 100m g/mL streptomycin,100U/mL penicillin and 10%(v/v)fetal bovineserum,and maintained in a humidified chamber with 5%CO 2at 37u C.Cytotoxicity of Hp1404against the mammalian cells was evaluated by MTT assays.Cells (6000cells/well)were pre-seeded on sterilized 96-well plates in 100m L medium and incubated for 24h.Dilutions of peptides in the same medium (100m L)were added and incubated for another 24h.Then 20m L of 3-(4,5-dimethylthiazol-2yl)-2,5-diphenyltetrazolium bromide (MTT)so-lution (5mg/mL)was added to each well and the plates were incubated for an additional 4h.The supernatant was removed,and 100m L dimethyl sulfoxide was added to the wells to dissolve any remaining precipitate.Absorbance at 570nm was measured using an ELISA reader.No peptide and 0.1%Triton X-100was served as negative and positive controls,respectively.The viabilityFigure 1.Sequence analysis,multiple alignment,and secondary structure analysis of Hp1404peptide.(A)cDNA and deduced amino acid sequences of Hp1404.The deduced amino acid residues are shown below the corresponding nucleotide sequences.The signal peptide residues are underlined.The mature peptide residues are shaded in gray.The potential cleavage site for prohormone convertase is marked in rectangle.The potential polyadenylation signal aataaa is double underlined.(B)Multiple alignment of Hp1404with other antimicrobial peptides.Sim%,the percentage of sequence identity relative to Hp1404.The different color represents the different homology level.StCT1and StCT2from the scorpion Scorpiops tibetanus ,CT1-NDBP-5.17from the scorpion Urodacus yaschenkoi ,IsCT from the scorpion Opisthacanthus madagascariensis ,OcyC2from the scorpion Opisthacanthus cayaporum ,pantinin 1from the scorpion Pandinus imperator .(C)Helical wheel diagram of Hp1404determined by the Heliquest method.The representation of Hp1404as a helical wheel shows the hydrophilic face and hydrophobic face.(D)CD spectra of Hp1404peptide (100m g/mL)in water alone or with 30or 70%aqueous TFE.doi:10.1371/journal.pone.0097539.g001人包皮成纤维细胞人胚肾细胞链霉素6292percentage was calculated according to the equation: Viability%~V sample{V positivenegative positive|100%.V:absorbance at 570nm.Acute toxicityThe acute systemic toxicity of Hp1404to mice was evaluated by the determination of the LD50(50%lethal dose).After being dissolved in0.9%saline,Hp1404was given to female balb-c mice (18–21g)by intraperitoneal(i.p.)or intravenous(i.v.)injection at the designated doses in0.4mL,six mice per group.The number of mice surviving at each group was monitored for up to a period of7days after treatment,and the values of LD50were calculated by the Karber method[21].Mouse peritonitis modelThe mouse peritonitis model was based on a previously described protocol[22].Female balb-c mice(18–21g)were infected by i.p.injection of0.5mL of the MRSA P1381inoculum (108CFU)in0.9%saline containing5%(wt/vol)mucin.One hour after bacterial administration,mice(6mice per group)were treated with a single dose of peptide by(i)i.p.or(ii)i.v.injection. Vancomycin and0.9%saline treatment were used as positive and negative controls,respectively.The mice were observed2–4times daily for7days.The mice were euthanized if they became moribund(unresponsive to external stimuli,can not swing the neck,and can not eat and drink),and counted as non-survivor.ResultsCharacterization and analysis of Hp1404After systemic screening the clones from the venom gland cDNA library of the scorpion H.petersii,we obtained a new peptide precursor named Hp1404.As shown in Figure1A,the cDNA sequence of Hp1404consists of a59UTR of62nt,an ORF of 210nt,and a39UTR of92nt.A single polyadenylation signal (AATAAA)was found16nt upstream of the poly(A)tail.The ORF of210nt encodes a precursor that consists of69amino acid residues,which contains a putative23-residue signal peptide (http://www.cbs.dtu.dk/services/SignalP/)and a46-residue pro-peptide.According to previous studies[23,24],further bioinfor-matic analysis indicated that the propeptide starts with a conserved posttranslational processing signal Gly-Lys-Arg at positions38to 40,and would result in a14-residue mature peptide(GILGKL-WEGVKSIF)with C-terminal amidation.Multiple sequence alignment revealed that Hp1404shared a high identity(Figure1B)with the AMPs StCT1[25],CT1-NDBP-5.17 [26],IsCT[27],StCT2[28],OcyC2[29],and pantinin1[30], which suggests that Hp1404peptide may have antibacterial activity. Secondary structure predicted by using the online program Heliquest(rs.fr/cgi-bin/ComputParams. py)indicated that Hp1404was a typical amphipathic a-helix,the helical wheel is divided into two parts:one part is the hydrophobic face,and the other is the hydrophilic face(Figure1C).Such structure was further confirmed by the CD spectral analysis.As shown in Figure1D,Hp1404exhibited only a large negative peak at198nm in water,indicating a random coil structure,while Hp1404exhibited a large positive peak at around195nm and large negative bands at208and220nm in TFE,indicating predominance of a-helices[31,32].These results suggested that Hp1404could form an amphipathic helical structure in the appropriate membrane environment.Antimicrobial activity in vitroAs shown in Table1,Hp1404has anti-bacterial activity with MICs of6.25–25m g/mL against gram-positive bacteria includingTable1.MICs of Hp1404against bacteria.Strains MICm g/mL m M Gram-positiveStaphylococcus aureus AB9400412.58.08 Staphylococcus aureus ATCC25923 6.25 4.04 Staphylococcus aureus ATCC653812.58.08 Staphylococcus aureus AB20819312.58.08 Staphylococcus epidermidis AB2081872516.16 Staphylococcus epidermidis AB2081882516.16 Micrococcus luteus AB9311312.58.08 Bacillus subtilis AB9102112.58.08 Staphylococcus aureus MRSA P138112.58.08 Staphylococcus aureus MRSA P1*******.16 Enterococcus faecium12.58.08 Streptococcus agalactiae2516.16 Gram-negativeEscherichia coli AB94012.100.64.6 Escherichia coli ATCC25922.100.64.6 Pseudomonas aeruginosa AB93066.100.64.6 Pseudomonas aeruginosa ATCC9027.100.64.6 Pseudomonas aeruginosa ATCC27853.100.64.6doi:10.1371/journal.pone.0097539.t001腹腔注射静脉注射腹膜炎粘蛋白万古霉素聚腺苷酸化翻译后的MRSA and other clinical strains,but it can not inhibit the growth of gram-negative bacteria at the concentration of 100m g/mL.LTA and LPS competition assaysTo determine whether LTA or LPS interfere with theantimicrobial activity of Hp1404,MICs of Hp1404mixed withLTA or LPS against S.aureus were measured.Our results (data not shown)showed that the MICs of Hp1404treated with LTA or LPS were 12.5m g/mL,which were the same as the untreated Hp1404.Antibacterial mechanismTo gain insights into the antibacterial mechanism of Hp1404,S.aureus AB94004was selected as the model bacteria.Firstly,the time-killing kinetics was studied.As shown in Figure 2A,the killing rate increased along with the increasing of the peptide concen-tration.The viable colony number only decreased 15%in 1h at the concentration of 26MIC,but it decreased approximately 85%in 30min at the concentration of 46MIC.To explore the target site of Hp1404in S.aureus AB94004,the distribution of Hp1404in bacteria was investigated by confocal laser-scanning microscopy.As shown in Figure 2B,FITC-labeled Hp1404penetrated the bacterial cell membrane and accumulated in the cytoplasm.To determine the direct influence of Hp1404on the bacteria,TEM was used to examine the ultrastructure changes of the bacteria.As shown in Figure 2C-a,untreated cells of S.aureus were round,proliferating cell with intact cell wall andwell-definedFigure 2.Antibacterial mechanism of the peptide Hp1404.(A)Time-kill kinetics of Hp1404against S.aureus AB94004.The assay was performed by determining the counts of surviving bacteria,0h represents bacteria before treated.(B)Confocal fluorescence microscopic images of S.aureus treated with FITC-Hp1404.Left:normal image;right:fluorescence image.(C)Transmission electron microscopy of S.aureus treated with Hp1404.a:negative control.b:treated with Hp1404at the concentration of 16MIC for 20min.c-d:treated with Hp1404at the concentration of 56MIC for 20min.