Apolipoprotein E modulates
- 格式:pdf
- 大小:229.00 KB
- 文档页数:8
载脂蛋白E在血栓相关性疾病及恶性肿瘤中的研究进展郭盛虎; 史健【期刊名称】《《中国全科医学》》【年(卷),期】2013(016)005【总页数】3页(P590-592)【关键词】载脂蛋白E; 冠心病; 阿尔茨海默病; 糖尿病肾病; 恶性肿瘤【作者】郭盛虎; 史健【作者单位】050000河北省石家庄市河北医科大学第四医院肿瘤内科【正文语种】中文【中图分类】R341.35载脂蛋白E(ApoE)是正常人血浆脂蛋白重要成分,在参与脂质的转运、储存、利用及排泄中发挥重要作用[1]。
ApoE是1973年Shore等在正常人的极低密度脂蛋白(VLDL)中首次发现的,其基因定位于人类第19对染色体长臂上,长37 kb,该基因包含4个外显子和3个内含子。
其cDNA长为1.63 kb,基因的最初产物为含317个氨基酸的蛋白质,被一个含18个氨基酸的信号肽裂解后,变成由299个氨基酸组成的成熟蛋白。
在血浆中存在3种主要异构体(E2、E3、E4),由位于同一基因位点上的3个等位基因(ε2、ε3、ε4)所编码,构成3种纯合子(E2/2、E3/3、E4/4),3种杂合子(E3/2、E4/3、E4/2)[2]。
ApoE存在于多种脂蛋白颗粒中,不同于其他载脂蛋白的是,其他载脂蛋白多由肝脏合成,而ApoE在肝、脑、肾、脾、表皮、子宫中都有合成。
ApoE是正常人血浆脂蛋白中重要的成分,在参与脂质的转运、储存、利用及排泄中发挥重要作用。
同时ApoE作为乳糜微粒(CM)、VLDL、高密度脂蛋白(HDL)的重要组分,有利于这些脂蛋白(LP)的结构稳定,并通过与低密度脂蛋白受体(LDL- R)和肝 ApoE受体结合,促进CM、低密度脂蛋白(LDL)、VLDL的降解,且有转运和调节脂类代谢的作用,特别是与机体总胆固醇(TC)的代谢密切相关[3]。
近年来有关ApoE与恶性肿瘤关系的研究也备受关注。
有研究表明ApoE与LDL-P的结合可以诱发细胞信号转导的变化,从而调节细胞的生长[4]。
17-Estradiol promotes the up-regulation of SR-BII in HepG2 cells and in rat liversGregory A. Graf, Kathy L. Roswell, and Eric J. Smart1University of Kentucky Medical School, Department of Physiology, Lexington, KY 40536Abstract The scavenger receptor class B type I (SR-BI) binds to HDL and mediates the selective uptake of cholesterol esters from HDL to cells. SR-BII is an alternatively spliced product of the SR-BI gene that only differs in the C-terminal cytoplasmic domain. Previous studies with male mice demonstrated that SR-BII comprises about 12% of the total SR-BI/SR-BII present in liver. In the current studies we used a liver cell line, HepG2, and a rat estrogen replacement model to examine the effects of estrogen on the expression of SR-BII. HepG2 cells express SR-BI but not SR-BII. SR-BI/SR-BII-blocking antibodies dem-onstrated that HepG2 cells selectively internalize cholesterol esters in a SR-BI-dependent manner. Incubation of HepG2 cells with 10 pM of 17-estradiol for 12 h eliminated the ex-pression of SR-BI and promoted the up-regulation of SR-BII. Radiolabeled HDL-binding studies demonstrated that 17-estradiol increased the number of HDL binding sites by 3-fold in HepG2 cells. However, 17-estradiol-treated cell internal-ized approximately 25% less cholesterol ester than vehicle-only-treated cells. The livers obtained from male rats and ovariec-tomized female rats contained SR-BI and a small amount of SR-BII. In contrast, the livers obtained from intact female rats and ovariectomized female rats receiving estrogen replace-ment contained SR-BII and a small amount of SR-BI. The amount of SR-BI and SR-BII in adrenal tissue was not affected by any of the experimental treatments. We conclude that es-trogen up-regulates SR-BII in HepG2 cells and rat liver.—Graf G. A., K. L. Roswell, and E. J. Smart. 17-Estradiol promotes the up-regulation of SR-BII in HepG2 cells and in rat livers.J. Lipid Res.2001. 42: 1444–1449.Supplementary key words HDL • cholesterol ester • scavenger receptor •selective uptakeThe scavenger receptor class B type I (SR-BI) is a physio-logical HDL receptor involved in the selective uptake of HDL cholesterol ester (1). Selective uptake refers to the internalization of HDL cholesterol esters without the in-ternalization of HDL apoproteins (2, 3). Webb et al. (4, 5) described a splice variant of SR-BI called SR-BII that is ex-pressed in rodents and humans. The amino acid se-quences of SR-BI and SR-BII are identical except for the C-terminal cytoplasmic tail (4, 5). Despite the remarkable amino acid similarity to SR-BI, SR-BII is approximately 4-fold less efficient at selective cholesterol ester uptake than SR-BI. SR-BII accounts for only about 12% of the total SR-BI/SR-BII found in mouse liver even though the mRNA for SR-BII comprises about 40% of the total SR-BI/SR-BII mRNA in liver (4, 5). It is unclear if SR-BII plays a signifi-cant role in selective cholesterol ester uptake in vivo.The expression of SR-BI appears to be coordinately reg-ulated with steroidgenesis in the adrenal gland, ovary, and testis. In mice, adrenocorticotropic hormone increases SR-BI expression, whereas glucocorticoids suppress ex-pression in the adrenal cortex (6). In rats, luteinization of the ovary with human chorionic gondadotropin or treat-ment of granulosa cells with follicle-stimulating hormone induces expression of SR-BI and uptake of HDL choles-terol esters (7). Similarly, human chorionic gondadotro-pin stimulated SR-BI expression and uptake of DiI-labeled HDL in the testis (8). It is interesting to note that the humanSR-BI promoter contains the recognition sequence (Ad4BP)for the orphan receptor steroidogenic factor-1, which induces the expression of several genes involved in ste-roidogeneis and modulates SR-BI promoter activity in adrenal cells (9).Supraphysiological doses of a synthetic estrogen, 17␣-ethinyl estradiol, decrease hepatic expression of SR-BI in rats, suggesting a role for estrogens in modulating SR-BI expression (8). However, high-dose estrogen also in-creases hepatic LDL-receptor activity (10, 11). Therefore, decreased hepatic expression of SR-BI may have been due to elevated cholesterol content, rather than a direct effect of estrogen. Alternatively, Landschulz et al. (8) noted that hepatic and adrenal expression of SR-BI were lower in fe-male rats relative to males, suggesting that endogenous estrogens may also modulate SR-BI.Little is known concerning the hormonal regulation of SR-BII. The goal of this study was to determine if physio-logical levels of 17-estradiol modulated the levels of SR-BII. We found that 17-estradiol promoted the down-regulation of SR-BI and the up-regulation of SR-BII in HepG2 cells and in the liver of a rat estrogen replacement model. The in vivo consequences of this switch between receptors will require additional studies.Abbreviations: SR-BI/SR-BII, scavenger receptor class B type I/type II.1To whom correspondence should be addressed.e-mail: ejsmart@ by on May 16, 2007 Downloaded from1444Journal of Lipid Research Volume 42, 2001MATERIALS AND METHODSMaterialsMEM Eagles with Earle salts (phenol red minus), fetal bovineserum (non-heat inactive), l-glutamine, trypsin-EDTA, and penicillin/streptomycin were from Life Technologies, Inc.(Grand Island, NY). Percoll, 17-estradiol, PVDF membrane, and Tween 20 were from Sigma (St. Louis, MO). BioRad Dcassay was from BioRad (Hercules, CA). Rabbit serum directedagainst SR-BI and SR-BII were a generous gift from Dr. Deneysvan der Westhuyzen (UK). The SR-BI/SR-BII-blocking antibodywas generated by Quality Control Biochemicals. Horseradishperoxidase conjugated IgGs were supplied by Cappel (WestChester, PA). Super Signal® chemiluminescent substrate waspurchased from Pierce (Rockford, IL). 1␣, 2␣ (n) [3H]choles-teryl-oleate (37 Ci/mmol) was supplied by Amersham (Arling-ton Heights, IL). 125I-Na (1 mCi/ml) was purchased from NEN (Boston, MA).BuffersLysis buffer consisted of 25 mM MES (pH 6.5), 0.15 M NaCl,1% (v/v) Triton X-100, and 60 mM octylglucoside. Samplebuffer (5ϫ) consisted of 0.31 M Tris (pH 6.8), 2.5% (w/v) SDS, 50% (v/v) glycerol, and 0.125% (w/v) bromophenol blue. Tris-buffered saline (TBS) consisted of 20 mM Tris (pH 7.6) and 137 mM NaCl. Blotting buffer consisted of TBS plus 0.5% Tween 20 and 5% dry milk. Wash buffer consisted of TBS plus 0.5% Tween 20 and 0.2% dry milk. Tris-saline consisted of 50 mM Tris (pH 7.4) and 150 mM NaCl.Cell cultureHepG2 cells were cultured in a monolayer and plated in a stan-dard format. On day 0, approximately 5 ϫ 105 cells were seeded into 100-mm dishes with 10 ml of MEM eagles (phenol red minus) with Earle salts supplemented with 100 unit/ml penicillin/ streptomycin, 0.5% (v/v) l-glutamine, and 10% (v/v) fetal bovine serum (non-heat inactive). The cells were used on day 4. Isolation and labeling of lipoproteinsHDL (dϭ1.063–1.21 g/ml) was isolated from fresh human plasma by density gradient ultracentrifugation as described pre-viously (12). The HDL3 subfraction (dϭ1.13–1.18 g/ml) was iso-lated from other HDL subfractions by ultracentrifugation. HDL3 apolipoproteins were then iodinated with iodine monochloride (13) to a specific radioactivity of 400–600 cpm/ng protein. 1␣,2␣(n) [3H]cholesteryl-oleate was then incorporated into 125I-labeled HDL3 as described previously (14). The specific radio-activity of [3H]cholesterol ester in the dually labeled particles ranged from 32–35 dpm/ng cholesterol.Ligand binding and uptake assaysThe selective uptake of cholesterol esters from HDL into cellswas determined using dual-labeled HDL3 as described previously(15). Cells were grown to 90% confluency in 12 well plates, thenrinsed twice with PBS (37ЊC for uptake assays and 4ЊC for bind-ing assays). Medium containing 5% human lipoprotein-deficient serum and 10 g/ml 125I-[3H]cholesteryl-oleate-HDL was added to the cells for the indicated intervals. Following the incubation, uptake was terminated by aspirating the medium and washing the cell monolayers four times with Tris-saline (4ЊC). Cholesterol ester was extracted from the cells with 3:2 hexane–isopropanol (v/v), and the cells were solubilized in 0.6 ml of 0.5 M NaOH. The total HDL3 associated with the cells was determined by counting aliquots of the 0.5 M NaOH homogenate. The total cholesterol ester associated with the cells was determined by liquid scintilla-tion counting of the organic extracts. The amount of HDL3degraded was determined by measuring the amount of non-TCA precipitatable 125I in the cell medium. To compare cell-associated125I-HDL with [3H]cholesterol ester HDL, [3H]cholesterol esteruptake was expressed as apparent HDL protein uptake, assumingthat uptake resulted from whole HDL particle uptake as describedby Knecht and Pittman (16).Animals and treatmentsFemale Sprague-Dawley rats were housed on a 14:10 hlight:dark cycle with unrestricted access to food and water. Ratswere bilaterally ovariectomized under isoflurane anesthesia at 8 weeks of age. For 17-estradiol studies, Silastic capsules (3.8 mm diameter, 30 mm length) containing either sesame oil (control)or 17-estradiol in sesame oil (1 mg/ml) were implanted subcu-taneously at the time of surgery as previously described (17). Im-plants were replaced after 7 days and rats were killed after 14days of treatment. Organs were harvested, snap frozen on dryice, and stored at Ϫ80ЊC. Trunk blood was collected and serum harvested for determination of total cholesterol and fraction-ation by FPLC as described previously (18). Estradiol concentra-tions were determined by radioimmunoassay as described (19).All surgical procedures were approved by the