(D)Fluorescence measurements.Negative control,0.9%saline.MSI78is an amphipathic a -helical peptide with an antibacterial mechanism of disrupting the membrane.The MIC of MSI78against S.aureus AB94004is 6.25m g/mL.doi:10.1371/journal.pone.0097539.g002Figure 3.Resistance development of S.aureus AB94004treated by Hp1404or kanamycin.doi:10.1371/journal.pone.0097539.g003酯磷壁酸脂多糖金葡菌穿透细胞质透射电镜超微结构界限清楚membrane,and the intracellular DNA region displayed a heterogeneous electron density.At the concentration of26 MIC,the cell wall and membrane had no changes(Figure2C-b). But,special structures could be observed in the cytoplasm (Figure2C-c:marked by arrow)and some cells were completely lysed(Figure2C-d)at the concentration of56MIC.To further assess whether Hp1404could influence the integrity of the membrane,the ability of permeabilizing the cell membrane of Hp1404was also tested with the fluorescent nucleic acid stain SYTOX.As shown in Figure2D,no fluorescence increase was apparent over30min when S.aureus cells were exposed toSYTOX and Hp1404,compared with a rapid fluorescence increase upon exposure to MSI78,which is an amphipathic a-helical peptide with a antibacterial mechanism of disrupting the membrane[33].Drug resistant assayThe emergence of drug resistance in bacteria for conventional antibiotic treatments is a major public-health concern.The development of resistance to Hp1404was evaluated in S.aureus AB94004.As shown in Figure3,the multiple treatments with kanamycin(MIC:6.25m g/mL)gave rise to drug resistance as early as passage2,and the MIC increased about32-fold after the course of9passages,while there was no change in the MIC for Hp1404during the course of15passages.These results indicated that it is difficult for S.aureus to develop resistance to Hp1404. Checkerboard assayTo evaluate whether Hp1404exhibits synergistic effects with kanamycin,the checkerboard assay was performed.Our results showed that the fractional inhibitory concentration index was0.5, which demonstrated that Hp1404exhibited synergistic effects with kanamycin.In vitro toxicityIn order to assess the toxicity of the peptide,the hemolytic activity to hRBCs and cytotoxicity to mammalian cell lines were tested.In comparison with BmKn2[34],a highly hemolytic peptide,Hp1404has a low hemolytic activity(about10%)at the concentration100m g/mL(Figure4A),and the HC50(50% hemolytic concentration)of the peptide Hp1404is226.6m g/mL (146.5m M).The MTT tests showed that the peptide only has about10–30%cytotoxicity to the cells be tested at the concentration of100m g/mL(Figure4B),and the CC50(50% cytotoxic concentration)is.100m g/mL(64.6m M).Taken together,the toxicity of Hp1404to the mammalian cells is lower than that to the sensitive bacteria.In vivo toxicityAcute toxicity was examined to evaluate the in vivo toxicity of Hp1404.With the dose up to80mg/Kg,the mice injected with Hp1404by i.p showed no immediate adverse events,and all the treated mice survived in the7-day study period(data not shown). In the i.v.injection treatment,the mice injected with Hp1404also showed no immediate adverse events at the dose of5or10mg/ Kg,but had a33.3%(2/6mice)mortality at the dose of80mg/ Kg and100%(6/6mice)mortality at the dose of160mg/Kg (Figure4C).Thus,the LD50of Hp1404to mice is89.8mg/Kg by i.v.injection.In vivo antibacterial activityTo further investigate the antibacterial activity of Hp1404 peptide in vivo,the potential therapeutic efficacy of the peptide Hp1404was evaluated by a mouse peritonitis model.As shown in Figure5A,all six mice in the negative control group died after inoculated with MRSA P1381for48h,but the mice treated with the dose of5or10mg/Kg Hp1404by i.p.injection showed a survival rate of100%.On the other hand,the mice treated with the dose of10mg/Kg Hp1404by i.v.injection also showed a survival rate of33.3%(2/6mice)in the7-day study period (Figure5B),but it was not as effective as the dose of5mg/Kg by i.p.All these data indicate that the peptide Hp1404still has anti-bacterial activity in vivo.DiscussionIn the present study,we indentified a new cationic AMP Hp1404with a net charge of+1from the scorpion H.petersii.In contrast to conventional AMPs with wide spectra of activity, Hp1404just has a specific potent antimicrobial activity againstFigure4.Toxicity of Hp1404in vitro and in vivo.(A)Hemolytic activity of Hp1404against human red blood cells.BmKn2is a highly hemolytic antimicrobial peptide from the scorpion Mesobuthus martensii Karsch.(B)Cytotoxicities of Hp1404against HFF,HEK293T and A375cell lines.Cytotoxicity was measured by MTT assay.(C)Acute toxicity of Hp1404to mice by intravenous injection.doi:10.1371/journal.pone.0097539.g004细胞内的透化CC50致半数细胞毒性所需浓度LD50的计算腹膜炎接种gram-positive bacteria(Table1).Previous studies showed that the interaction between AMPs and LPS or LTA is important to the activity of AMPs[12].Our results showed that the antibacterial activity of Hp1404was not interfered with LTA or LPS,which was different from the peptide Kn2–7,a scorpion AMP derivative studied in our previous work[35].It implied that LPS or LTA might not be the primary antimicrobial action site of Hp1404.Our further data by biolayer interferometry experiments showed that Hp1404did not interact with LPS or LTA indeed(data not shown).Thus,the mode of action of Hp1404may be different from that of most classical AMPs[35].Though Hp1404do not interact with LTA,two factors may let it successful reach the action sites and perform its activity against gram-positive bacteria: (i)The structure of the gram-positive bacteria cell wall.Such as S. aureus cell,which has a loosely arrayed network on the cell surface, consisting of fibrils and pores ranging in size from50to500A˚[36,37],while the Hp1404molecule(14aa)is about20–50A˚. These pores are large enough to let the peptide molecules to pass through them.(ii)Hp1404is a cationic peptide,while there are a lot of negatively charged molecules outside the bacterial cell,the electrostatic bonding between Hp1404and bacteria will encour-age the cationic peptide to pass through the cell wall[11]. Structure analysis suggested that Hp1404was an amphipathic and a-helical peptide(Figure1C,1D).Usually,natural AMPs with such structure are considered to be membrane-lytic peptides [38,39],which kill bacteria in2to3min after initial exposure [40,41].The killing of S.aureus by Hp1404peptide was a rather slow process(Figure2A),which suggested that Hp1404might not be a classical membrane-lytic peptide.Such assumption was confirmed by TEM,which showed that the membrane was not lysed by Hp1404at low MIC concentration(Figure2C-b).Our Fluorescence measurements also showed that Hp1404did not disrupt the membrane at low MIC concentration(Figure2D).But, it is interestingly that the FITC-Hp1404can penetrate the cell membrane of S.aureus and accumulate in the cytoplasm at low concentration(Figure2B),which is similar to the linear a-helical peptide Buforin II[19].As a short peptide with the length of14 amino acid residues,Hp1404may be via a floodgate mechanism to translocate to the cytoplasm[42].As we know,after penetrating the membrane,AMPs can inhibit the macromolecular biosynthesis or interact with specific vital components inside the microorgan-isms(43,46,47).Thus,Hp1404may have an unknown intracellular target,which needs further study to be confirmed in the future.On the other hand,Hp1404may interact with the membrane and lead to the lateral expansion of the membrane area at high MIC concentration,which result in forming mesosome-like structures(Figure2C-c),and lead to cell lysis(Figure2C-d).It is the same as PGLa[40].Due to the complex mechanism of action, it is difficult for S.aureus to develop resistance to Hp1404(Figure3), compared to conventional antibiotics.Figure5.Antibacterial activity of Hp1404in vivo.(A)Therapeutic efficacy of Hp1404on MRSA infected mice by intraperitoneal injection.(B) Therapeutic efficacy of Hp1404on MRSA infected mice by intravenous injection.doi:10.1371/journal.pone.0097539.g005万古霉素膜裂解肽。
抗菌肽概况
抗菌肽概况1.1 抗菌肽的基本概况抗菌肽又称抗微生物肽(antimicrobialpeptide)或肽抗生素(peptide antibiotics),在动植物体内分布广泛,是天然免疫防御系统的一部分。
抗菌肽是近年来发现的广泛存在于自然界的一类阳离子抗菌活性肽。
越来越多的证据表明它们在宿主先天性免疫和适应性免疫中有着重要的作用。
目前国内外对抗菌肽的研究开发正不断深入。
抗菌肽(antibacterialpeptides)广义上是指存在于生物体内具有抵抗外界微生物侵害、消除体内突变细胞的一类小分子多肽。
抗菌肽是由生物细胞特定基因编码,经特定外界条件诱导产生的一类多肽。
1972年,瑞典科学家Boman对惜古比天蚕(Hyalophoracecropia)蛹注射蜡状芽孢杆菌(Bacilluscereus),首次发现了抗菌肽cecropin。
此后,对抗菌肽的研究取得了很大的进展,目前在昆虫、植物、哺乳动物、病毒、两栖类以及人类中已发现类似的抗菌活性物质达2000多种。
抗菌肽广泛存在于动物的免疫细胞(如吞噬细胞)、各种脏器的粘膜、皮肤以及植物的花、果、叶中。
有专家推测,抗菌肽在进化意义上最早可能参与了早期真核细胞的噬菌作用,这种作用既是细胞自身防御的需要,而且有可能通过降解微生物为自身生长提供需要的营养,并且最终在生物进化过程中作为防御分子被保留下来。
由于抗菌肽具有小分子的特点,可以快速合成并易于大量存储,与特异性免疫反应相比能更加迅速地对病原菌作出反应,使其成为生物机体先天性非特异性防御系统的重要组分,此外,抗菌肽还具有稳定、水溶性好、抗菌机制独特、对高等动物正常细胞无害等特点,显示了在医学和农业上潜在的研究价值和应用价值。
近年来,有关抗菌肽及其应用逐渐成为动物学、植物学、药理学及生理学等领域的研究热点。
1.2 抗菌肽的理化性质包括细菌、真菌、昆虫、被囊动物(tunicate)、两栖类动物、甲壳类动物、鸟类、鱼类、哺乳动物(包括人类)以及植物在内的所有生物体都可产生抗菌肽。
苏格兰 围术期预防的抗菌药物使用
KEY TO EVIDENCE STATEMENTS AND GRADES OF RECOMMENDATIONS
LEVELS OF EVIDENCE 1++ 1+ 12++ 2+ 23 4 High quality meta-analyses, systematic reviews of RCTs, or RCTs with a very low risk of bias Well conducted meta-analyses, systematic reviews, or RCTs with a low risk of bias Meta-analyses, systematic reviews, or RCTs with a high risk of bias High quality systematic reviews of case control or cohort studies High quality case control or cohort studies with a very low risk of confounding or bias and a high probability that the relationship is causal Well conducted case control or cohort studies with a low risk of confounding or bias and a moderate probability that the relationship is causal Case control or cohort studies with a high risk of confounding or bias and a significant risk that the relationship is not causal Non-analytic studies, eg case reports, case series Expert opinion
抗菌肽的抗菌机制及其在反刍动物中应用的研究进展
抗菌肽(antimicrobial peptides,AMPs )因其独特的抗细菌、真菌、病毒以及抗癌细胞等生物学功能且不易产生耐药性,使其成为最有前景的抗生素替代品之一。
从20世纪80年代瑞典科学家Hulmark 从惜古比天蚕(Hyalophora cecropia )中分离出第一种抗菌肽,命名为天蚕素(Cecropin )[1],到目前为止抗菌肽数据库中已注册的抗菌肽序列已经超过3000个[2]。
抗菌肽是包括植物、动物和人类在内的所有生物体天然免疫反应的保守部分,是许多脊椎动物免疫系统的主要组成部分[3],被定义为能够保护宿主免受细菌、病毒或真菌入侵的关键防御分子[4]。
抗菌肽是由基因编码、核糖体合成的多肽,通常具有短肽(30~60个氨基酸)、强阳离子(pI 8.9~10.7)、热稳定性(100℃,15min )、不易产生耐药性、对真核细胞无影响等共同特征[5]。
根据其来源可以分为:植物源抗菌肽,如硫素(thionins )、植物防御素(plant defensins);动物源抗菌肽,如天蚕素、防御素;微抗菌肽的抗菌机制及其在反刍动物中应用的研究进展■纵瑞1胡忠泽1*张乃锋2段心明3(1.安徽科技学院动物科学学院,动物营养调控与健康安徽省重点实验室,安徽滁州233100;2.中国农业科学院饲料研究所,北京100081;3.农发苑(浙江)农业发展有限公司,浙江湖州313000)作者简介:纵瑞,硕士,研究方向为动物营养与饲料科学。
通讯作者:胡忠泽,教授。
收稿日期:2021-03-25基金项目:国家自然科学基金[31872385];安徽省高校协同创新项目[GXXT-2019-035];安徽省现代牛羊产业技术体系[AHCYTX-7];滁州市科技计划项目[2019ZN003]摘要:抗菌肽(antimicrobial peptides,AMPs )是自然界中广泛存在的多肽物质。
作为机体先天免疫的关键组成部分,具有抗细菌、真菌、肿瘤、病毒等生物学功能。
预防兽医学的一级学科
预防兽医学的一级学科英文回答:Preventive veterinary medicine is a discipline that focuses on preventing diseases and promoting the overall health and well-being of animals. As a veterinarian, myrole is to work proactively to identify potential health risks and implement measures to prevent them from occurring.One of the key aspects of preventive veterinarymedicine is vaccination. Vaccines are a crucial tool in preventing the spread of infectious diseases among animals. For example, in the case of dogs, vaccines against diseases such as rabies, distemper, and parvovirus are essential to protect their health. By ensuring that pets are up to date with their vaccinations, we can significantly reduce therisk of outbreaks and keep them healthy.Another important aspect of preventive veterinary medicine is regular health check-ups. Just like humans,animals can develop various health issues that may go unnoticed without proper examination. During check-ups, I conduct a thorough physical examination, including checking the animal's weight, temperature, heart rate, and overall condition. This helps me detect any underlying health problems early on, allowing for prompt treatment and prevention of further complications.Diet and nutrition also play a significant role in preventive veterinary medicine. Providing animals with a balanced and nutritious diet is essential for their overall health and well-being. For example, feeding a cat with adiet that meets its specific nutritional needs can help prevent obesity, diabetes, and other diet-related health issues. Additionally, I may recommend dietary supplementsto address specific nutritional deficiencies or support certain health conditions.Regular dental care is another important preventive measure in veterinary medicine. Dental problems, such as periodontal disease, can lead to serious health issues ifleft untreated. Regular teeth cleaning, dental examinations,and the use of dental chews or toys can help maintain good oral hygiene and prevent dental diseases in animals.Furthermore, preventive veterinary medicine alsoinvolves educating pet owners on responsible pet care. This includes providing information on proper nutrition, exercise, grooming, and parasite control. By empowering pet owners with knowledge and resources, we can ensure that animals receive the care they need to stay healthy and prevent diseases.中文回答:预防兽医学是一门专注于预防疾病并促进动物整体健康的学科。
如何避免浪费药材英语作文
如何避免浪费药材英语作文Title: How to Avoid Wasting Medicinal Herbs。
In the realm of traditional medicine and herbal remedies, the significance of preserving and utilizing medicinal herbs efficiently cannot be overstated. The sustainability of these resources and the efficacy of treatments heavily rely on judicious usage and conservation efforts. In this essay, we will explore various strategiesto prevent the wastage of medicinal herbs.First and foremost, proper cultivation practices play a pivotal role in minimizing wastage. Cultivating medicinal herbs in controlled environments such as botanical gardensor organic farms ensures optimal growth conditions and reduces the risk of pests and diseases. Additionally, employing sustainable farming techniques like crop rotation, companion planting, and natural pest control methods can enhance yield while preserving the integrity of the ecosystem.Furthermore, harvesting techniques significantly impact the sustainability of medicinal herbs. Harvesting at the right time, when the plants have reached their peak potency, ensures maximum therapeutic benefits. It is essential to employ gentle harvesting methods to minimize damage to the plants and facilitate regrowth. Moreover, selective harvesting, where only the necessary parts of the plant are collected, prevents overexploitation and allows for continued regeneration.Proper post-harvest handling is equally crucial in preventing wastage. Immediate processing or drying of harvested herbs helps maintain their potency and prevents spoilage. Adequate storage in airtight containers, awayfrom direct sunlight and moisture, preserves the quality of medicinal herbs for an extended period. Implementing inventory management systems can also prevent surplus stock from expiring and going to waste.In addition to cultivation and harvesting practices, responsible consumption habits contribute to theconservation of medicinal herbs. Practitioners and consumers should be educated about the appropriate dosage and frequency of herbal remedies to avoid excessive use and unnecessary wastage. Furthermore, promoting the use of alternative treatments or herbal substitutes when possible can help alleviate pressure on endangered plant species.Collaborative efforts among stakeholders are essential for effective conservation and sustainable utilization of medicinal herbs. Governments, research institutions, farmers, herbalists, and consumers must work together to establish regulations, guidelines, and awareness campaigns aimed at preserving medicinal plant diversity and promoting responsible usage.Research and innovation also play a vital role in addressing the challenges of medicinal herb wastage. Developing efficient extraction methods, formulating stable herbal preparations, and exploring novel uses for underutilized plant species can enhance the value chain and reduce losses along the supply chain.In conclusion, preventing the wastage of medicinal herbs requires a multi-faceted approach encompassing cultivation, harvesting, post-harvest handling, consumption practices, and collaborative efforts among stakeholders. By adopting sustainable practices, promoting responsible consumption, and fostering innovation, we can ensure the conservation of medicinal plant resources for generations to come.。
抗微生物药物(英文PPT)Antimicrobial Agents
• Antibiotics work to kill bacteria.Antibiotics are specific to certain bacteria and disrupt their function.