University of Kentucky Institutional Animal Care and Use Committee.SDS-PAGE and immunoblottingTissues (0.1 g) were homogenized in 1.0 ml of lysis buffer witha Teflon-coated homogenizer on ice. Homogenates were incu-bated on ice for 20 min and centrifuged at 12,000 g for 15 min at4ЊC. Supernatants were collected and protein concentration de-termined by Dc assay. Proteins were diluted in 1ϫ sample bufferplus 1.2% (v/v) -mercaptoethanol and heated to 95ЊC for 5min immediately prior to loading. Proteins were separated on a12.5% polyacrylamide gel at 50 mA (constant current) and sub-sequently transferred to PVDF membrane at 50 V (constant volt-age) for 2 h. Membranes were blocked with blotting buffer for60 min at 22ЊC. Primary antibodies were diluted in blottingbuffer and incubated with blocked membranes for 60 min at22ЊC. Membranes were washed four times for 10 min in wash buffer. Horseradish peroxidase conjugated IgGs directed againstthe appropriate host IgGs were diluted and incubated with mem-branes as described for primary antibodies. Membranes were washed four times for 10 min in wash buffer and visualized using chemoluminescence. The relative signal intensities were deter-mined by densitometry. Increasing amounts of a single samplewere analyzed to verify that signal intensity varied linearly withprotein loading.Nuclease protection assayRat liver total RNA was used to reverse transcribe 120 nucleo-tide fragments corresponding to the C-terminal regions of SR-BIor SR-BII. The C-terminal regions of SR-BI and SR-BII are unique. An RNA Polymerase Promoter Addition Kit, Ribonu-clease Protection Assay Kit, and Nonisotopic Detection Kit from Ambion, Inc., were used to conduct the nuclease protection assay. The assay was performed according to the manufacturer’s instructions.RESULTS17-Estradiol alters the expression ofSR-BI and SR-BII in HepG2 cellsWe first determined if HepG2 cells contained SR-BI and/or SR-BII by immunoblotting cell lysates with SR-BI andSR-BII specific antibodies (4). Figure 1 demonstrates thatby on May 16, 2007Downloaded fromGraf, Roswell, and Smart17-Estradiol regulation of SR-BII14451446Journal of Lipid Research Volume 42, 2001HepG2 cells contained immunodetectable levels of SR-BI but not SR-BII. However, SR-BI and SR-BII were detected in cell lysates prepared from CHO cells transfected with either SR-BI or SR-BII.We next determined if HepG2 cells were capable of SR-BI-dependent selective cholesterol ester uptake. HDL par-ticles, dually labeled with [ 3 H]cholesterol ester and 125 I-apoprotein, were incubated with HepG2 cells ( Fig. 2 ) orwith HepG2 cells in the presence of 50 g/ml of SR-BI/SR-BII blocking antibody (Fig. 2) for the indicated times and the amount of selective cholesterol ester uptake de-termined. HepG2 cells selectively internalized a relatively large amount of cholesterol ester. The selective uptake of cholesterol ester was abolished in the presence of a SR-BI/SR-BII blocking antibody. Non-specific IgG did not affect the selective uptake of cholesterol ester (data not shown). Less than 0.3% of the total added radio-labeled HDL particles were degraded during the time course of the experiment.To determine the effect of 17  -estradiol on SR-BI and SR-BII expression, HepG2 cells were cultured in 10 pM 17  -estradiol for the indicated times (phenol red minus medium). At the end of the incubation, cell lysates were generated, resolved by SDS-PAGE and immunoblotted with SR-BI- and SR-BII-specific antibodies ( Fig. 3 ). 17  -Estradiol induced a time-dependent decrease in SR-BI levels and increase in SR-BII levels. SR-BI was completely down-regulated by 12 h and SR-BII was maximally up-regulated by 20 h of 17  -estradiol treatment. Quantification mea-surements done with an antibody that recognizes both SR-BI and SR-BII (4) demonstrated that SR-BII was increased 3.2-fold more than the starting level of SR-BI (data not shown).We next determined if the alterations in SR-BI and SR-BII levels affected the ability of HDL to associate with HepG2cells. Cells were incubated with 10 pM 17  -estradiol for the indicated times. The cells were then chilled to 4 Њ C and incubated with 125 I-HDL for 60 min. The cells were washed and processed to determine the extent of 125 I-HDL cell association and degradation. Figure 4A demonstrates that the amount of associated 125 I-HDL declines for the first 8 h of 17  -estradiol treatment. After 20 h of 17  -estradiol treatment the extent of 125 I-HDL that associated with the cells was approximately 3-fold greater than un-treated cells. Less than 0.3% of the total added radio-labeled HDL particles were degraded during the course of the experiment.To determine if alterations in SR-BI and SR-BII levels af-fected the ability of HepG2 cells to take up cholesterol ester selectively, cells were incubated with 10 pM 17 -estradiol forFig.1.HepG2 cells express SR-BI but not SR-BII. HepG2 cells were cultured for four days, then collected, and a cell lysate gener-ated. 20 g of protein was resolved by SDS-PAGE and transferred to nylon. As controls for SR-BI and SR-BII, 1 g of cell lysate protein generated from CHO cells transfected with SR-BI or transfected with SR-BII were also resolved by SDS-PAGE and transferred to nylon. SR-BI and SR-BII specific antiserum (4) were used to probe the membranes. The immunoblots were developed by the method of chemoluminescence (30 s exposures). Longer exposure times (20 min) did not allow the detection of SR-BII in HepG2 cells (data not shown). The data are representative of three independentexperiments.Fig.2.HepG2 cells selectively internalize cholesterol ester in a SR-BI-dependent manner. HepG2 cells (᭹) or HepG2 cells plus 50 g/ml of SR-BI/SR-BII blocking antibody () were incubated with 10 g/ml of 125I and [3H]cholesterol ester labeled HDL for the indicated times. The cells were then washed and the amount of cell associated 125I-HDL determined with a gamma counter, and the amount of [3H]cholesterol ester quantified with a beta counter. Se-lective uptake was determined by subtracting the values obtained for 125I from the values obtained for the [3H]cholesterol ester (CE). Less than 0.3% of the total added radiolabeled HDL particles were degraded during the course of the experiment. The data are from four independent experiments, mean Ϯ S.E.M, n ϭ3.Fig.3.17-Estradiol down-regulates SR-BI and up-regulates SR-BII in HepG2 cells. HepG2 cells were cultured for four days and then incubated for the indicated times (hours) with 10 pM 17-estradiol. At the end of the incubation, cell lysates were generated and 20 g of protein was resolved by SDS-PAGE, transferred to ny-lon and immunoblotted with SR-BI and SR-BII antiserum. The im-munoblots were developed by the method of chemoluminescence (30 s exposures). Longer exposure times (20 min) did not signifi-cantly alter the data (data not shown). The data are representative of five independent experiments.by on May 16, 2007Downloaded fromGraf, Roswell, and Smart 17  -Estradiol regulation of SR-BII 1447the indicated times (Fig. 4B). Three hours before each mea-surement, 10 g/ml of dually radiolabeled HDL was added to the cells. At the indicated times the cells were processed to determine the extent of selective cholesterol ester uptake.The amount of selective cholesterol ester internalized ini-tially decreased with minimal uptake occurring at 9 h of 17  -estradiol treatment. After 24 h of 17  -estradiol treatment the amount of cholesterol ester internalized was approximately 75% of that obtained in the absence of 17  -estradiol.The effect of 17  -estradiol on the expression of SR-BI and SR-BII in rat liverTo determine if 17  -estradiol influences the expression of SR-BII in vivo, we used an established rat estrogen re-placement model (17). Twelve rats were ovariectomized on day 0 of the study and immediately given implants con-taining either 17 -estradiol in sesame oil or vehicle only.Proestrus estradiol concentrations in intact cycling rats range from 40 to 80 pg/ml (20). The mean serum estra-diol concentration in the ovariectomized rats was 6.9 Ϯ0.5 pg/ml. Estrogen replacement increased estradiol con-centrations to 97.7 Ϯ 7.4 pg/ml. Four male rats and four intact cycling female rats were also studied for compari-son. On day 14 the animals were sacrificed, and the livers and the adrenal glands were harvested and immunoblot-ted with SR-BI- and SR-BII-specific antibodies. Figure 5demonstrates that none of the treatments altered the amount of SR-BI and SR-BII associated with adrenal glands. However, the experimental groups had different amounts of SR-BI and SR-BII associated with the livers.Livers from male rats and ovariectomized female rats con-tained predominantly SR-BI and a small amount of SR-BII. In contrast, livers from intact cycling female rats and ovariectomized rats receiving estrogen replacement had significantly less SR-BI and a dramatic increase in the amount of SR-BII. Because different antibodies were used to detect SR-BI and SR-BII, it is not possible to compare the relative mass amounts of SR-BI to SR-BII in Fig. 5.To determine if the changes in hepatic SR-BI and SR-BII protein levels correlated with changes in mRNA levels,total RNA was isolated from hepatic tissues and the rela-tive levels of SR-BI and SR-BII message quantified by a ri-bonuclease protection assay (Fig. 6). C-terminal specific nucleotide fragments were used so that SR-BI and SR-BIIcould be distinguished. In parallel, expression of GAPDHFig.4.The effects of 17-estradiol on HDL cell association and selective cholesterol ester uptake. HepG2 cells were cultured for four days and then incubated for the indicated times with 10 pM 17-estradiol. A: At the indicated times of 17-estradiol treatment,the cells were chilled to 4ЊC and then incubated with 10 g/ml of dually radiolabeled HDL for 1 h. The cells were washed and pro-cessed to determine the extent of HDL cell association and degra-dation. Radiolabeled HDL-binding was prevented with 50-fold ex-cess unlabeled HDL or in the presence of 50 g/ml blocking antibody (data not shown). The data are from six independent ex-periments, mean Ϯ S.E.M, n ϭ 3. B: Three hours before the indi-cated times of 17-estradiol treatment, 10 g/ml of dually radio-labeled HDL was added (2, 6, 10, 14, 21 h respectively). At the indicated times of 17-estradiol treatment, the cells were then washed and the amount of cell associated 125I-HDL determined with a gamma counter, and the amount of [3H]cholesterol ester quantified with a beta counter. Selective uptake was determined by subtracting the values obtained for 125I from the values obtained for the [3H]cholesterol ester (CE). Less than 0.3% of the total added radiolabeled HDL particles were degraded during the course of the experiment. Selective cholesterol ester uptake was inhibited with 50-fold excess unlabeled HDL or in the presence of 50 g/ml blocking antibody (data not shown). The data are from six inde-pendent experiments, means Ϯ S.E.M, n ϭ3.Fig.5.Effects of ovariectomy and ovariectomy with estrogen re-placement on the levels of liver SR-BI and SR-BII protein. Livers and adrenals from male rats, intact cycling female rats, and ovari-ectomized rats receiving a subcutaneous Silastic implant containing sesame oil or 17-estradiol (1 mg/ml) for 14 days were harvested.Cell lysates were generated and resolved by SDS-PAGE, transferred to nylon, and immunoblotted with SR-BI and SR-BII specific anti-serum. 