Susceptibility vs. Resistance (cont.)
• Dose of drug has to be sufficient to produce effect inhibit or kill the microorganism:
• However concentration of the drug must remain below those that are toxic to human cells –
• Limitation of in vitro tests • In vitro sensitivity tests are based on non-toxic
plasma concentrations –cut off • Do not reflect concentration at the site of infection • E.g.: G- aer.bacilli like Ps.aeruginosa inhibited by
What is an Antibiotic?
• An antibiotic is a selective poison.
• It has been chosen so that it will kill the desired bacteria, but not the cells in your body. Each different type of antibiotic affects different bacteria in different ways.
EP 5.1.4 非无菌制剂和药用物质的微生物质量标准(中英文)
01/2011:50104 5.1.4. MICROBIOLOGICAL QUALITY OF NON-STERILE PHARMACEUTICAL PREPARATIONS AND SUBSTANCES FOR PHARMACEUTICAL USE (1) 非无菌制剂和药用物质的微生物质量The presence of certain micro-organisms in non-sterile preparations may have the potential to reduce or even inactivate the therapeutic activity of the product and has a potential to adversely affect the health of the patient.在非无菌制剂中如果存在某些微生物,可能会降低或抑制药物的治疗活性,可能会对病人健康有潜在不良影响。
Manufacturers therefore have to ensure a low bioburden of finished dosage forms by implementing current guidelines on Good Manufacturing Practice during the manufacture, storage and distribution of pharmaceutical preparations.因此,生产商应在药品生产、存贮和销售过程中遵守现行GMP指南,来保证制剂的微生物在一个较低的水平。
Microbial examination of non-sterile products is performed according to the methods given in general chapters 2.6.12 and 2.6.13. Acceptance criteria for non-sterile pharmaceutical products based upon the total aerobic microbial count (TAMC) and the total combined yeasts/moulds count (TYMC) are given in Tables 5.1.4-1 and 5.1.4-2. Acceptance criteria are based on individual results or on the average of replicate counts when replicate counts are performed (e.g. direct plating methods).非无菌产品微生物检查应依据通则2.6.12和2.6.13中指定的方法进行。
加拿大拟定肟菌酯的最大残留限量
加拿大拟定肟菌酯的最大残留限量
2008年5月20日,加拿大卫生部有害生物管理局(PMRA)拟定肟菌酯(Trifloxystrobin)最大残留限量。
目前,加拿大的最大残留限量是通过官方公报进行磋商后,根据食品药物法规(FDR)制定的。
通过Bill C-28对食品药物法的修订,预计于2008年生效,这将允许按照有害生物控制产品法合法的制定杀虫剂最大残留限量,而无须经过FDA所属法规的批准。
本文件的目的是对所列肟菌酯(Trifloxystrobin),包括代谢物CGA-321113的MRLs进行咨询,这些限量是在PCPA于2008年6月28日生效后由PMRA拟定的。
咨询活动已经在Bill C-28生效之前开始,以便在FDA被修改后尽快合法的制定本文件所列的MRLs。
(注意:在将有关杀虫剂最大残留限量立法从食品药物法案过渡为有害生物控制产品法案(对拟定最大残留限量的磋商(PMRL2006-01))的文件中拟定的0.04ppm最大残留限量在G/SPS/N/CAN/276中通报)。
所列产品补充了作物14组核果的最大残留限量。
抗微生物药物英文PPTAntimicrobial
Antibacterial Antibiotics Inhibitors of Cell Wall Synthesis
• Polypeptide antibiotics
– Bacitracin
• Topical application • Against gram-positives
– Gram-positives
• Binds 50S, prevents translocation
• Oxazolidinones
– Linezolid
• Gram-positives
– Binds 50S subunit, prevents formation of 70S ribosome
Figure 20.12
Antibacterial Antibiotics Inhibitors of Cell Wall Synthesis
Figure 20.8
Antibacterial Antibiotics Inhibitors of Cell Wall Synthesis
• Cephalosporins
– 2nd, 3rd, and 4th generations more effective against gramnegatives
Antibacterial Antibiotics Injury to the Plasma Membrane
• Polymyxin B
– Topical – Combined with bacitracin and neomycin in
– Changes shape of 30S subunit
Antibacterial Antibiotics Inhibitors of Protein Synthesis
预防兽医学英语
预防兽医学英语English:Preventive veterinary medicine focuses on strategies and practices aimed at maintaining the health and well-being of animals through preventive measures rather than treating diseases after they occur. This field encompasses various approaches such as vaccination programs, regular health screenings, parasite control, nutrition management, environmental enrichment, and behavioral interventions. Vaccination plays a crucial role in preventive veterinary medicine by providing immunity against common infectious diseases, thereby reducing the risk of outbreaks and transmission within animal populations. Regular health check-ups allow veterinarians to detect potential health issues early, enabling prompt intervention and treatment. Effective parasite control involves the use of anthelmintics and ectoparasiticides to prevent infestations and minimize the spread of parasitic diseases. Nutrition management ensures that animals receive balanced diets tailored to their specific nutritional requirements, promoting overall health and disease resistance. Environmental enrichment strategies aim to create stimulating and comfortable living environments for animals,reducing stress and preventing behavioral problems. Behavioral interventions address issues such as aggression, anxiety, and compulsive behaviors, promoting mental well-being and reducing the risk of injury. By implementing these preventive measures, veterinarians can significantly contribute to the overall health and welfare of animals under their care.中文翻译:预防兽医学专注于通过预防措施而不是在疾病发生后进行治疗,旨在维护动物的健康和福祉。
抗菌肽
抗菌肽抗菌肽概述抗菌肽(Antibacterial Peptide) 又叫抗微生物肽(Antimicrobial Peptides),是由多种生物细胞特定基因编码经外界条件诱导产生的一类具有广谱抗细菌、真菌、病毒、寄生虫和肿瘤细胞等活性作用的多肽。
由于抗生素的广泛使用导致了微生物耐药性,新型抗菌药物研发成为近年研究热点,而抗菌肽是生物天然免疫重要的效应分子,具有调节宿主免疫的作用,所以有极大的临床应用潜力。
目前已有2200 多种的抗菌肽,来源有细菌、真菌、人体、昆虫等,其中有些抗菌肽的基因序列已得到测定。
抗菌肽具有多种优点,如无毒副作用(仅少数种类具有溶血活性)、无污染、无残留,难以产生耐药性,能抑制癌细胞扩增,调节机体免疫反应、抗炎功能,已经成为近些年来生物学及药学领域研究的热点。
抗菌肽是带有正电荷的、螺旋的、序列较短的具有抑菌作用物质的总称,一般是指从各个生物体包括单细胞生物、植物、昆虫、鱼类、鸟类以及哺乳动物等分离出来的,由12~60个氨基酸残基组成的多肽,分子质量一般小于 10 ku,被认为是先天免疫的重要组成部分,因为其可以抵抗外来微生物的入侵。
抗菌肽由于富含疏水基团,通常存在疏水区和亲水区,并且对细胞膜显现出两亲性。
目前已经有很多学者从食源性蛋白质中分离出抗菌肽,其中研究最早的是 1966 年从牛乳酪蛋白中分离得到的对多种细菌均具有抑菌作用的 抗菌肽。
在 1972 和 1980 年,瑞典科学家Boman 等和 Hultmark 等分别通过诱导眉纹天蚕蛾蛹和惜古比天蚕( Hyalophora cecropia) 发现了类似的具有抑菌活性的物质。
而第一个真正意义上的抗菌肽就是由 Boman 命名的天蚕素( cecropins) , 该物质对革兰氏阳性和阴性菌都有抑制作用。
此 后各种抗菌物质陆续被发现,其中昆虫和某些非 脊椎动物被广泛研究,迄今为止已发现超过 2 000种天然存在的抗菌肽。
抗菌肽创新 合成生物学
抗菌肽创新合成生物学英文回答:Antimicrobial peptides (AMPs) are a class of small proteins that play a crucial role in the innate immune response of various organisms, including humans. These peptides have attracted significant attention due to their potential as novel therapeutic agents against antibiotic-resistant bacteria. As a researcher in the field of synthetic biology, I am excited about the opportunitiesthat lie in the innovation of antimicrobial peptides.Synthetic biology is an interdisciplinary field that combines engineering principles with biological sciences to design and construct new biological parts, devices, and systems. It offers a powerful toolkit for the engineering of antimicrobial peptides with enhanced properties, such as increased potency, broader spectrum of activity, and improved stability. By manipulating the genetic code, we can introduce specific modifications in the amino acidsequence of AMPs to optimize their antimicrobial activity.For example, one approach is to use rational design to modify the peptide sequence based on known structure-activity relationships. By studying the three-dimensional structure of AMPs and their interactions with microbial membranes, we can identify key amino acid residues that contribute to their antimicrobial activity. Through targeted mutagenesis, we can introduce specific amino acid substitutions to enhance the peptide's potency orselectivity against certain types of bacteria.Another approach is to employ directed evolution techniques to generate libraries of AMP variants with diverse sequences. By subjecting these libraries to high-throughput screening or selection methods, we can identify peptides with improved antimicrobial properties. This process mimics the natural process of evolution, where mutations and selection lead to the emergence of new traits in living organisms.Furthermore, synthetic biology enables the productionof antimicrobial peptides through recombinant DNA technology. By introducing the genes encoding AMPs intohost organisms, such as bacteria or yeast, we can harness their cellular machinery to produce large quantities of peptides. This approach offers a cost-effective andscalable method for peptide production, which is crucialfor their development as therapeutic agents.中文回答:抗菌肽是一类小分子蛋白,在包括人类在内的各种生物体的先天免疫反应中起着重要作用。
抗倍特板英文术语