45 g of liver lysate and 10 g of adrenal lysate were used.The immunoblots were developed by the method of chemolumi-nescence (60-sec exposures). Data from one animal in each experi-mental group is shown. All of the animals had nearly identical SR-BI and SR-BII profiles. Ovx, ovariectomy; E, 17-estradiol.by on May 16, 2007Downloaded from1448Journal of Lipid Research Volume 42, 2001was determined to validate equality of RNA loading. The relative abundance of SR-BI and SR-BII mRNA did not change in the presence of 17-estradiol.DISCUSSIONSR-BI and SR-BII are two isoforms of a class B scavenger receptor that differ only in the C-terminal cytoplasmic tail (4, 5). Estrogen has been reported to induce the down-regulation of hepatic SR-BI (8, 21). In the present study we demonstrated that as little as 10 pM of 17-estradiol could induce the complete loss of SR-BI and the dramatic up-regulation of SR-BII in HepG2 cells, a liver-derived cell line. Significantly, the amount of SR-BI and SR-BII corre-lated with the extent of 125I-HDL that associated with the cells. The 17-estradiol-induced switch from SR-BI to SR-BII did not inhibit selective cholesterol ester uptake al-though uptake did decrease by approximately 25%. Simi-lar to other studies we observed less SR-BI in intact cycling female rats compared with male rats (8). However, we re-port for the first time that intact cycling female rats contain considerably more SR-BII than male rats. Ovariectomized female rats contained as much SR-BI as male rats and down-regulated SR-BII to levels comparable to male rats.Ovariectomized female rats receiving estrogen replace-ment contained SR-BI and SR-BII protein levels compara-ble to intact cycling females.The functions of SR-BI and SR-BII have been studied in transfected cell lines. SR-BI and SR-BII both bind HDL with similar affinities and both receptors mediate the se-lective uptake of cholesterol ester from HDL (4). How-ever, SR-BII is much less efficient at selectively internaliz-ing HDL-derived cholesterol ester (4). Webb et al. (4)have estimated that SR-BII is approximately 4-fold less effi-cient at selective uptake than SR-BI. This estimate of SR-BII efficiency matches well with our current data. If SR-BII is 4-fold less efficient than SR-BI than a 4-fold increase in SR-BII would be required to obtained the same efficiency of selective cholesterol ester uptake. The final amount of 17-estradiol induced SR-BII up-regulation in HepG2cells was about 3-fold more than the starting SR-BI levels.Significantly, the amount of HDL-derived cholesterol ester internalized in 3 h was 75% of that obtained by SR-BI.The mechanism by which class B scavenger receptors mediate the selective uptake of HDL-derived cholesterol ester is not understood. SR-BI and SR-BII are identical at the amino acid level with the exception of the C-terminal cytosolic tail. Both proteins are enriched in caveolae in transfected CHO cells (4, 22) and both proteins are acylated (4, 22). The extracellular domain and C-terminal cytosolic tail of SR-BI have been postulated to be critical domains involved in selective uptake (23). How the C-terminal cytosolic tail of SR-BII decreases the efficiency of selective cholesterol ester uptake is not known. One possibility is that the cytoplasmic tail of SR-BII interacts with a cellular protein that regulates selective uptake. This speculative mechanism has not been tested.We have demonstrated that 17-estradiol induces the loss of SR-BI and the accumulation of SR-BII in HepG2cells. These observations confirm previous reports of an estrogen-induced decline in hepatic SR-BI expression and are the first to demonstrate an estrogen-induced up-regulation of SR-BII. The mechanism of how 17-estradiol causes a switch from SR-BI to SR-BII is unclear. SR-BI and SR-BII are encoded by the same gene (5). The precursor mRNA is alternatively spliced such that a 129-nucleotide exon is either included or removed to yield SR-BI or SR-BII transcripts (5). Previous studies with adrenocorticotropic hormone demonstrated that this hormone did not regulate alternative splicing. The data presented here suggest that 17-estradiol may regulate the splicing of the SR-BI/SR-BII transcript, however, RNase protection experiments did not detect significant changes in the amount of SR-BI and SR-BII specific transcripts. Alternatively, 17-estradiol may affect the stability of the proteins. Both apoAI and apoE can be post-transcriptionally modified in an estrogen-dependent process (24). Indeed, apoE mRNA shifts to the fast translating poly-somal pool in response to estrogen treatment (25).In conclusion, the data demonstrate that 17-estradiol can induce the down-regulation of SR-BI and the up-regulation of SR-BII in HepG2 cells. Despite the lower efficiency of SR-BII in selective cholesterol ester uptake, the large in-crease in SR-BII allowed the cells to internalize about 75%of the cholesterol ester that SR-BI internalized in 3 h. We have provided data demonstrating that in a rat estrogen replacement model SR-BII is up-regulated and SR-BI is down-regulated in the liver. This in vivo model will allowfuture studies to examine the impact of 17-estradiol on the metabolism of HDL.This work was supported by NIH grants HL62844, HL58475,HL64056 (EJ S), and P20RR15592 (Wise). We thank Dr. WiseFig.6.Effects of ovariectomy and ovariectomy with estrogen re-placement on the levels of liver SR-BI and SR-BII mRNA. Fifty mi-crograms of total RNA from male rats (Male), intact cycling female rats (Intact), and ovariectomized rats receiving a subcutaneous Si-lastic implant containing sesame oil (Ovx) or 17-estradiol (Ovx ϩE 2) (1 mg/ml) for 14 days were hybridized with an antisense 120nucleotide fragment of SR-BI or SR-BII and digested with RNAse A/T1. Adrenal RNA (Ad) (25 g) was used as a positive control for SR-BI and SR-BII. The antisense fragment of SR-BI or SR-BII in the absence of cellular RNA was also included with (ϩ) or without (Ϫ)treatment with RNAse A/T1. In parallel, levels of GADPH mRNA were determined to verify equal loading.by on May 16, 2007 Downloaded fromand the Atherosclerosis Research Group for invaluable advice and assistance.Manuscript received 20 February 2001 and in revised form 1 May 2001.REFERENCES1.Trigatti, B., A. Rigotti, and M. Krieger. 2000. The role of the high-density lipoprotein receptor SR-BI in cholesterol metabolism. Curr Opin Cell Biol.11: 123–131.2.Glass, C., R. C. Pittman, M. Civen, and D. Steinberg. 1985. Uptakeof high-density lipoprotein-associated apoprotein A-I and choles-terol esters by 16 tissues of the rat in vivo and by adrenal cells and hepatocytes in vitro. J. Biol. Chem.260: 744–750.3.Johnson, W. J., F. H. Mahlberg, G. H. Rothblat, and M. C. Phillips.1991. Cholesterol transport between cells and high-density lipo-proteins. Biochim. Biophys. Acta.1085: 273–298.4.Webb, N. R., P. M. Connell, G. A. Graf, E. J. Smart, W. J. de Villiers,F. C. de Beer, and D. R. van der Westhuyzen. 1998. SR-BII, an iso-form of the scavenger receptor BI containing an alternate cyto-plasmic tail, mediates lipid transfer between high density lipopro-tein and cells. J. Biol. Chem.273: 15241–15248.5.Webb, N. R., W. J. S. de Villiers, P. M. Connell, F. C. de Beer, andD. R. van der Westhuyzen. 1997. Alternative forms of the scavengerreceptor BI (SR-BI). J. Lipid Res.38: 1490–1495.6.Rigotti, A., E. R. Edelman, P. Seifert, S. N. Iqbal, R. B. DeMattos,R. E. Temel, M. Krieger, and D. L. Williams. 1996. Regulation by adrenocorticotropic hormone of the in vivo expression of scaven-ger receptor class B type I (SR-B1), a high density lipoprotein re-ceptor, in steroidogenic cells of the murine adrenal gland. J. Biol.Chem.271: 33545–33549.7.Reaven, E., A. Nomoto, S. Leers-Sucheta, R. Temel, D. L. Williams,and S. Azhar. 1998. Expression and microvillar localization of scav-enger receptor, class B, type I (a high density lipoprotein recep-tor) in luteinized and hormone-desensitized rat ovarian models.Endocrinology.139: 2847–2856.ndschulz, K., R. K. Pathak, A. Rigotti, M. Krieger, and H. H.Hobbs. 1996. Regulation of scavenger receptor, class B, type 1, a high density lipoprotein receptor, in liver and steroidogenic tis-sues of the rat. J. Clin. Invest.98: 984–995.9.Cao, G., C. K. Garcia, K. L. Wyne, R. A. Schultz, K. L. Parker, andH. H. Hobbs. 1997. Structure and localization of the human geneencoding SR-BI/CLA-1. J. Biol. Chem.272: 33068–33076.10.Windler, E. E., P. T. Kovanen, Y. S. Chao, M. S. Brown, R. J. Havel,and J. L. Goldstein. 1980. The estradiol-stimulated lipoprotein re-ceptor of rat liver. J. Biol. Chem.255: 10464–10471.11.Kovanen, P. T., M. S. Brown, and J. L. Goldstein. 1979. Increased bind-ing of low density lipoprotein to liver membranes from rats treated with 17 alpha-ethinyl estradiol. J. Biol. Chem.254: 11367–11373. 12.Strachan, A. F., F. C. de Beer, D. R. van der Westhuyzen, and G. A.Coetzee. 1988. Identification of three isoform patterns of human serum amyloid A proteins. Biochem. J.250: 203–207.13.Bilheimer, D. W., S. Eisenberg, and R. I. Levy. 1972. The metabo-lism of very low density lipoprotein. Biochim. Biophys. Acta.260:212–221.14.Gwynne, J. T., and D. D. Mahaffee. 1989. Rat adrenal uptake andmetabolism of high density lipoprotein cholesteryl ester. J. Biol.Chem.264: 8141–8150.15.Glass, C., R. C. Pittman, D. W. Weinstein, and D. Steinberg. 1983. Dis-sociation of tissue uptake of cholesterol from that of apoprotein A-Iof rat high density lipoproteins: selective delivery of cholesterol toliver, adrenal, and gonad. Proc. Natl. Acad. Sci. USA.80: 5435–5439.16.Knecht, T. P., and R. C. Pittman. 1989. A plasma membrane poolof cholesteryl esters that may mediate the selective uptake of cho-lesteryl esters from high density lipoproteins. Biochim. Biophys.Acta.1002: 365–375.17.Wise, P. M., P. Camp-Grossman, and C. A. Barraclough. 1981. Ef-fects of estradiol and progesterone on plasma gonadotropins, pro-lactin, and LHRH in specific brain areas of ovariectomized rats.Biol. Reprod.24: 820–830.18.Daugherty, A., E. Puré, D. Delfel-Butteiger, S. Chen, J. Leferovich,S. E. Roselaar, and D. J. Rader. 1997. The effects of total lympho-cyte deficiency on the extent of atherosclerosis in apolipoproteinEϪ/Ϫ mice. J. Clin. Invest.100: 1575–1580.19.Dubal, D. B., M. L. Kashon, L. C. Pettigrew, J. M. Ren, S. P. Fink-lestein, S. W. Rau, and P. M. Wise. 1998. Estradiol protects againstischemic injury. J. Cereb. Blood Flow Metab.18: 1253–1258.20.Smith, M. S., M. E. Freeman, and J. D. Neill. 1975. The control ofprogesterone secretion during the estrous cycle and earlypseudopregnancy in the rat: prolactin, gonadotropin and steroidlevels associated with rescue of the corpus luteum of pseudopreg-nancy. Endocrinology.96: 219–226.21.Fluiter, K., D. R. van der Westhuyzen, and T. J. C. van Berkel. 1998.In vivo regulation of scavenger receptor BI and the selective up-take of high density lipoprotein cholesteryl esters in rat liver pa-renchymal and kupffer cells. J. Biol. Chem.273: 8434–8438.22.Babitt, J., B. Trigatti, A. Rigotti, E. J. Smart, R. G. Anderson, S. Xu,and M. Krieger. 1997. Murine SR-BI, a high density lipoprotein re-ceptor that mediates selective lipid uptake, is N-glycosylated andfatty acylated and colocalizes with plasma membrane caveolae. J.Biol. Chem.272: 13242–13249.23.Connelly, M. A., S. M. Klein, S. Azhar, N. A. Abumrad, and D. L.Williams. 1999. Comparsion of class B scavenger receptors, CD36and scavenger receptor BI (SR-BI), shows that both receptorsmediate high density lipoprotein-cholesteryl ester selective uptakebut SR-BI exhibits a unique enhancement of cholesteryl ester up-take. J. Biol. Chem.274: 41–47.24.Srivastava, R. A., E. S. Krul, R. C. Lin, and G. Schonfeld. 1997. Reg-ulation of lipoprotein metabolism by estrogen in inbred strains ofmice occurs primarily by posttranscriptional mechanisms. Mol. CellBiochem.173: 161–168.25.Srivastava, R. A., N. Srivastava, M. Averna, R. C. Lin, K. S. Korach,D. B. Lubahn, and G. Schonfeld. 1997. Estrogen up-regulates apoli-poprotein E (ApoE) gene expression by increasing ApoE mRNAin the translating pool via the estrogen receptor alpha-mediatedpathway. J. Biol. Chem.272: 33360–33366.by on May 16, 2007Downloaded fromGraf, Roswell, and Smart17-Estradiol regulation of SR-BII1449。
肿瘤坏死因子-α在非酒精性脂肪性肝病进展中的作用郭悦承; 陆伦根【期刊名称】《《胃肠病学》》【年(卷),期】2019(024)010【总页数】4页(P623-626)【关键词】肿瘤坏死因子α; 非酒精性脂肪性肝病; 非酒精性脂肪性肝炎; 胰岛素抵抗; 脂代谢【作者】郭悦承; 陆伦根【作者单位】上海交通大学附属第一人民医院消化科 200080【正文语种】中文肝细胞内脂肪沉积是发生非酒精性脂肪性肝病(non-alcoholic fatty liver disease, NAFLD)的标志性病理改变,肝细胞中的脂滴主要由三酰甘油组成。
病理上,若超过5%的肝细胞内含有脂滴,即可诊断为脂肪肝。
细胞因子如肿瘤坏死因子(TNF)-α、白细胞介素(IL)-1、转化生长因子-β(TGF-β)等可参与NAFLD进程,直接或间接地对肝细胞造成损伤。
有研究指出,脂肪组织中IL-6和TNF-α表达与NAFLD 的严重程度呈正相关[1]。
TNF-α是一种由巨噬细胞和单核细胞产生的促炎细胞因子,在恶性肿瘤、败血症、慢性炎症等病理状态下显著增多。
TNF-α可能是单纯性脂肪肝进展为非酒精性脂肪性肝炎(non-alcoholic steatohepatitis, NASH)的重要参与因子。
高浓度TNF-α可促进脂肪动员、抑制外周脂肪组织分解、下调胰岛素活化受体能力,在介导脂肪肝形成、胰岛素抵抗方面发挥重要作用。
本文就肝细胞损伤与凋亡、脂代谢、胰岛素抵抗、肝细胞线粒体障碍、脂质过氧化损伤等对TNF-α在NAFLD进展中作用的影响作一综述。
一、TNF-α的生理功能TNF-α已被证实可调节多种炎症和自身免疫过程。
TNF-α可通过促进T细胞增殖、损伤血管内皮细胞等作用杀伤肿瘤。
在炎症反应中,TNF-α可提高中性粒细胞的吞噬能力,刺激巨噬细胞和单核细胞分泌IL-1等炎症因子。
TNF-α也可通过募集免疫细胞、诱导产生前列腺素和环氧合酶、诱导氧化应激等途径促进细胞变性与炎症进展[2]。
ACE 基因多态性与不同类型脑梗死的研究进展韩丽雅;黄小亚;茅新蕾;黄向东【期刊名称】《心脑血管病防治》【年(卷),期】2015(000)001【总页数】3页(P59-61)【关键词】脑梗死亚型;ACE基因多态性【作者】韩丽雅;黄小亚;茅新蕾;黄向东【作者单位】325000 浙江省温州市中心医院神经内科;325000 浙江省温州市中心医院神经内科;325000 浙江省温州市中心医院神经内科;325000 浙江省温州市中心医院神经内科【正文语种】中文【中图分类】R743.3血管紧张素转换酶(angiotensin converting enzyme,ACE)基因是肾素-血管紧张素系统的关键基因,ACE主要分布在肺、肾近曲小管、小肠绒毛和毛细血管上皮细胞,属肽酰二肽水解酶。
ACE能影响血管紧张素Ⅱ的形成和缓激肽的灭活,继而影响机体内的水钠代谢、醛固酮分泌、血管收缩以及血管平滑肌细胞增殖,然后产生一系列病理生理学改变[1]。
近年来的研究显示,ACE基因可能是脑梗死遗传易感性的分子学基础[2]。
1.1 ACE的概述:ACE是一种位于内皮细胞和上皮细胞膜的 Zn2+依赖性羧二肽酶,相对分子质量为14000~16000,结构类似于蛋白羧化酶A,含有两个同源结构域,每一结构域中有一个潜在的Zn2+结合区和催化区,催化寡肽释放羧基端二肽,主要底物有血管紧张素I(angiotensin,Ⅰ)和缓激肽[3]。
另外ACE的羧基端有一疏水区,可能是ACE与细胞膜结合的部位,可使血管紧张素Ⅰ转变成血管紧张素Ⅱ,还可以使缓激肽灭活[4]。
ACE通过肾上腺素¯血管紧张素系统以及激肽释放酶¯激肽系统,调节血压、水电解质平衡以及血管内皮发育。
ACE将无活性的十肽血管紧张素Ⅰ转换为具有活性的八肽血管紧张素Ⅱ,后者为肾素¯血管紧张素系统的主要活性产物,除具有强烈血管收缩活性之外,还可作用于肾上腺皮质促进醛固酮释放。
生物药剂学与药物动力学专业词汇※<A>Absolute bioavailability, F 绝对生物利用度Absorption 吸收Absorption pharmacokinetics 吸收动力学Absorption routes 吸收途径Absorption rate 吸收速率Absorption rate constant 吸收速率常数Absorptive epithelium 吸收上皮Accumulation 累积Accumulation factor 累积因子Accuracy 准确度Acetylation 乙酰化Acid glycoprotein 酸性糖蛋白Active transport 主动转运Atomic absorption spectrometry 原子吸收光谱法Additive 加和型Additive errors 加和型误差Adipose 脂肪Administration protocol 给药方案Administration route 给药途径Adverse reaction 不良反应Age differences 年龄差异Akaike’s information criterion, AIC AIC判据Albumin 白蛋白All-or-none response 全或无效应Amino acid conjugation 氨基酸结合Analog 类似物Analysis of variance, ANOVA ANOVA方差分析Anatomic Volume 解剖学体积Antagonism 拮抗作用Antiproliferation assays 抑制增殖法Apical membrane 顶端表面Apoprotein 载脂蛋白脱辅基蛋白Apparatus 仪器Apparent volume of distribution 表观分布容积Area under the curve, AUC 曲线下面积Aromatisation 芳构化Artery 动脉室Artifical biological membrane 人工生物膜Aryl 芳基Ascorbic acid 抗坏血酸维生素C Assistant in study design 辅助实验设计Average steady-state plasma drug concentration 平均稳态血浆药物浓度Azo reductase 含氮还原酶※<B>Backward elimination 逆向剔除Bacteria flora 菌丛Basal membrane 基底膜Base structural model 基础结构模型Basolateral membrane 侧底膜Bayesian estimation 贝易斯氏评估法Bayesian optimization 贝易斯优化法Bile 胆汁Billiary clearance 胆汁清除率Biliary excretion 胆汁排泄Binding 结合Binding site 结合部位Bioactivation 生物活化Bioavailability, BA 生物利用度Bioequivalence, BE 生物等效性Biological factors 生理因素Biological half life 生物半衰期Biological specimen 生物样品Biomembrane limit 膜限速型Biopharmaceutics 生物药剂学Bioequivalency criteria 生物等效性判断标准Biotransformation 生物转化Biowaiver 生物豁免Blood brain barrier, BBB BBB血脑屏障Blood clearance 血液清除率Blood flow rate-limited models 血流速度限速模型Blood flux in tissue 组织血流量Body fluid 体液Buccal absorption of drug 口腔用药的吸收Buccal mucosa 口腔粘膜颊粘膜Buccal spray formulation 口腔喷雾制剂※<C>Capacity limited 容量限制Carrier mediated transport 载体转运Catenary model 链状模型Caucasion 白种人Central compartment 中央室Characteristic 特点Chelate 螯合物Chinese Traditional medicine products 中药制剂Cholesterol esterase 胆固醇酯酶Chromatogram 色谱图Circulation 循环Classification 分类Clearance 清除率Clinical testing in first phase I期临床试验Clinical testing in second phase Ⅱ期临床试验Clinical testing in third phase Ⅲ期临床试验Clinical trial 临床试验Clinical trial simulation 临床实验计划仿真Clockwise hysteresis loop 顺时针滞后回线Collection 采集Combined administration 合并用药Combined errors 结合型误差Common liposomes, CL 普通脂质体Compartment models 隔室模型Compartments 隔室Competitive interaction 竞争性相互作用Complements 补体Complex 络合物Confidential interval 置信区间Conjugation with glucuronic acid 葡萄糖醛酸结合Controlled-release preparations 控释制剂Control stream 控制文件Conventional tablet 普通片Convergence 收敛Convolution 卷积Corresponding relationship 对应关系Corticosteroids 皮质甾体类Counter-clockwise hysteresis loop 逆时针滞后回线Countermeasure 对策Course in infusion period 滴注期间Covariance 协方差Covariates 相关因素Creatinine 肌酐Creatinine clearance 肌酐清除率Cytochrome P450, CYP450 细胞色素P450 Cytoplasm 细胞质Cytosis 胞饮作用Cytosol 胞浆胞液质※<D>Data File 数据文件Data Inspection 检视数据Deamination 脱氨基Deconvolution 反卷积Degree of fluctuation, DF DF波动度Delayed release preparations 迟释制剂Desaturation 降低饱和度Desmosome 桥粒Desulfuration 脱硫Detoxication 解毒Diagnosis 诊断Diffusion 扩散作用Dietary factors 食物因素Displacement 置换作用Disposition 处置Dissolution 溶解作用Distribution 分布Dosage adjustment 剂量调整Dosage form 剂型Dosage form design 剂型设计Dosage regimen 给药方案Dose 剂量dose-proportionality study 剂量均衡研究Dropping pills 滴丸Drug absorption via eyes 眼部用药物的吸收Drug binding 药物结合Drug concentration in plasma 血浆中药物浓度Drug Delivery System, DDS 药物给药系统Drug interaction 药物相互作用Drug-plasma protein binding ratio 药物—血浆蛋白结合率Drug-Protein Binding 药物蛋白结合Drug transport to foetus 胎内转运※<E>Efficient concentration range 有效浓度范围Efflux 外排Electrolyte 电解质Electro-spray ionization, ESI 电喷雾离子化Elimination 消除Elimination rate constant 消除速度常数Elongation 延长Emulsion 乳剂Endocytosis 入胞作用Endoplasmic reticulum 内质网Enterohepatic cycle 肠肝循环Enzyme 酶Enzyme induction 酶诱导Enzyme inhibition 酶抑制Enzyme-linked immunosorbent assays ELISA 酶联免疫法Enzymes or carrier-mediated system 酶或载体—传递系统Epithelium cell 上皮细胞Epoxide hydrolase 环化物水解酶Erosion 溶蚀Excretion 排泄Exocytosis 出胞作用Exons 外显子Experimental design 实验设计Experimental procedures 实验过程Exponential errors 指数型误差Exposure-response studies 疗效研究Extended least squares, ELS 扩展最小二乘法Extended-release preparations 缓控释制剂Extent of absorption 吸收程度External predictability 外延预见性Extraction ratio 抽取比Extract recovery rate 提取回收率Extrapolation 外推法Extravascular administration 血管外给药※<F>F test F检验Facilitated diffusion 促进扩散Factors of dosage forms 剂型因素Fasting 禁食Fibronectin 纤粘连蛋白First order rate 一级速度First Moment 一阶矩First order absorption 一级吸收First-order conditional estimation, FOCE 一级条件评估法First-order estimation, FO 一级评估法Fiest-order kinetics 一级动力学First pass effect 首过作用首过效应Fixed-effect parameters 固定效应参数Flavoprotein reductaseNADPH-细胞色素还原酶附属黄素蛋白还原酶Flow-through cell dissolution method 流室法Fluorescent detection method 荧光检测法Fraction of steady-state plasma drug concentration 达稳分数Free drug 游离药物Free drug concentration 游离药物浓度※<G>Gap junction 有隙结合Gas chromatography, GC 气相色谱法Gasrtointestinal tract, GI tract 胃肠道Gender differences 性别差异Generalized additive modeling, GAM 通用迭加模型化法Glimepiride 谷胱甘肽Global minimum 整体最小值Glomerular filtration 肾小球过滤Glomerular filtration rate, GFR 肾小球过滤率Glucuonide conjugation 葡萄糖醛酸结合Glutathione conjugation 谷胱甘肽结合Glycine conjugation 甘氨酸结合Glycocalyx 多糖—蛋白质复合体Goodness of Fit 拟合优度Graded response 梯度效应Graphic method 图解法Gut wall clearance肠壁清除率※<H>Half life 半衰期Health volunteers 健康志愿者Hemodialysis 血液透析Hepatic artery perfusion administration 肝动脉灌注给药Hepatic clearance, Clh 肝清除率Hierarchical Models 相同系列药物动力学模型High performance liquid chromatography, HPLC 高效液相色谱Higuchi equation Higuchi 方程Homologous 类似Human liver cytochrome P450 人类肝细胞色素P450 Hydrolysis 水解Hydroxylation 羟基化Hysteresis 滞后Hysteresis of plasma drug concentration 血药浓度滞后于药理效应Hysteresis of response 药理效应滞后于血药浓度※<I>Immunoradio metrec assays, IRMA 免疫放射定量法Incompatibility 配伍禁忌Independent 无关,独立Individual parameters 个体参数Individual variability 个体差异Individualization of drug dosage regimen 给药方案的个体化Inducer 诱导剂Induction 诱导Infusion 输注Inhibition 抑制Inhibitor 抑制剂Initial dose 速释部分Initial values 初始值Injection sites 注射部位Insulin 胰岛素Inter-compartmental clearance 隔室间清除率Inter-individual model 个体间模型Inter-individual random effects 个体间随机效应Inter-individual variability 个体间变异性Intermittence intravenous infusion 间歇静脉输液Internal predictability 内延预见性Inter-occasion