抗倍特板英文术语Antimicrobial Resistance: A Global Public Health ChallengeAntimicrobial resistance (AMR) has emerged as a pressing global health concern, posing a significant threat to the effective treatment of infectious diseases. The term "antimicrobial resistance" refers to the ability of microorganisms, such as bacteria, viruses, fungi, and parasites, to resist the effects of antimicrobial agents, including antibiotics, antivirals, and antifungals. This phenomenon has far-reaching consequences, as it can lead to the failure of standard treatments, prolonged illnesses, increased healthcare costs, and even the spread of untreatable infections.The rise of AMR can be attributed to various factors, including the overuse and misuse of antimicrobial agents, poor infection control practices, inadequate sanitation and hygiene, and the lack of new antimicrobial drug development. In many parts of the world, antibiotics are readily available without prescription, leading to their indiscriminate use, which contributes to the selection and spread of resistant strains. Furthermore, the lack of effective surveillance systems and limited access to diagnostic tools in some regions make it challenging to monitor and address the problem effectively.The consequences of AMR are far-reaching and can have a significant impact on global health and economic stability. Resistant infections can result in longer hospital stays, increased mortality rates, and higher healthcare expenditures. The World Health Organization (WHO) has identified AMR as one of the top 10 global public health threats facing humanity, with the potential to cause up to 10 million deaths per year by 2050 if left unchecked.To address this global challenge, a comprehensive and coordinated approach is necessary, involving various stakeholders, including governments, healthcare providers, the pharmaceutical industry, and the general public. The WHO has developed a global action plan on AMR, which outlines five strategic objectives: improving awareness and understanding of AMR, strengthening the knowledge and evidence base through surveillance and research, reducing the incidence of infection through effective sanitation, hygiene, and infection prevention measures, optimizing the use of antimicrobial agents, and promoting investment in new medicines, diagnostic tools, vaccines, and other interventions.At the national level, many countries have developed their own action plans and strategies to combat AMR. These efforts often include the implementation of antimicrobial stewardship programs, the regulation of antimicrobial use in human and animal health, thepromotion of infection prevention and control measures, and the investment in research and development of new antimicrobial agents.One of the key strategies in the fight against AMR is the concept of "One Health," which recognizes the interconnectedness of human, animal, and environmental health. This approach emphasizes the need for collaboration across different sectors, including human medicine, veterinary medicine, and environmental sciences, to address the multifaceted nature of AMR. By adopting a holistic perspective, stakeholders can develop and implement more effective interventions that target the various drivers of AMR.Another important aspect of the global response to AMR is the need for international cooperation and coordination. The WHO, the Food and Agriculture Organization (FAO), and the World Organisation for Animal Health (OIE) have established the Tripartite Collaboration on AMR, which aims to foster collaboration and harmonize efforts across different sectors and regions. This collaboration has led to the development of guidelines, tools, and capacity-building initiatives to support countries in their efforts to combat AMR.The challenges posed by AMR are complex and multifaceted, requiring a sustained and concerted effort from all stakeholders. Addressing this global health crisis will require a combination of policy interventions, technological advancements, and behavioralchanges. By working together, the global community can strive to preserve the effectiveness of antimicrobial agents, protect public health, and safeguard the future of healthcare.。
避免刺猬生病英文作文
避免刺猬生病英文作文英文:As a hedgehog owner, it is important to take preventative measures to ensure that your pet does not become sick. Here are some tips to keep your hedgehog healthy:1. Keep their cage clean: Regularly clean their cage and bedding to prevent the buildup of bacteria and fungi. This can lead to respiratory infections and skin irritations.2. Provide a balanced diet: Hedgehogs require a diet high in protein and low in fat. Avoid feeding them foods that are high in sugar or salt as this can lead to obesity and other health issues.3. Monitor their weight: Weigh your hedgehog regularly to ensure that they are maintaining a healthy weight.Obesity can lead to a variety of health problems including diabetes and heart disease.4. Give them exercise: Hedgehogs are active animals and require daily exercise. Provide them with toys and a wheel to run on to keep them healthy and happy.5. Watch for signs of illness: Keep an eye out for symptoms such as lethargy, loss of appetite, and changes in behavior. If you notice any of these, take your hedgehog to the vet immediately.中文:作为一个刺猬的主人,保持你的宠物健康非常重要。
抗菌肽
抗菌肽
科技名词定义
中文名称:
抗菌肽
英文名称:
antimicrobial peptide;antibiotic peptide
定义1:一种参与固有免疫的小分子多肽,具有广谱抗菌活性,构成宿主防御细菌、真菌等入侵的重要分子屏障。
人抗菌肽有数种,以防御素最为重要。
所属学科:免疫学(一级学科);概论(二级学科);固有免疫(三级学科)
定义2:具有抗菌活性的肽类,其中研究得较多的有天蚕杀菌肽和防御肽等。
所属学科:生物化学与分子生物学(一级学科);氨基酸、多肽与蛋白质(二级学科)
水产抗菌新药抗菌肽问世
[2010-03-08] 来源于:中国化工信息网
国家海洋局第三海洋研究所采用先进分子生物学技术,克隆鱼类抗菌肽基因,构建出高效表达基因工程菌。
该所研制的重组抗菌肽市场前景广阔,有望获得我国第一个具有自主知识产权的水产抗菌新药。
目前,该所已完成基因工程抗菌肽生产,建立了基因工程抗菌肽的中试生产线,研制开发了无公害环保型水产抗菌新药。
据介绍,抗菌肽是由30~50个氨基酸组成的小分子多肽,具有杀菌力强、抗菌谱广、稳定性好、无残留、无毒副作用等优点。
开发和利用抗菌肽将为新型抗菌药物的研制提供一条新途径,也是国内外医药界关注的焦点。
- 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)。
1The equine antimicrobial peptide eCATH1 is effective against the facultative intracellular 1pathogen Rhodococcus equi in mice 23Running title: eCATH1 for treatment of Rhodococcus equi infection 45Margot Schlusselhuber 1, Riccardo Torelli 2, Cecilia Martini 2, Matthias Leippe 3, Vincent Cattoir 4,5, 6Roland Leclercq 4,5, Claire Laugier 1, Joachim Grötzinger 6, Maurizio Sanguinetti 2*, Julien 7Cauchard 1#* 891 Anses, Dozulé laboratory for equine diseases, Bacteriology & Parasitology unit, Goustranville, 10F-14430 Dozulé, France 112 Istituto di Microbiologia, Università Cattolica del Sacro Cuore, Rome, Italy 123 Department of Zoophysiology, Zoological Institute, Christian-Albrechts University of Kiel, 13Olshausenstrasse 40, 24098 Kiel, Germany 144 Caen University Hospital, Department of Clinical Microbiology, F-14033 Caen, France 155 University of Caen Basse-Normandie, Research Unit EA4655 (Team “Antibioresistance”), 16Medical School, F-14032 Caen, France 176 Institute of Biochemistry, Christian-Albrechts University of Kiel, Olshausenstrasse 40, 24098 18Kiel, Germany 1920* Co-last authors 21# Corresponding author and mailing address: 22Dr. Julien Cauchard23Copyright © 2013, American Society for Microbiology. All Rights Reserved.Antimicrob. Agents Chemother. doi:10.1128/AAC.02044-12 AAC Accepts, published online ahead of print on 1 July 20132Anses, Dozulé laboratory for equine diseases, Goustranville, F-14430 Dozulé, FRANCE. 24 Phone: +33 2 31 79 22 76 25 Fax: +33 2 31 39 21 37 26 E-mail: julien.cauchard@anses.fr273ABSTRACT 28 Rhodococcus equi, the causal agent of rhodococcosis, is a major pathogen of foals and is also 29 responsible of severe infections in immunocompromised humans. Of great concern, resistant 30 strains to currently used antibiotics have emerged. As the number of drugs that are efficient in 31 vivo is limited because of the intracellular localization of the bacterium inside macrophages, new 32 active but cell-permeant drugs will be needed in the near future. In the present study, we 33 evaluated, by in vitro and ex vivo experiments, the ability of the alpha helical equine 34 antimicrobial peptide eCATH1 to kill intracellular bacterial cells . Moreover, the therapeutic 35 potential of the peptide was assessed in an experimental rhodococcosis induced in mice while the 36 in vivo toxicity was evaluated by behavioral and histopathological analysis. The study revealed 37 that eCATH1 significantly reduced the number of bacteria inside macrophages. Furthermore, the 38 bactericidal potential of the peptide was maintained in vivo at doses that appeared to have no 39 visible deleterious effects for the mice even after 7 days of treatment. Indeed, daily subcutaneous 40 injections of 1 mg/kg body weight of eCATH1 led to a significant reduction of the bacterial load 41 in organs comparable to that obtained after treatment with 10 mg/kg body weight of rifampin. 