random effects 实验间随机效应Intestinal bacterium flora 肠道菌丛Intestinal metabolism 肠道代谢Intra-individual model 个体内模型Intra-individual variability 个体内变异性Intramuscular administration 肌内给药Intramuscular injection 肌内注射Intra-peritoneal administration 腹腔给药Intravenous administration 静脉给药Intravenous infusion 静脉输液Intravenous injection 静脉注射Intrinsic clearance固有清除率内在清除率Inulin 菊粉In vitro experiments 体外试验In vitro–In vivo correlation, IVIVC 体外体内相关关系In vitro mean dissolution time, MDT vitro 体外平均溶出时间In vivo Mean dissolution time, MDT vivo 体内平均溶出时间Ion exchange 离子交换Isoform 异构体Isozyme 同工酶※<K>Kerckring 环状皱褶Kidney 肾※<L>Lag time 滞后时间Laplace transform 拉普拉斯变换Lateral intercellular fluid 侧细胞间隙液Lateral membrane 侧细胞膜Least detection amount 最小检测量Linearity 线性Linear models 线性模型Linear regression method 线性回归法Linear relationship 线性关系Lipoprotein 脂蛋白Liposomes 脂质体Liver flow 肝血流Local minimum 局部最小值Loading dose 负荷剂量Logarithmic models 对数模型Long circulation time liposomes 长循环脂质体Loo-Riegelman method Loo-Riegelman法Lowest detection concentration 最低检测浓度Lowest limit of quantitation 定量下限Lowest steady-state plasma drug concentration 最低稳态血药浓度Lung clearance 肺清除率Lymphatic circulation 淋巴循环Lymphatic system 淋巴系统※<M>Maintenance dose 维持剂量Mass balance study 质量平衡研究Masticatory mucosa 咀嚼粘膜Maximum likelihood 最大似然性Mean absolute prediction error, MAPE 平均绝对预测误差Mean absorption time, MAT 平均吸收时间Mean disintegration time, MDIT 平均崩解时间Mean dissolution time, MDT 平均溶出时间Mean residence time, MRT 平均驻留时间Mean sojourn time 平均逗留时间Mean squares 均方Mean transit time 平均转运时间Membrane-limited models 膜限速模型Membrane-mobile transport 膜动转运Membrane transport 膜转运Metabolism 代谢Metabolism enzymes 代谢酶Metabolism locations 代谢部位Metabolites 代谢物Metabolites clearance, Clm 代谢物清除率Method of residuals 残数法剩余法Methylation 甲基化Michaelis-Menten equation 米氏方程Michaelis-Menten constant 米氏常数Microbial assays 微生物检定法Microsomal P-450 mixed-function oxygenases 肝微粒体P-450混合功能氧化酶Microspheres 微球Microvilli 微绒毛Minimum drug concentration in plasma 血浆中最小药物浓度Mixed effects modeling 混合效应模型化Mixed-function oxidase, MFO 混合功能氧化酶Models 模型Modeling efficiency 模型效能Model validation 模型验证Modified release preparations 调释制剂Molecular mechanisms 分子机制Mono-exponential equation 单指数项公式Mono-oxygenase 单氧加合酶Mucous membrane injury 粘膜损伤Multi-compartment models 多室模型延迟分布模型Multi-exponential equation 多指数项公式Multifactor analysis of variance, multifactor ANOVA 多因素方差分析Multiple dosage 多剂量给药Multiple-dosage function 多剂量函数Multiple-dosage regimen 多剂量给药方案Multiple intravenous injection 多次静脉注射Myoglobin 肌血球素※<N>Naive average data, NAD 简单平均数据法Naive pool data, NPD 简单合并数据法Nanoparticles 纳米粒Nasal cavity 鼻腔Nasal mucosa 鼻粘膜National Institute of Health 美国国立卫生研究所Nephron 肾原Nephrotoxicity 肾毒性No hysteresis 无滞后Non-compartmental analysis, NCA 非隔室模型法Non-compartmental assistant Technology 非隔室辅助技术Nonionized form 非离子型Nonlinear mixed effects models, NONMEM 非线性混合效应模型Nonlinear pharmacokinetics 非线性药物动力学Non-linear relationship 非线性关系Nonparametric test 非参数检验※<O>Objective function, OF 目标函数Observed values 观测值One-compartment model 一室模型(单室模型)Onset 发生Open randomized two-way crossover design 开放随机两路交叉实验设计Open crossover randomized design 开放交叉随机设计Oral administration 口服给药Ordinary least squares, OLS 常规最小二乘法Organ 器官Organ clearance 器官清除率Original data 原始数据Osmosis 渗透压作用Outlier 偏离数据Outlier consideration 异常值的考虑Over-parameterized 过度参数化Oxidation 氧化Oxidation reactions 氧化反应※<P>Paracellular pathway 细胞旁路通道Parameters 参数Passive diffusion 被动扩散Pathways 途径Patient 病人Peak concentration 峰浓度Peak concentration of drug in plasma 血浆中药物峰浓度Poly-peptide 多肽Percent of absorption 吸收百分数Percent of fluctuation, PF 波动百分数Perfused liver 灌注肝脏Period 周期Peripheral compartments 外周室Peristalsis 蠕动Permeability of cell membrane 细胞膜的通透性P-glycoprotein, p-gp P-糖蛋白Phagocytosis 吞噬Pharmaceutical dosage form 药物剂型pharmaceutical equivalents 药剂等效性Pharmacokinetic models 药物动力学模型Pharmacokinetic physiological models 药物动力学的生理模型Pharmacological effects 药理效应Pharmacologic efficacy 药理效应Pharmacokinetics, PK 药物动力学Pharmacokinetic/pharmacodynamic link model 药物动力学-药效动力学统一模型Pharmacodynamics, PD 药效动力学Pharmacodynamic model 药效动力学模型Phase II metabolism 第II相代谢Phase I metabolism 第I相代谢pH-partition hypothesis pH分配假说Physiological function 生理功能Physiological compartment models 生理房室模型Physiological pharmacokinetic models 生理药物动力学模型Physiological pharmacokinetics 生理药物动力学模型Pigment 色素Physicochemical factors 理化因素Physicochemical property of drug 药物理化性质Physiological factors 生理因素Physiology 生理Physiological pharmacokinetic models 生理药物动力学模型Pinocytosis 吞噬Plasma drug concentration 血浆药物浓度Plasma drug concentration-time curve 血浆药物浓度-时间曲线Plasma drug-protein binding 血浆药物蛋白结合Plasma metabolite concentration 血浆代谢物浓度Plasma protein binding 血浆蛋白结合Plateau level 坪浓度Polymorphism 多态性Population average pharmacokinetic parameters 群体平均动力学参数Population model 群体模型Population parameters 群体参数Population pharmacokinetics 群体药物动力学Post-absorptive phase 吸收后相Post-distributive phase 分布后相Posterior probability 后发概率practical pharmacokinetic program 实用药代动力学计算程序Precision 精密度Preclinical 临床前的Prediction errors 预测偏差Prediction precision 预测精度Predicted values 拟合值Preliminary structural model 初始结构模型Primary active transport 原发性主动转运Principle of superposition 叠加原理Prior distribution 前置分布Prodrug 前体药物Proliferation assays 细胞增殖法Proportional 比例型Proportional errors 比例型误差Prosthehetic group 辅基Protein 蛋白质Pseudo-distribution equilibrium 伪分布平衡Pseudo steady state 伪稳态Pulmonary location 肺部Pulsatile drug delivery system 脉冲式释药系统※<Q、R>QQuality controlled samples 质控样品Quality control 质量控制Quick tissue 快分布组织RRadioimmuno assays, RIA 放射免疫法Random error model 随机误差模型Rapid intravenous injection 快速静脉注射Rate constants 速度常数Rate method 速度法Re-absorption 重吸收Receptor location 受体部位Recovery 回收率Rectal absorption 直肠吸收Rectal blood circulation 直肠部位的血液循环Rectal mucosa 直肠黏膜Reductase 还原酶Reduction 还原Reductive metabolism 还原代谢Reference individual 参比个体Reference product 参比制剂Relative bioavailability, Fr 相对生物利用度Release 释放Release medium 释放介质Release standard 释放度标准Renal 肾的Renal clearance, Clr 肾清除率Renal excretion 肾排泄Renal failure 肾衰Renal impairment 肾功能衰竭Renal tubular 肾小管Renal tubular re-absorption 肾小管重吸收Renal tubular secretion 肾小管分泌Repeatability 重现性Repeated one-point method 重复一点法Requirements 要求Research field 研究内容Reside 驻留Respiration 呼吸Respiration organ 呼吸器官Response 效应Residuals 残留误差Residual random effects 残留随机效应Reversal 恢复Rich Data 富集数据Ritschel one-point method Ritschel 一点法Rotating bottle method 转瓶法Rough surfaced endoplasmic reticulum 粗面内质网Routes of administration 给药途径※<S、T>SSafety and efficacy therapy 安全有效用药Saliva 唾液Scale up 外推Scale-Up/Post-Approval Changes, SUPAC 放大/审批后变化Second moment 二阶矩Secondary active transport 继发性主动转运Secretion 分泌Sensitivity 灵敏度Serum creatinine 血清肌酐Sigma curve 西格玛曲线Sigma-minus method 亏量法(总和减量法)Sigmoid curve S型曲线Sigmoid model Hill’s方程Simulated design 模拟设计Single-dose administration 单剂量(单次)给药Single dose response 单剂量效应Sink condition 漏槽条件Skin 皮肤Slow Tissue 慢分布组织Smooth surfaced endoplasmic reticulum 滑面内质网Soluble cell sap fraction 可溶性细胞液部分Solvent drag effect 溶媒牵引效应Stability 稳定性Steady-state volume of distribution 稳态分布容积Sparse data 稀疏数据Special dosage forms 特殊剂型Special populations 特殊人群Specialized mucosa 特性粘膜Species 种属Species differences 种属差异Specificity 特异性专属性Square sum of residual error 残差平方和Stagnant layer 不流动水层Standard curve 标准曲线Standard two stage, STS 标准两步法Statistical analysis 统计分析Statistical moments 统计矩Statistical moment theory 统计矩原理Steady state 稳态Steady state plasma drug concentration 稳态血药浓度Stealth liposomes, SL 隐形脂质体Steroid 类固醇Steroid-sulfatases 类固醇-硫酸酯酶Structure 结构Structure and function of GI epithelial cells 胃肠道上皮细胞的构造与功能Subcutaneous injections 皮下注射Subgroup 亚群体Subjects 受试者Sublingual administration 舌下给药Sublingual mucosa 舌下粘膜Subpopulation 亚群Substrate 底物Sulfate conjugation 硫酸盐结合Sulfation 硫酸结合Sum of squares 平方和Summation 相加Superposition method 叠加法Susceptible subject 易受影响的患者Sustained-release preparations 缓释制剂Sweating 出汗Synergism 协同作用Systemic clearance 全身清除率TTargeting 靶向化Taylor expansion 泰勒展开Tenous capsule 眼球囊Test product 试验制剂Therapy drug monitoring, TDM 治疗药物监测Therapeutic index 治疗指数Thermospray 热喷雾Three-compartment models 三室模型Though concentration 谷浓度Though concentration during steady state 稳态谷浓度Thromboxane 血栓素Tight junction 紧密结合Tissue 组织Tissue components 组织成分Tissue interstitial fluid 组织间隙Tolerance 耐受性Topping effect 尖峰效应Total clearance 总清除率Toxication and emergency treatment 中毒急救Transcellular pathway 经细胞转运通道Transdermal absorption 经皮肤吸收Transdermal drug delivery 经皮给药Transdermal penetration 经皮渗透Transport 转运Transport mechanism of drug 药物的转运机理Trapezoidal rule 梯形法Treatment 处理Trial Simulator 实验计划仿真器Trophoblastic epithelium 营养上皮层Two-compartment models 二室模型Two one sided tests 双单侧t检验Two period 双周期Two preparations 双制剂Two-way crossover bioequivalence studies 双周期交叉生物等效性研究Typical value 典型值※<U~Z>UUnwanted 非预期的Uniformity 均一性Unit impulse response 单位刺激反应Unit line 单位线Urinary drug concentration 尿药浓度Urinary excretion 尿排泄Urinary excretion rate 尿排泄速率VVagina 阴道Vaginal Mucosa 阴道黏膜Validation 校验Variance of mean residence time, VRT 平均驻留时间的方差Vein 静脉室Villi 绒毛Viscre 内脏Volumes of distribution 分布容积volunteers or patients studies 人体试验WWagner method Wagner法Wagner-Nelson method Wagner-Nelson法Waiver requirements 放弃(生物等效性研究)要求Washout period 洗净期Weibull distribution function Weibull分布函数Weighted Least Squares WLS加权最小二乘法Weighted residuals 加权残留误差XXenobiotic 外源物, 异生素ZZero Moment 零阶矩Zero-order absorption 零级吸收Zero-order kinetics 零级动力学Zero order rate 零级速度Zero-order release 零级释放。
Minutes101552025M o n e n s i n BM o n e n s i n AS a l i n o m y c i nN a r a s i nPolyether Antibiotics聚醚类抗生素Monensin, Narasin, and Salinomycin聚醚类抗生素,例如莫能菌素(Monensin)、Narasin 和盐霉素(Salinomycin),存在于原料、预混合料、液体补充剂和饲料产品中,采用高效液相色谱和柱后衍生仪能够对其进行定量分析。
聚醚类抗生素在硫酸存在的条件下与香草醛(Vanillin)反应,其衍生产物能够在520 nm 处被检测。
但是,这种腐蚀性的试剂会严重地损坏大部分仪器。
然而,Pickering 公司的PCX5200柱后衍生设备,主要由非金属零件和清洗泵头组成,能够耐受硫酸的腐蚀性,同时提供特殊的操作说明。
需要获得PCX5200柱后衍生设备用于聚醚类抗生素分析的更多资料,可联系Pickering Laboratories 。
方法分析条件色谱柱: 聚醚类反相柱, C18, 4.6 x 250 mm, 货号2381750温度: 40 °C流速: 0.7 mL/min流动相: 甲醇/5%醋酸水溶液(90/10), 等度洗脱柱后衍生条件柱后衍生系统: Pinnacle PCX (要求非金属柱后衍生系统)试剂1: 浓硫酸/甲醇(4:96 v/v)反应器1: 室温, 0.1 mL试剂2: 60 g 香草醛(或者p-dimethylaminoben-zaldehyde, DMAB) 溶于950 mL 甲醇反应器2: 90 °C, 1.4 mL 流速: 0.3 mL/min检测器: 紫外可见分光光度计:香草醛λ=520 nm, DMAB λ=450 nm货号 描述2381750 聚醚类反相柱, C 18, 4.6×250 mm 18ECG001 保护柱卡套, 带3个保护柱芯 3700-2200 香草醛, 1x30 g/瓶3700-0400p-Dimethylaminobenzaldehyde (DMAB), 1x5 g/瓶色谱柱和试剂。
基于TaqMan-TAMRA探针技术的ApoE基因快速分型方法研究摘要】目的建立基于TaqMan-TAMRA探针技术的快速、准确的载脂蛋白E(ApoE)基因型检测方法,为临床诊断、治疗和预防ApoE基因相关疾病提供依据。
方法对1049例血液样本提取DNA后,利用TaqMan-TAMRA探针对ApoE基因进行检测和分型,并利用测序法进行验证。
结果 1049例样本中,共检出1048例,ApoE基因的6种基因型(ε3/ε3、ε2/ε3、ε3/ε4、ε2/ε4、ε2/ε2、ε4/ε4)所占比例分别为68.64%、13.54%、14.97%、1.72%、0.76%、0.38%。
TaqMan-TAMRA探针法分型结果与测序结果完全一致。
结论 TaqMan-TAMRA探针法操作简便,分型结果准确度高,是一种适用于临床的ApoE基因快速分型方法。
【关键词】载脂蛋白ETaqMan-TAMRA基因分型测序【中图分类号】R319 【文献标识码】A 【文章编号】2095-1752(2013)35-0132-02Development of a fast genotyping forApoEbased on TaqMan-TAMRAprobes【Abstract】Objective:To establish a method for fast and accurategenotyping ApoE with TaqMan-TAMRA probes and to support the clinical diagnosis、treatment and prevention.Methods:DNA was extracted from 1049 blood samples, then detected and genotyped with TaqMan TAMRA probes. The genotypes were compared with sequencing results. Results: DNA were detected in 1048 out of 1049 samples.Of which, six genotypes of ApoE gene (ε3/ε3, ε2/ε3, ε3/ε4, ε2/ε4, ε2/ε2, ε4/ε4) accounted for 68.64%, 13.54%, 14.97%, 1.72%, 0.76%, 0.38%.TaqMan-TAMRA probe genotypes were consistent with sequencing results. Conclusion:TaqMan-TAMRA is a fast and accurate method for genotyping ApoE.【Key words】 Apolipoprotein E TaqMan-TAMRA genotyping sequencing 载脂蛋白(ApoE)基因位于人的第19号染色体,编码由299个氨基酸组成的ApoE蛋白,该蛋白具有脂蛋白转运、代谢及修复细胞膜等功能[1,2]。
小动物活体光学成像技术在糖尿病研究中的应用PerkinElmer小动物活体光学成像技术已在生命科学基础研究、临床前医学研究及药物研发等领域得到广泛应用。
在众多应用领域中,糖尿病相关研究是近几年又一兴起的应用热点之一。
将活体光学成像技术应用于糖尿病研究的主要方向包括:1、从特异构建的发光转基因小鼠中获取具有发光特性的胰岛,进行胰岛移植相关研究;2、利用荧光素酶基因标记相关治疗用细胞,观测治疗用细胞在活体动物体内的分布、器官靶向及对糖尿病的治疗效果;3、通过构建荧光素酶基因表达载体或转基因动物,研究糖尿病相关基因表达及信号通路。
下面结合一些具体实例进行阐述:一.胰岛移植相关研究I型糖尿病,即胰岛素依赖性糖尿病,是由感染、毒物等因素诱发机体产生异常自身体液和细胞免疫应答,导致胰岛β细胞损伤,胰岛素分泌减少。
胰岛移植的主要适应症为胰岛素依赖型糖尿病。
众多实验研究证实,胰岛移植不仅可以纠正实验动物的糖尿病状态,而且可有效地防止糖尿病微血管病变的发生、发展,促进糖代谢内环境稳定,降低死亡率。
应用活体光学成像技术可以在活体动物水平长期监测胰岛移植的存活。
为了实现这一应用,研究者首先需要对胰岛进行光学标记,通常采用的方法是利用荧光素酶基因标记胰岛素基因启动子,而构建胰腺特异性发光的转基因动物,从该转基因动物体内即可直接提取具备发光特性的胰岛。
上图:左,Tg(RIP-luc)转基因小鼠表达载体示意图;应用IVIS系统对发光转基因小鼠及对照的成像结果。
从转基因小鼠获取发光的胰岛之后,便可开展胰岛移植的相关观测实验。
如观测不同数量或不同部位胰岛移植的存活情况。
如下图所示,应用IVIS系统可以观测胰岛在不同部位的移植情况,并且基于IVIS系统的超高灵敏度,可以观测到少量胰岛移植后的发光情况。
上图:分别将10个或150个从FVB-Tg(RIP-luc)转基因小鼠中取出的胰岛移植于同种小鼠的不同部位,应用IVIS系统进行观测。
uderback et al./Brain Research924(2002)90–9791 increased risk of Alzheimer’s disease(AD)[46],and 2.2.Animalscorrelates with increased oxidative damage in the AD brain[38,43,44,52].AD is characterized by the overproduction All protocols used in this study were approved by the and accumulation of39–43amino acid beta-amyloid University of Kentucky Institutional Animal Care and Use peptides(A b),which are thought to be central to the Committee.The apoE gene replacement animals were disorder[47].A b induces protein and lipid oxidation in generated as described in Refs.[50,51].Briefly,for all neurons and ultimately results in cell death[7,31,55,56,58–these apoE gene replacement mice,the mouse apoE coding 60].Because reactive oxygen species(ROS)are thought to exons2–4were replaced with the human counterparts for mediate these effects[3,14–16,56],the oxidative damage the apoE e2,e3or e4.Fig.1shows that human apoE is and toxicity of A b can be inhibited by antioxidants present in synaptosomes isolated from each of these [12,21,55,58,60].This evidence suggests that A b may knock-in mice.contribute to the oxidative damage in the AD brain andthat apoE,by acting as an antioxidant,may modulate these 2.3.Synaptosomal preparationseffects in an allelic-specific manner.Synaptosomes are a good model of the synapse and Cortical synaptosomes were prepared from knock-in display markers of neurodegeneration upon treatment with mice by ultracentrifugation on discontinuous sucrose gra-oxidants such as A b[35],and were used in the current dients as described previously[28].Synaptosomes were study to investigate the effects of the different alleles of resuspended and washed in Locke’s buffer(154mM NaCl, apoE on A b-induced oxidation.Synaptosomes were iso- 5.6mM KCl,2.3mM CaCl,1.0mM MgCl,3.6mM22lated from mice that expressed only one of the e2,e3or e4NaHCO,5mM glucose,5mM HEPES)before protein3human apoE alleles(human apoE alleles in place of mouse concentrations were determined by the Pierce BCA method apoE via targeted gene replacement),and the modulation and adjusted to1mg/ml.Samples were treated with10 of A b-induced ROS generation and biomarkers for lipid m M A b(1–42)for up to3h at378C.A b was dissolved in and protein oxidation were measured.PBS at a concentration of1mg/2ml,and preincubated for8–9h at378C prior to treatment of synaptosomes.2.4.Measurement of reactive oxygen species(ROS)2.Materials and methodsThe measurement of ROS formation in synaptosomes 2.1.Materials treated with A b(1–42)was performed with dich-lorodihydrofluoroscin-diacetate(DCFH-DA)as described A b(1–42)was purchased from Bachem(Torrance,CA)previously[20].DCFH-DA was dissolved in ethanol at a or U.S.Peptides(Fullerton,CA).Human apoE knock-in concentration of10mM.Aliquots of synaptosomes(1mg) mice were prepared as described previously[50,51].were incubated with10m M DCFH-DA in1ml of Locke’s Protease inhibitors used in the isolation buffer were buffer for30min.Excess dye was removed by washing purchased from ICN(Aurora,OH).Western and slot twice with Locke’s buffer and the synaptosomes were blotting materials including apparatus,nitrocellulose(0.45-resuspended in0.5ml of Locke’s.Intracellular esterases m m pore size),and transferfilter papers were purchased convert DCFH-DA to the anionic DCFH,which is unable from Bio-Rad(Hercules,CA).Anti-HNE was purchased to diffuse out of the synaptosomes.Reaction with ROS from Calbiochem(San Diego,CA).All other reagents converts DCFH to dichlorofluoroscein(DCF),which is were purchased from Sigma(St.Louis,MO).highlyfluorescent.A10-m M sample of A b(1–42)wasFig.1.Western blot of human ne1,authentic human apoE from Panvera ne2,synaptosomes from apoE e2knock-in ne 3,synaptosomes from apoE e3knock-in ne4,synaptosomes from apoE e4knock-in mice.uderback et al./Brain Research924(2002)90–97added to200m g of synaptosomes in a96-well plate and protein were precipitated with0.4ml of ice cold10%the resultingfluorescence was measured(l5485nm;TCA.After centrifugation for5min at6000rpm(30003exl5530nm).g),0.4ml of the supernatant was incubated with0.2ml emthiobarbituric acid(0.335%TBA in50%glacial acetic 2.5.Spin labeling and electron paramagnetic resonance acid)for1h at1008C.Samples were allowed to cool toRT before adding0.4ml of butanol.After mixing each A protein specific spin label,Mal-6(2,2,6,6-tetramethyl-sample with a pipette,the organic layer was allowed to 4-maleimido piperidin-1-oxyl),was used to determine separate from the aqueous layer,and100m l was immedi-conformational changes of synaptosomal membrane pro-ately removed from the top organic phase and added to a teins after treatment with A b[28].Synaptosomes were96-well plate.TBARS were detected by measuring theincubated with10m M of A b(1–42)for2h at378C,andfluorescence with a l of518nm and l of588nm.ex emafter incubation washed in PBS prior to spin labeling.Mal-6was dissolved in100m l of acetonitrile and dilutedto afinal concentration of200m M in50ml of lysing 3.Resultsbuffer(2mM EDTA,2mM EGTA,10mM HEPES,pH7.4).Samples were labeled overnight at48C with12.5m g Mice deficient in apoE exhibit increased markers of Mal-6/mg protein(50m Mfinal concentration)[28].After neuronal oxidative damage[20,28,34,39,42,45]and vul-labeling,synaptosomes were centrifuged and the superna-nerability to oxidative insults[9,20,26,28],while the three tant removed and replaced with fresh lysing buffer.After alleles of apoE possess differential antioxidant and cyto-mixing,the cycle was repeated.Samples were washed six protective activities[8,37,48].We have previously shown times to ensure complete removal of all unbound spin label that apoE is associated with synaptosomes and that apoE-before acquiring EPR spectra.Spectra were acquired on a deficient synaptosomes are more susceptible to A b-in-Bruker EMX spectrometer with the following instrumental duced oxidation[20,28],suggesting an antioxidant role for parameters:microwave power,20mW;microwave fre-apoE.In this study,the role of the different apoE alleles in quency,9.77GHz;modulation amplitude,1.0G;modula-attenuating A b(1–42)-induced oxidative damage to5tion frequency,100KHz;receiver gain,1310;time synaptosomes was investigated.To measure ROS gene-constant,0.64ms.ration by A b(1–42)and possible ROS scavenging byapoE,synaptosomes were loaded with non-fluorescent 2.6.Assessment of protein and lipid oxidation DCFH-DA which,upon oxidation,becomes highlyfluores-cent DCF.Upon treatment of synaptosomes with A b(1–Levels of protein-bound HNE and protein carbonyls42),there is a significant increase in ROS in all samples were determined by slot blot analyses of synaptosomes(Fig.2);however,ROS are significantly elevated(P, before and after treatment with10m M A b(1–42)as0.05)in apoE4synaptosomes when compared to apoE3 described previously[28].Proteins were vacuum-filtered and apoE2synaptosomes.Although there is no statistical onto a nitrocellulose membrane which was subsequently difference between ROS levels in apoE2and apoE3 blocked with PBS containing3%BSA.Detection of samples,a trend suggests that A b(1–42)-induced ROS protein carbonyls required sample derivatization prior to formation is allele-dependent(e2,e3,e4)with ROS being filtration.Sample aliquots(30–50m g protein)were incu-less in apoE2synaptosomes as compared to apoE3 bated with2,4-dinitrophenylhydrazine in the presence of synaptosomes.These data are consistent with previous SDS for20min at room temperature,before neutralization reports of apoE acting as an antioxidant in an allele-with a2M Tris–HCl buffer.The2,4-dinitrophenyl dependent fashion[37]and may possibly explain allele-hydrazone(DNP)adduct of the carbonyls is detected on specific responses to injury that have been reported[8,48]. the nitrocellulose paper using a rabbit antibody specific for ROS generation leads to increased protein and lipid DNP-protein adducts(1:150).HNE-protein adducts were oxidation[6].Therefore,studies were performed to de-detected on the nitrocellulose membrane using a rabbit termine the extent of these parameters in synaptosomes anti-HNE antibody(1:4000).After incubation with respec-treated with A b(1–42)and to assess the effects of apoE tive primary antibodies,nitrocellulose papers were incu-allele type in this process.HNE and TBARS are indices of bated with a secondary antibody(goat anti-rabbit;lipid oxidation and are formed by free radical oxidation of 1:15,000)alkaline phosphatase-conjugate.SigmaFast was unsaturated lipids[11].TBARS measure general,non-used as the colorimetric substrate for alkaline phosphatase.specific oxidation of lipids,while HNE is a specific Blots were scanned into Adobe Photoshop and quantitated product of lipid oxidation whose neurotoxic effects have with Scion Image(PC version of Macintosh compatible been widely studied[4,23,40,49].HNE modification of NIH Image).proteins by reaction with cysteine,histidine,and lysine Levels of thiobarbituric acid reactive substances residues of the protein backbone disrupts protein function (TBARS)were determined in synaptosomes before and and may ultimately result in cell death[6,11,19].Because after treatment with10m M A b(1–42).Aliquots of250m g apoE modulates ROS generation by A b(1–42)in an allele-uderback et al./Brain Research924(2002)90–9793Fig.2.ApoE modulates A b-induced ROS formation in an allele-depen-dent manner.Synaptosomes were loaded with non-fluorescent DCFH-DAwhich becomesfluorescent after oxidation.Upon treatment of synapto-somes with A b(1–42),ROS increase significantly in all samples;however,ROS are significantly elevated in apoE4synaptosomes whencompared to apoE3and apoE2synaptosomes.Data are the mean andS.E.M.