42 Interestingly, the combination of the peptide with rifampin showed a synergistic interaction in 43 both ex vivo and in vivo experiments. These results emphasize the therapeutic potential that 44 eCATH1 represents in the treatment of rhodococcosis.454INTRODUCTION 46 Rhodococcus equi (R. equi ), previously known as Corynebacterium equi , was initially described 47 in 1923 by Magnusson in foals presenting pyogranulomatous pneumonia (1). Rhodococcosis is 48 now typically reported in foals from one to six month of age and is one of the most common 49 causes of mortality at this age (2-5). The pathogen, however, can infect a wide variety of other 50 animal species including humans. Since the first case described in 1967, the number of reports on 51 the infection have increased substantially in humans, with ~200 cases in the last three decades, 52 preferentially in persons whose immune system was compromised, either naturally, or due to 53 illness or medical treatment (6-9). 54 Many antimicrobials are effective against the extracellular pathogen in vitro . However, the 55 number of antimicrobials available for treatment of rhodococcosis is limited because of the 56 intracellular location of the bacterium. The combination of rifampin and a macrolide such as 57 erythromycin, or more recently of azithromycin and clarithromycin, is considered today as the 58 treatment of choice, as these antibiotics have the ability to penetrate abscesses and cells due their 59 lipophilic nature and to accumulate in granulocytes and macrophages to kill the bacterium (10). 60 Since the introduction of these antibiotics in rhodococcosis treatment, the survival rate of foals 61 has increased dramatically from less than 20% up to 97% (reviewed in (K. Chaffin, presented at 62 the 49th Annual Ocala Equine Conference, Ocala, FL, 21 to 24 October 2011)). Unfortunately, 63 some observations suggest that there might be a problem in the treatment of R. equi infections in 64 the near future due to the emergence of rhodococci resistant to these antibiotics and macrolides´ 65 potentially fatal side effects for horses (11-14).665With the rise of bacterial resistance to antibiotics, there is a growing interest in anti-infective 67 agents with fundamentally different mechanisms of action than those of conventional antibiotics 68 (15, 16). The concept of using antimicrobial peptides (AMPs) as therapeutic tools dates back to 69 the 1990s and they are now emerging as particularly promising candidates as new anti-infective 70 agents in the antimicrobial research area (17). In the nature, these peptides participate in the first 71 line of host defense against pathogens by combining antimicrobial activity with 72 immunomodulatory properties (18). 73 We recently reported the in vitro activity of the alpha helical equine AMP, eCATH1, against 74 antibiotic-resistant and antibiotic-susceptible strains of R. equi . The peptide proved to be 75 extracellularly active at low micromolar concentrations (0.16-1.27 µM) against the pathogen 76 independently of the antibiotic resistance profile (19) (M. Schlusselhuber, K. Guldbech, C. Sévin, 77 C. Laugier, M. Leippe, J. Grötzinger, R. Leclercq, S. Giguère and J. Cauchard, submitted for 78 publication). Aiming at extending the therapeutic potential of eCATH1 against rhodococcosis, 79 the present study was designed to analyze the efficacy of the peptide to kill intracellular R. equi 80 and to combat the pathogen in an infection-model in mice. 81 82 MATERIALS AND METHODS 83 Cell line, bacterial strain and growth conditions 84 The mouse macrophage cell line J774.2 (ECACC reference number: 85011428) purchased from 85 Sigma-Aldrich was grown in Dulbecco’s modified Eagle’s medium (DMEM; Gibco, Rockville, 86 MD, USA) supplemented with 10% (v/v) Fetal bovine serum (FBS; Pan Biotech GmbH, 87 Aidenbach, Germany ), 100 µg/ml streptomycin, 100 units/ml penicillin (complete medium) and 88 incubated at 37 °C in a humidified atmosphere containing 5% CO 2. Before assays, cell number896and viability of cells were determined by exclusion of the vital dye Trypan blue. Virulent, vapA -90 positive R. equi ATCC 33701 was used in this study. Pure strain freshly thawed was grown in 91 Brain Heart Infusion medium (BHI; BD Difco, Franklin Lakes, NJ, USA) before each experiment 92 to avoid the lost of the virulence plasmid. Cultures were incubated aerobically at 37 °C for 24 to 93 48 h. For cell line infection, bacteria were suspended with an appropriate volume of DMEM 94 supplemented with 5% FBS. The actual number of cells was confirmed by plating serial dilutions 95 on BHI agar plates. For mice inoculation, bacteria were harvested from a liquid culture by 96 centrifugation, washed, and resuspended in phosphate-buffered saline (PBS, Gibco, Rockville, 97 MD, USA) at the required bacterial concentration. 98 99 Antimicrobials used 100 eCATH1 was chemically synthesized at a purity grade of at least 96% by GenScript USA Inc. 101 (Piscataway, NJ, USA) and dissolved in 10 mM acetic acid at the final concentration of 1 mg/ml. 102 Rifampin stock solution was prepared by dissolving antibiotic powder (Ref R7382, Sigma-103 Aldrich, St. Louis, MO, USA) in methanol at a final concentration of 50 mg/kg. Stock solutions 104 of antimicrobials were stored at -20 °C until use. 105 106 Survival of R. equi in mouse macrophage cell line 107 Before the use of animals, the activity of eCATH1 against intracellular rhodococci was evaluated 108 in vitro . J774.2 murine monocyte/macrophage-like cells were washed with PBS, detached by 109 scraping and resuspended in complete medium at a final cell concentration of 5 x 105 cell/ml. 110 Monocytes were allowed to attach for 3 h on 8-wells Lab-tek chamber slide (200 µl of cell 111 suspension per well) at 37 °C in a humidified atmosphere containing 5% CO 2. Infection of1127mammalian cells was performed by washing cells with PBS before adding 200 µl of bacterial 113 suspension per well in a bacterium-to-monocyte ratio of 2:1. Chamber slides were incubated 45 114 min at 37 °C in a humidified atmosphere containing 5% CO 2 to allow serum opsonisation of 115 rhodococci. Infected monolayers were carefully washed three times with PBS to remove unbound 116 bacteria. After the last wash, 200 µl of DMEM supplemented either with eCATH1 (final 117 concentration: 20 µg/ml) or the same volume of peptide solvent (acetic acid, final concentration 118 of 0.2 mM) was added to infected cells before incubation at 37 °C in a humidified atmosphere 119 containing 5% CO 2. Viable intracellular bacteria were assessed after 24 h by staining with the 120 LIVE/DEAD Baclight bacterial viability kit (Molecular Probes Europe BV, Leiden, The 121 Netherlands) in saponin solution (final concentration of 0.05% (w/v)) to permeabilize 122 macrophages membranes as previously described (20). Slides were examined by fluorescence 123 microscopy (microscope Axioskop 40, Zeiss). The bacterial viability kit is composed of 124 propidium iodide that stains dead cells in red and Syto 9 that stains viable cells in green. 125 Therefore, nuclei of permeabilized macrophages appear red. Before the experiment, the effect of 126 20 µg/ml of eCATH1 on plasma membrane integrity of J774.2 cell line was assessed by the 127 lactate dehydrogenase (LDH) release assay after 24h of treatment. One hundred percent lysis was 128 achieved b using 1% (vol/vol) Triton X00. The LDH release assay was carried out according to 129 the manufacturer’s instructions using a commercially available kit (TOX-7, Sigma-Aldrich, Saint 130 Louis, MO, USA). 131 132 Animal studies 133 Pathogen-free BALB/c mice (Harlan Italy S.r.l., San Pietro al Natisone, Udine, Italy) were 134 housed with free access to water and standard mouse food at animal facilities of the Sacred Heart1358University (Rome, Italy). All animals used for experiments were females of 10 weeks of age 136 weighing approximately 20-25 g. For the needs of experiments, animals were sacrificed by 137 cervical dislocation. The animal experiments were performed under a protocol approved by the 138 Institutional Animal Use and Care Committee at Università Cattolica del S. Cuore, Rome, Italy 139 (Permit number: N21, 12/05/2010) and authorized by the Italian Ministry of Health, according to 140 Legislative Decree 116/92, which implemented the European Directive 86/609/EEC on 141 laboratory animal protection in Italy. Animal welfare was routinely checked by veterinarians of 142 the Service for Animal Welfare. 143 144 145 Survival of R. equi in mouse peritoneal macrophages 146 Survival of R. equi in mouse peritoneal macrophages after treatment with eCATH1, rifampin, or 147 eCATH1 combined with rifampin was assessed by using an ex vivo infection model as described 148 previously (21, 22). Briefly, mice were infected with 107 to 108 CFU of R. equi by intraperitoneal 149 injection. After an 8 h-infection period, the peritoneal macrophages were collected by peritoneal 150 lavage, centrifuged, and suspended in DMEM containing 10 mM HEPES, 2 mM glutamine, 10% 151 (v/v) FBS, and 1× non-essential amino acids supplemented with vancomycin (10 μg/ml) and 152 gentamicin (150 μg/ml). The cell suspension was dispensed into 24-well tissue culture plates and 153 incubated at 37 °C under 5% CO 2 for 2 h. After exposure to antibiotics to kill extracellular 154 bacteria (i.e., at 10 h post-infection), the infected macrophages were washed, and triplicate wells 155 of macrophages were lysed with detergent. After dilution with BHI broth, the lysates were plated 156 on BHI agar to quantify the viable intracellular bacteria. The remaining wells containing infected 157 macrophages were maintained in DMEM and treated with eCATH1 (20 µg/ml), rifampin (0.5, 51589and 10 µg/ml), or eCATH1 (20 µg/ml) combined with rifampin (5 µg/ml) for the duration of the 159 experiment. At 24 h post-infection, supernatant fluid was removed from each well, and 160 intracellular bacteria were quantified by counting on BHI agar plates. Before the experiment, 161 toxicity of eCATH1 and rifampin on mouse peritoneal macrophages was assessed with a range 162 concentration up to 80 µg/ml in order to determine the concentration of antimicrobials used in the 163 study. To assess cell viability, macrophages treated for 24 h were analyzed using the AlamarBlue 164 test (Invitrogen, Milan, Italy) following the manufacturer’s instructions. 165 166 Activity of eCATH1 against R. equi -infected mice 167 The activity of eCATH1 alone and in combination with rifampin was assessed on 168 rhodocococcosis induced in mice. Before therapeutic trials, comparison of the natural clearance 169 of R. equi CFU from organs was established in our experimental conditions to determine time 170 points of treatment and the sub-lethal dose. Briefly, ten mice per group were inoculated 171 intravenously with 107, 108 and 109 CFU of R. equi and two animals of each group were 172 sacrificed at days 1, 2, 6, 8, and 10 post-infection for bacterial count in liver, spleen, and lung. 173 The intravenous route was chosen based on Takai et al., who reported a higher virulence of the R. 174 equi ATCC 33701 strain by this route compared to the intraperitoneal route (23). All the mice 175 were monitored for survival throughout the experimental period. According to these preliminary 176 results, mice were intravenously challenged with 200 µl of a sub-lethal dose (108 CFU) of R. 177 equi . Therapeutic trials with BALB/c mice were initiated one day after bacterial inoculation to let 178 the infection develop and symptoms appear and before natural clearance of rhodococcoci from 179 mouse organs. Stock solutions of antimicrobials were appropriately freshly diluted in isotonic 180 sodium chloride before subcutaneous injection of 200 µl to animals. Infected mice were treated18110once a day with eCATH1 (1 mg/kg body weight, Group I), rifampin (10 mg/kg body weight, 182 Group II), a combination of both in the same concentrations as mentioned above (Group III), or 183 the same volume of isotonic sodium chloride (Control group) during 7 days. The doses, 184 periodicities and route of injections were chosen based on previous reports (24-26). Five mice per 185 group were sacrificed 1, 4, and 8 days post-infection (i.e. start-, mid- and end- treatment) and 186 their spleens and livers aseptically removed. Organs were weighed and separately homogenized 187 in PBS. The R. equi concentrations in organs (CFU/g organ) were determined by plating 10-fold 188 serial dilutions of the tissue homogenates on BHI agar. The plates were incubated at 37 °C for 24 189 to 48 h before bacterial enumeration. 190 191 In vivo toxicity of eCATH1 192 Non-infected mice were used to assess the in vivo toxicity of eCATH1 by behavioral and 193 histopathological analysis. Five mice was treated once a day with eCATH1 for 7 days (1 mg/kg 194 body weight) by subcutaneous injection while a second group of five not treated mice was used 195 as a control group. All the mice were monitored for survival and presence of any drug-related 196 adverse effect (local signs of inflammation, weight loss, diarrhea and behavioral alterations) 197 throughout the experimental period. Mice were sacrificed after 7 days of treatment, and organs 198 were aseptically removed (intestine, spleen, liver, lung, kidney, and stomach). Tissue samples 199 were fixed in 3.7% formaldehyde solution, processed for paraffin embedding, and stained with 200 Hematoxylin/eosin before analysis at the Veterinary Pathological Anatomy laboratory (Amboise, 201 France) by pathologists. 202 203 Statistics20411Ex vivo and in vivo experiments were performed in triplicate and duplicate, respectively, and the 205 results were subjected to statistical analysis by using one-way analysis of variance (ANOVA) 206 with a Bonferroni correction posttest with GraphPad Prism version 5.04 for Windows (GraphPad 207 Software, San Diego, CA). 208 209 RESULTS 210 Survival of R. equi in mouse macrophage cell line after addition of eCATH1 211 Before the assessment of the bactericidal activity of eCATH1 on R. equi inside J774.2 mouse 212 macrophages, the effects of the peptide on plasma membrane integrity of the host cells were 213 evaluated by the LDH release assay. At a concentration of 20 µg/ml, eCATH1 exerted no 214 detectable cytotoxic effect on J774.2 cells (data not shown). 215 The activity of eCATH1 against the intracellular pathogen was first assessed by fluorescent 216 microscopy. J774.2 macrophages were infected with R. equi for 45 min, and cells were 217 subsequently incubated for 24 h either with 20 µg/ml of eCATH1 or the peptide solvent (negative 218 control). Fluorescent staining of intracellular R. equi with the LIVE/DEAD baclight bacterial 219 viability kit revealed clear differences in rhodococcal survival inside macrophages between both 220 conditions. Indeed, very few macrophages were found to be infected in the presence of peptide 221 and those cells contained substantially lower numbers of viable bacteria (stained in green) as 222 observed in the negative control (Fig. 1). Moreover, the overall number of macrophages was 223 higher in the presence of eCATH1 compared to the non-treated sample. In the negative control, 224 no dead bacteria (stained in red) were observed, indicating that the virulent strain used in this 225 study is able to successfully survive inside macrophages (Fig. 1 c, d),. Dead rhodococci were also 226 not observed in the presence of eCATH1, but it is most likely the result of a rapid lysis of bacteria22712by the peptide. Extracellular bacteria were observed in the negative control, but not in the treated 228 sample. Before adding the peptide or its solvent, infected cells were carefully washed several 229 times to remove any non-internalized bacteria. Therefore, the presence of extracellular bacteria 230 might be either due to the necrotic death of macrophages after bacterial multiplication during 24 231 h, to the chemical action of saponin on the macrophage membrane that may release bacteria or to 232 a mechanical break of eukaryotic membranes by the positioning of the coverslip. 233 234 Survival of R. equi in mouse peritoneal macrophages 235 To confirm the in vitro study, complementary ex vivo experiments were performed. For this, 236 mouse macrophages were naturally infected in vivo and further collected for in vitro treatment 237 with eCATH1 and rifampin before CFU enumeration. As both antimicrobials exhibited a 238 synergistic interaction in vitro against extracellular R. equi (19), the combination 239 eCATH1/rifampin in the present ex vivo model was also assessed. Before the ex vivo experiment, 240 cytotoxicity of eCATH1 and rifampin on mouse peritoneal macrophages revealed that, similarly 241 to the J774.2 cell line, eCATH1 did not exhibit significant toxicity at 20 µg/ml, whereas rifampin 242 was found to exert a significant toxicity at 20 µg/ml (data not shown, ANOVA test p<0.05). 243 Concentrations of antimicrobials used in the ex vivo experiment were therefore chosen as 20 244 µg/ml for eCATH1 and 0.5, 5, and 10 µg/ml for rifampin. Supporting the in vitro data, it was 245 observed that eCATH1 resulted in a significant killing (p<0.001) of intracellular R. equi at a 246 concentration of 20 µg/ml after 14 h of treatment, i.e. ~60% decrease of R. equi in macrophages 247 compared to the control (Fig. 2). The same killing rate was found already at 5 µg/ml of rifampin. 