(n54–6;*P,0.05vs.e2and e3,paired t-test).dependent manner,the effects of apoE allele on A b(1–42)-induced lipid oxidation were analyzed by measuringTBARS and HNE-immunoreactive proteins.Levels of bothTBARS and protein-bound HNE increased in synapto-somes after treatment with A b(1–42)following a similarallele-specific response described for ROS generation(e2,e3,e4).Increases in TBARS were significant in allsynaptosomal preparations with significant differencesbetween all alleles(Fig.3A).Interestingly,synaptosomesdeficient in apoE paralleled the response of apoE4synapto-somes to A b(1–42)-induced TBARS formation(data not Fig.3.Synaptosomal lipid peroxidation induced by A b(1–42)is me-diated by apoE in an allele-specific fashion.(A)TBARS are markers for shown).Due to large scatter,allele-dependent differencesgeneral lipid oxidation and increase in synaptosomes upon treatment with in HNE-immunoreactivity were insignificant(Fig.3B);A b(1–42).Between each apoE allele,there are significant differences in however,mean increases in HNE followed the same trendTBARS with the e4allele being most vulnerable to A b-induced lipid that was evident in both TBARS and ROS,i.e.HNE oxidation.(B)HNE is a specific marker of lipid oxidation.Although no increased the most in apoE4synaptosomes.These data significant changes occur,an allele-specific trend parallels TBARSformation and suggests that HNE levels are highest when apoE4is support the role of apoE as a modulator of A b-inducedpresent.Data are the mean and S.E.M.(n53–5;*P,0.05,paired t-test). oxidative stress and are consistent with the correlation ofthe e4allele with increased lipid oxidation in AD[38,43,44,52].strongly hindered motion of Mal-6results in broad lines. Protein carbonyls and protein conformational changes The resulting intensities of the respective W and S peaks ofare direct and indirect markers of protein oxidation,the M511lowfield resonance lines give rise to the W/SIrespectively[6].Changes in protein conformations are ratio,a parameter highly sensitive to protein conformation-assessed by EPR in conjunction with a protein specific spin al changes.Because the W/S ratio consistently decreases label,Mal-6.The motion of covalently bound Mal-6is with oxidative insult[13,22,24,28]and inversely correlates dependent upon protein structure and can be classified into with protein oxidation,this parameter has been used as a two main environments:weakly(W)-and strongly(S)-measure of indirect protein oxidation[6].Analysis of immobilized.Weakly immobilized Mal-6residues are protein conformations reveals no difference in untreated manifested as narrow lines in the EPR spectrum,whereas apoE3and apoE4synaptosomes;however,the W/S ratiouderback et al./Brain Research924(2002)90–97significantly decreases(P,0.05)in apoE4synaptosomesafter a2-h incubation with A b(1–42),but remains un-changed in apoE3synaptosomes(Fig.4).This suggeststhat apoE4synaptosomes are more vulnerable to proteinoxidation than apoE3synaptosomes.To further investigatethis suggestion,protein carbonyls were measured insynaptosomes after treatment with A b(1–42).Proteincarbonyls arise from free radical oxidation of amino acidside chains or protein backbone and can also result fromnucleophilic addition reactions with products of lipidoxidation,e.g.HNE[6].Measurement of protein carbonyllevels in synaptosomes indicates no difference amongapoE alleles at basal levels;however,after a3-h treatmentwith A b(1–42),protein carbonyls increase in an allele-dependent manner and similar to the trend reported forROS generation and lipid oxidation,i.e.e2,e3,e4.Protein carbonyls are significantly increased(P,0.05)in Fig.4.A b(1–42)induces protein conformational changes in apoE4,butapoE3and apoE4synaptosomes,but not in apoE2synapto-not apoE3,synaptosomal membranes.Changes in the motion of Mal-6spin labeled proteins are due to protein conformational changes and are somes(Fig.5).These data suggest that apoE modulates manifested as changes in the W/S ratio.Because decreased W/S ratios A b-induced protein oxidation,and that the e4allele of correlate with increased protein oxidation,decreases in the W/S ratio are apoE is less protective than either the e2or e3alleles.an indirect measure of protein oxidation.After a2-h incubation at378Cwith10m M A b(1–42),a decrease in the W/S ratio occurs in apoE4synaptosomal proteins that is not observed in apoE3synaptosomes,suggesting that apoE4synaptosomes have an increased susceptibility to 4.DiscussionA b(1–42)-induced protein oxidation.Data are the mean and S.E.M.(n53;*P,0.05,paired t-test).In the present study,the roles of the three alleles ofapoE in modulating oxidative damage to synaptosomesinduced by A b(1–42)was analyzed.Oxidative damage isthought by many to play a causative role in the neurode-generation in AD.Overproduction of A b is characteristicof AD[47]and,based upon ROS generation and oxidativecapabilities of these peptides[3,7,14–16,56],may berelated to the oxidative damage present in AD brain[5].Further,the e4allele of apoE is a risk factor for AD andcorrelates with increased oxidative damage in AD brain[38,43,44,52].Our data are consistent with these two ideasin that A b-induced oxidation is consistently greatest insynaptosomes containing the e4allele of apoE.Thissupports a role for apoE in the modulation of A b-inducedoxidative damage and also the idea that the e4allele ofapoE may be less effective in this role than either the e2ore3alleles.The concept that apoE plays a role as an antioxidant hasbeen proposed[37].In the absence of apoE,mice arevulnerable to oxidative insults such as stroke[26],headinjury[9],or A b-peptides[20,28],and levels of lipidperoxidation products increase[20,28,39,42,45].Oxidativedamage and specifically lipid peroxidation products arecapable of inducing neuron death[4,19,23],which mayexplain the neurodegeneration evident in the absence of Fig.5.A b(1–42)-induced protein carbonyl formation in synaptosomalmembranes is modulated by apoE in an allele-dependent manner.Protein apoE[33].Interestingly,both neurodegeneration and lipid carbonyls are markers for protein oxidation and increase in synaptosomes peroxidation products in apoE deficient mice are amelior-upon treatment with A b-peptides.Allele-specific increases in protein ated by infusion or transfection with apoE[32,53],sup-oxidation correlate with increases in ROS and lipid oxidation and suggest porting both neurorestorative and scavenger roles for apoE. that the e4allele is more vulnerable than e2or e3to A b-induced proteinThefirst report of apoE as an antioxidant described an oxidation.Data are the mean and S.E.M.(n54–6;*P,0.05vs.untreatedsynaptosomes,paired t-test).allele-dependency to these effects in the order of e2.e3.uderback et al./Brain Research924(2002)90–9795 e4[37].Additional studies have confirmed that models Acknowledgementsexpressing the e4allele of apoE are more susceptible thanthe e3allele to kainic acid[8]or stroke[48],injuries in This work was supported in part by grants from NIH which oxidative damage is known to play a role[10,36].(AG-10836;AG-05119;AG-12423)to D.A.B.and(AG-Our data are consistent with these reports in that A b-12981;NS-32221)to M.S.K.induced oxidative damage correlates to apoE allele in theorder e2,e3,e4(i.e.increases in oxidative markerscorrelate with decreased antioxidant potential of respective ReferencesapoE allele).This result supports the decreased antioxidantcapability of the e4allele.Increases in ROS formation and[1]U.Beffert,J.S.Cohn,C.Petit-Turcotte,M.Tremblay,N.Aumont, protein and lipid oxidation follow this trend with ROS,C.Ramassamy,J.Davignon,J.Poirier,Apolipoprotein E andbeta-amyloid levels in the hippocampus and frontal cortex of TBARS,and protein conformational changes being sig-Alzheimer’s disease subjects are disease-related and apolipoprotein nificantly altered in apoE4synaptosomes as compared toE genotype dependent,Brain Res.843(1999)87–94.apoE3synaptosomes.[2]U.Beffert,N.Aumont,D.Dea,S.Lussier-Cacan,J.Davignon,J. An alternative explanation of the apoE allele-specific Poirier,Apolipoprotein E isoform-specific reduction of extracellular modulation of A b(1–42)-induced oxidation to synapto-amyloid in neuronal cultures,Mol.Brain Res.68(1999)181–185.[3]C.Behl,J.B.Davis,R.Lesley,D.Schubert,Hydrogen peroxide somes invokes the role of apoE in the clearance andmediates amyloid beta protein toxicity,Cell77(1994)817–827. catabolism of A b.ApoE binds A b in an allele-specific[4]A.Bruce-Keller,Y.J.Li,M.A.Lovell,P.J.Kraemer,D.S.Gary,R.R. fashion[25];moreover,the clearance of apoE/A b com-Brown,W.R.Markesbery,M.P.Mattson,4-Hydroxynonenal,a plexes is allele-dependent,based upon their respective product of lipid peroxidation,damages cholinergic neurons and affinities for the LRP receptor[2,18,41,57].Levels of A b impairs visuospatial memory in rats,J.Neuropathol.Exp.Neurol.57(1998)257–267.inversely correlate to levels of apoE in the AD brain in an[5]D.A.Butterfield,J.Drake,C.Pocernich,A.Castegna,Evidence of allele-dependent fashion,suggesting that in the absence ofoxidative damage in Alzheimer’s disease brain:central role of apoE,A b accumulates in the brain[1].Competitiveamyloid b-peptide,Trends Mol.Med.7(2001)548–554. inhibitors of LRP enhance A b toxicity and negate protec-[6]D.A.Butterfield,E.R.Stadtman,Protein oxidation processes in the tive effects of the e3allele of apoE,presumably by aging brain,Adv.Cell Aging Gerontol.2(1997)161–191.