248 Interestingly, the combination of both antimicrobials led to an even higher significant decrease of 249 the overall number of R. equi residing in macrophages (~90%, p<0.00001) (Fig. 2). However,25013from these data it was not possible to conclude whether this is a synergy or only an additive 251 effect. 252 253 Activity of eCATH1 against R. equi -infected mice 254 Before the therapeutic trials, comparison of the natural clearance of R. equi CFU from mice 255 organs for various bacterial inoculums (107 to 109 CFU) was assessed (data not shown). 256 Intravenous injection of 109 CFU of R. equi led to the death of mice after 2 to 4 days. In contrast, 257 an inoculum of 108 CFU led to the onset of symptoms from days 1 to 6 post-infection, the in vivo 258 multiplication of the pathogen, and a progressive clearance of bacteria from spleen, liver and 259 lungs after 4 days. Therefore, this sub-lethal dose was selected for the therapeutic trials. 260 Moreover, the lung was no longer used for bacterial counting in the therapeutic trials of our study 261 as a lower concentration of bacteria was found in this organ compared to spleen and liver. 262 For therapeutic trials, mice were infected with the sub-lethal dose of R. equi . One day later, 263 therapy started with daily injections of antimicrobials during 7 days. The results of CFU 264 enumeration in spleen and liver are presented in Figure 3. After three days of treatment, bacterial 265 loads were significantly decreased by comparison to the control in a similar manner in spleen and 266 liver. Indeed, 1mg/kg of body weight of eCATH1 as well as 10 mg/kg of body weight of rifampin 267 led to a decrease of ~2 log 10 CFU in both organs (i.e. >99%) compared to the control. eCATH1 268 combined with rifampin (1 mg/kg and 10 mg/kg body weight, respectively) showed the highest 269 antimicrobial activity in mice with a decrease of ~3 log 10 CFU in organs (i.e. >99.9%). 270 271 In vivo toxicity of eCATH127214Daily injection of 1 mg/kg body weight of eCATH1 during 7 days did not result in any 273 behavioral changes, differences in body weight or any clinical evidence of antimicrobials-related 274 adverse effects (data not shown). Moreover, the histopathology analysis performed on the 275 intestine, spleen, liver, lung, kidney, and stomach of treated and non-treated mice did not reveal 276 any histological changes between the groups (Fig. 4). 277 278 DISCUSSION 279 The data presented in this study represent the first demonstration of the intracellular 280 antirhodoccoccal properties of an antimicrobial peptide by application of in vitro , ex vivo and in 281 vivo methods. Indeed, the present in vitro and ex vivo studies proved that eCATH1 remained 282 active against bacteria inside the host cell although a higher concentration was needed to reach 283 the intracellular pathogen. These results support similar work in Mycobacterium species. Sharma 284 et al. demonstrated in vitro the direct bactericidal activity of the human neutrophil peptide (HNP-285 1) against intracellular Mycobacterium tuberculosis. The number of cells per macrophage was 286 reduced by 1 log 10 with 20 µg/ml and by 2 log 10 with 40 µg/ml of peptide after three days of 287 treatment (27). More recently, Jena et al., reported the activity of NK-2, a shortened peptide 288 derived from the porcine NK-lysin, against intracellular Mycobacterium smegmatis. This study 289 revealed that 10 µM of peptide (~30 µg/ml) decreased the number of bacteria per macrophage by 290 ~40% after 8 h of treatment (28). 291 From the mouse rhodococcosis model, the main conclusion is that eCATH1 remained effective 292 against the facultative intracellular pathogen.. Interestingly, the peptide appeared to be more 293 active in vivo than in vitro in comparison to rifampin activity. A four-fold-lower concentration of 294 rifampin led to a similar decrease of rhodococci per macrophage than eCATH1 in vitro , while, in29515vivo , a ten-fold-higher concentration of rifampin than the peptide is required to equally decrease 296 the bacterial load in organs. Previously, similar data were reported by Sharma et al., (26). As 297 little as 1-5 µg of the HNP-1 per mouse was found to decrease tuberculosis infection while 40 298 µg/ml of peptide was needed to kill intramacrophage mycobacteria in ex vivo experiments (26, 299 27). These observations might indicate that eCATH1 has a more complex effect in vivo than in 300 vitro . As previously raised by Sharma et al., we hypothesize that the in vivo activity of eCATH1 301 is a combination of i) immunomodulatory properties leading to an indirect killing of bacteria and 302 ii) a direct killing effect on intra- and extracellular bacteria. Some reports in the literature about 303 immunomodulatory properties of human and bovine cathelicidins could support our hypothesis. 304 The human cathelicidin LL-37 exerts a chemoattractive activity on immune cells through direct 305 interaction with granulocytes and mononuclear cells receptors and through induction of 306 chemokines production which hypothetically increase the numbers of neutrophils and monocytes 307 at sites of infection (29-33). Interestingly, some bovine cathelicidins were also found to be 308 chemoattractants for neutrophils and had the property of enhancement of phagocytic and 309 degranulation activities of these cells (B. W. Paget, J. L. Harper and B. J. Haigh, presented at the 310 3rd Antimicrobial peptide symposium, Lille, France, 13 to 15 June 2012). Moreover, 311 macrophages have the ability to uptake granules of apoptotic neutrophils to acquire AMPs that 312 traffic via early endosomes to intracellular pathogens-containing vacuoles where they exert their 313 bactericidal activity (34). Chemoattraction of immune cells such as neutrophils by eCATH1 314 might limit the spread of R. equi infection in the host in a similar way. Furthermore, this 315 hypothesis would be in accordance with the observation that neutrophils are critical for the 316 control of rhodocococcosis (35). Nevertheless, the immunomodulatory properties of eCATH1 317 remain to be elucidated to an better understanding of the in vivo activities of this peptide.31816The mechanism by which exogenously applied AMPs might access to intracellular bacteria is not 319 fully understood. Some clues, however, were first given by Sharma et al., in 2000 (27). The 320 studied peptide was initially found to localize at the macrophage membrane and then to be taken 321 up inside the cell (27). In a similar pathway to that of neutrophil’s AMPs uptake by macrophages 322 discussed above (34), eCATH1 might be internalized by macrophages in early endosomes that 323 would further fuse with R. equi-containing vacuoles, where the peptide could exert its 324 bactericidal activity. 325 Interestingly, the slight positive interaction between eCATH1 and rifampin against extracellular 326 rhodococci described previously, was observed against the facultative intracellular bacterium in 327 the in vivo setting as well (19). Our observations are consistent with the work of Cirioni et al., 328 who similarly reported a synergistic effect between rifampin and α-helical AMPs (magainin II 329 and cecropin A) in Pseudomonas aeruginosa infection models in rats (25). It was hypothesized 330 that AMPs allow rifampin, a RNA polymerase inhibitor, to access its intracellular target by 331 permeabilization of the bacterial membrane (19, 36). 332 In vivo degradation by proteases, inactivation by physiological salt concentration and rapid 333 clearance by kidneys and toxicity are often described in the literature as major limiting factors 334 that hamper the development of AMPs as therapeutic options. In the present study we proved that 335 the bactericidal activity of eCATH1 was conserved in vivo at doses that might be compatible with 336 clinical use, without detectable deleterious effects for the host even after 7 days of daily 337 subcutaneous injections. The results of the in vivo toxicity and activity presented here agree with 338 our previous investigations of eCATH1. Indeed, we recently reported that the in vitro activity of 339 eCATH1 was not hampered by physiological salt concentration and that the peptide was not toxic 340 for various mammalian cell types in vitro (19).341。