[7]D.A.Butterfield,K.Hensley,M.Harris,M.Mattson,J.Carney, inhibiting the clearance of A b[17,54].Thus,removal ofBeta-amyloid peptide free radical fragments initiate synaptosomal extracellular A b by an LRP-mediated pathway may be alipoperoxidation in a sequence-specific fashion:implications to mechanism to prevent protein and lipid oxidation inducedAlzheimer’s disease,mun.200(1994) by A b,and may explain the differential increases in710–715.oxidative markers reported in the current study.Our data[8]M.Buttini,M.Orth,S.Bellosta,H.Akeefe,R.E.Pitas,T.Wyss-Coray,L.Mucke,R.W.Mahley,Expression of human apolipoprotein are consistent with this hypothesis in that increasedE3or E4in the brains of apoE2/2mice:isoform-specific effects markers for protein and lipid oxidation correlate withon neurodegeneration,J.Neurosci.19(1999)4867–4880.apoE-mediated uptake of A b,i.e.delaying the removal of[9]Y.Chen,L.Lomnitski,D.M.Michaelson,E.Shohami,Motor andA b results in increased oxidative damage.Additionally,by cognitive deficits in apolipoprotein E-deficient mice after closed simply binding to A b,apoE may alter its oxidative head injury,Neuroscience80(1997)1255–1262.[10]J.T.Coyle,P.Puttfarcken,Oxidative stress,glutamate,and neurode-potential,before receptor-mediated clearance takes place.generative disorders,Science262(1993)689–695.Thus,in the interpretation of the presented data,these[11]H.Esterbauer,H.Zollner,R.J.Schaur,Chemistry and biochemistry mechanisms for reducing A b-induced oxidative injuryof4-hydroxynonenal,malonaldehyde and related aldehydes,Free cannot be overlooked.However,apoE also modulates Radic.Biol.Med.11(1991)81–128.neuronal injury when A b is not involved(e.g.stroke or[12]K.E.Gridley,P.S.Green,J.W.Simpkins,Low concentrations ofestradiol reduce beta-amyloid(25–35)-induced toxicity,lipid perox-H O)or when apoE uptake is inhibited[8,37],suggesting22idation and glucose utilization in human SK-N-SH neuroblastoma other mechanisms of apoE-mediated protection may becells,Brain Res.778(1997)158–165.responsible for neuroprotection.[13]N.C.Hall,J.M.Carney,M.S.Cheng,D.A.Butterfield,Ischemia/ Overall,our data suggest that apoE can modulate A b(1–reperfusion induced changes in membrane proteins and lipids of 42)-induced oxidative damage in synaptic terminals.Re-gerbil cortical synaptosomes,Neuroscience64(1995)81–89.[14]M.E.Harris,K.Hensley, D.A.Butterfield,R.A.Leedle,J.M. gardless of whether these results are due to an antioxidant-Carney,Direct evidence of oxidative injury by the Alzheimer’s related or A b clearance-related mechanism,it is clear thatamyloid peptide in cultured hippocampal neurons,Exp.Neurol.131 apoE is important in maintaining synaptic integrity by(1995)193–202.modulating oxidative damage.Allele-dependent(e2,e3,[15]K.Hensley,J.M.Carney,M.P.Mattson,M.Aksenova,M.E.Harris, e4)increases of several oxidative markers in synaptosomes J.F.Wu,R.A.Floyd, D.A.Butterfield,A model for b-amyloidaggregation and neurotoxicity based on free radical generation by suggest that the e4allele of apoE is less effective inthe peptide:relevance to Alzheimer disease,Proc.Natl.Acad.Sci. modulating this damage.Thesefindings are particularlyUSA91(1994)3270–3274.relevant to Alzheimer’s disease where A b is overproduced,[16]X.Huang,C.Atwood,M.Hartshorn,G.Multhaup,L.Goldstein,R. oxidative damage is evident,and synapses are lost,and for Scarpa,M.Cuajungco,D.Gray,J.Lim,R.Moir,R.Tanzi,A.Bush, which apoE4is a risk factor.The A b peptide of Alzheimer’s disease directly produces hydrogenuderback et al./Brain Research924(2002)90–97peroxide through metal ion reduction,Biochemistry38(1999)apoptosis-related events in synapses and dendrites,Brain Res.807 7609–7616.(1998)167–176.[17]J.Jordan,M.F.Galindo,ler,C.Reardon,G.Getz,Du,[36]E.K.Michaelis,Molecular biology of glutamate receptors in theIsoform-specific effect of apolipoprotein E on cell survival and central nervous system and their role in excitotoxicity,oxidative beta-amyloid-induced toxicity in rat hippocampal pyramidal neuro-stress and aging,Prog.Neurobiol.54(1998)369–415.nal cultures,J.Neurosci.18(1998)195–204.[37]M.Miyata,J.D.Smith,Apolipoprotein E allele-specific antioxidant [18]D.E.Kang,C.Pietrzik,L.Baum,N.Chevallier,D.E.Merriam,M.activity and effects on cytotoxicity by oxidative insults and beta-Kounnas,S.L.Wagner,J.C.Troncoso,C.H.Kawas,R.Katzman,amyloid peptides,Nat.Genet.14(1996)55–61.E.H.Koo,Modulation of amyloid b-protein clearance and Alzheim-[38]K.S.Montine,E.Reich,M.D.Neely,K.R.Sidell,S.J.Olson,W.R.er’s disease susceptibility by the LDL receptor-related protein Markesbery,T.J.Montine,Distribution of reducible4-hydroxy-pathway,J.Clin.Invest.106(2001)1159–1166.nonenal adduct immunoreactivity in Alzheimer disease is associated [19]J.N.Keller,M.P.Mattson,Roles of lipid peroxidation in modulation with APOE genotype,J.Neuropathol.Exp.Neurol.57(1998)of cellular signaling pathways,cell dysfunction,and death in the415–425.nervous system,Rev.Neurosci.9(1998)105–116.[39]T.J.Montine,K.S.Montine,S.J.Olson,D.G.Graham,L.J.Roberts, [20]J.N.Keller,uderback,D.A.Butterfield,M.Kindy,J.Yu,J.D.Morrow,M.F.Linton,S.Fazio,L.L.Swift,Increased cerebralW.R.Markesbery,Amyloid beta-peptide effects on synaptosomes cortical lipid peroxidation and abnormal phospholipids in aged from apolipoprotein E-deficient mice,J.Neurochem.74(2000)homozygous apoE-deficient C57BL/6J mice,Exp.Neurol.158 1579–1586.(1999)234–241.[21]J.N.Keller,Z.Pang,J.W.Geddes,J.G.Begley,A.Germeyer,G.[40]M.D.Neely,K.R.Sidell,D.G.Graham,T.J.Montine,The lipidWaeg,M.P.Mattson,Impairment of glucose and glutamate transport peroxidation product4-hydroxynonenal inhibits neurite outgrowth, and induction of mitochondrial oxidative stress and dysfunction in disrupts neuronal microtubules,and modifies cellular tubulin,J.synaptosomes by amyloid beta-peptide:role of the lipid peroxidation Neurochem.72(1999)2323–2333.product4-hydroxynonenal,J.Neurochem.69(1997)273–284.[41]J.Poirier,Apolipoprotein E and Alzheimer’s disease:a role in [22]T.Koppal,J.Drake,S.Yatin, B.Jordan,S.Varadarajan,L.amyloid catabolism,Ann.NY Acad.Sci.924(2000)80–90.Bettenhausen,D.A.Butterfield,Peroxynitrite-induced alterations in´[42]D.Pratico,J.Rokach,R.K.Tangirala,Brains of aged apolipoproteinsynaptosomal membrane proteins:insight into oxidative stress in E-deficient mice have increased levels of F2-isoprostanes,in vivo Alzheimer’s disease,J.Neurochem.72(1999)310–317.markers of lipid peroxidation,J.Neurochem.73(1999)736–741.[23]I.Kruman,A.J.Bruce-Keller,D.Bredesen,G.Waeg,M.P.Mattson,[43]C.Ramassamy,D.Averill,U.Beffert,S.Bastianetto,L.Theroux,S.Evidence that4-hydroxynonenal mediates oxidative stress-induced Lussier-Cacan,J.S.Cohn,Y.Christen,J.Davignon,R.Quirion,J.neuronal apoptosis,J.Neurosci.17(1997)5089–5100.Poirer,Oxidative damage and protection by antioxidants in the [24] Fontaine,J.W.Geddes, A.Banks, D.A.Butterfield,3-frontal cortex of Alzheimer’s disease is related to the apolipoproteinNitropropionic acid induced in vivo protein oxidation in striatal and E genotype,Free Radic.Biol.Med.27(1999)544–553.cortical synaptosomes:insights into Huntington’s disease,Brain[44]C.Ramassamy, D.Averill,U.Beffert,L.Theroux,S.Lussier-Res.858(2000)356–362.Cacan,J.S.Cohn,Y.Christen,A.Schoofs,J.Davignon,J.Poirer,[25]Du,M.T.Falduto,A.M.Manelli,C.A.Reardon,G.S.Getz,Oxidative insults are associated with apolipoprotein E genotype inD.E.Frail,Isoform-specific binding of apolipoprotein E to beta-Alzheimer’s disease brain,Neurobiol.Dis.7(2000)23–37.amyloid,J.Biol.Chem.269(1994)23403–23406.[45]C.Ramassamy,P.Krzywkowski,D.Averill,S.Lussier-Cacan,L.[26]skowitz,H.Sheng,R.D.Bart,K.A.Joyner,A.D.Roses,D.S.Theroux,Y.Christen,J.Davignon,J.Poirier,Impact of apoEWarner,Apolipoprotein E-deficient mice have increased suscep-deficiency on oxidative insults and antioxidant levels in the brain, tibility to focal cerebral ischemia,J.Cereb.Blood Flow Metab.17Mol.Brain Res.86(2001)76–83.(1997)753–758.[46]A.D.Roses,Apolipoprotein E alleles as risk factors in Alzheimer’s [27]skowitz,K.Horsburgh,A.D.Roses,Apolipoprotein E and disease,Annu.Rev.Med.47(1996)387–400.the CNS response to injury,J.Cereb.Blood Flow Metab.18(1998)[47]D.J.Selkoe,Alzheimer’s disease:genes,proteins,and therapy, 465–471.Physiol.Rev.81(2001)741–766.[28]uderback,J.Hackett,J.N.Keller,S.Varadarajan,L.Szweda,[48]H.Sheng,skowitz,E.Bennett,D.E.Schmechel,R.D.Bart,M.S.Kindy,D.A.Butterfield,Apoliprotein E knock-out synapto- A.M.Saunders,R.D.Pearlstein,A.D.Roses,D.S.Warner,Apolipo-somes are vulnerable to A b(1–40)-induced oxidative injury,Bio-protein E isoform-specific differences in outcome from focal is-chemistry40(2001)2548–2554.chemia in transgenic mice,J.Cereb.Blood Flow Metab.18(1998) [29]R.Mahley,Apolipoprotein E:cholesterol transport protein with361–366.expanding role in cell biology,Science240(1988)622–630.[49]R.Subramaniam,F.Roediger,B.Jordan,M.P.Mattson,J.N.Keller, [30]R.Mahley,S.C.Rall,Apolipoprotein E:far more than a lipid G.Waeg,D.A.Butterfield,The lipid peroxidation product,4-hy-transport protein,Annu.Rev.Genomics Hum.Genet.1(2000)droxy-2-trans-nonenal,alters the conformation of cortical synap-507–537.tosomal membrane proteins,J.Neurochem.69(1997)1161–1169.[31]R.J.Mark,M.A.Lovell,W.R.Markesbery,K.Uchida,M.P.Mattson,[50]P.M.Sullivan,H.Mezdour,Y.Aratani,C.Knuff,J.Najib,R.L.A role for4-hydroxynonenal,an aldehydic product of lipid peroxi-Reddick,S.H.Quarfordt,N.Madea,Targeted replacement of thedation,in disruption of ion homeostasis and neuronal death induced mouse apolipoprotein E gene with the common human apoE3allele by amyloid beta-peptide,J.Neurochem.68(1997)255–264.enhances diet-induced hypercholesterolemia and atherosclerosis,J.[32]E.Masliah,W.Samuel,I.Veinbergs,M.Mallory,M.Mante,T.Biol.Chem.272(1997)17972–17980.Saitoh,Neurodegeneration and cognitive impairment in apoE-de-[51]P.M.Sullivan,H.Mezdour,S.H.Quarfordt,N.Madea,Type III ficient mice is ameliorated by infusion of recombinant apoE,Brain hyperlipoproteinemia and spontaneous atherosclerosis in mice re-Res.751(1997)307–314.sulting from gene replacement of mouse apoE with human apoE2,J.[33]E.Masliah,M.Mallory,N.Ge,M.Alford,I.Veinbergs,A.D.Roses,Clin.Invest.102(1998)130–135.Neurodegeneration in the central nervous system of apoE-deficient[52]A.Takaoma, F.Miyatake,S.Matsuno,K.Ishii,S.Nagase,N.mice,Exp.Neurol.136(1995)107–122.Sahara,S.Ono,H.Mori,K.Wakabayashi,S.Tsuji,H.Takahashi,S.[34]R.T.Matthews,M.F.Beal,Increased3-nitrotyrosine in brains of Shoji,Apolipoprotein E allele-dependent antioxidant activity inapoE-deficient mice,Brain Res.718(1996)181–184.brains with Alzheimer’s disease,Neurology54(2000)2319–2321.[35]M.P.Mattson,J.Partin,J.G.Begley,Amyloid beta-peptide induces[53]R.K.Tangirala, D.Pratico,G.A.FitzGerald,S.Chun,K.。