• No results found

HDL is redundant for adrenal steroidogenesis in LDLR knockout mice with a human-like lipoprotein profile

N/A
N/A
Protected

Academic year: 2021

Share "HDL is redundant for adrenal steroidogenesis in LDLR knockout mice with a human-like lipoprotein profile"

Copied!
7
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

to adrenocortical cells under high steroidogenic pressure conditions, like stress. More specifi cally, probucol-induced lowering of both HDL-cholesterol and LDL-cholesterol levels in mice is associated with a >55% decrease in the glucocorticoid response to endotoxemia ( 1 ). Studies in LCAT and APOA1 knockout mice, respectively, have indi- cated that a specifi c decrease in plasma HDL-cholesterol levels is associated with a 25–50% decrease in the maximal adrenal glucocorticoid output ( 2, 3 ). Moreover, disrup- tion of (adrenal-specifi c) HDL receptor function in mice is associated with a 40–50% decrease in the adrenocortical steroidogenic capacity ( 4, 5 ).

Male carriers of functional mutations in the HDL bio- genesis genes, ABCA1 and LCAT, display a decrease in the 24 h urinary excretion rate of adrenal-derived steroids ( 6 ).

However, basal and stimulated plasma cortisol levels are similar in HDL-defi cient male ABCA1 and LCAT mutation carriers and their normolipidemic controls ( 6 ). Further- more, both the urinary glucocorticoid excretion rate and cortisol response to corticotropin are unaltered in females with genetically low HDL ( 7 ). The presence of relatively low HDL-cholesterol levels is, thus, in striking contrast to what is observed in mice: not consistently associated with glucocorticoid insuffi ciency in humans.

In vitro studies have suggested that both HDL and APOB-containing lipoproteins, i.e., VLDL and LDL, can theoretically supply cholesterol to adrenocortical cells ( 8–

11 ). Importantly, human subjects exhibit a markedly dif- ferent lipoprotein profi le as compared with mice. The majority of cholesterol in humans is carried by LDL, while the murine lipoprotein profi le is characterized by rela- tively low to absent levels of cholesterol associated with VLDL/LDL in the context of normal HDL-cholesterol lev- els. The relative importance of HDL-associated cholesterol as steroidogenic substrate can, thus, hypothetically be dif- ferent between these two specifi c species due to the fact that human plasma, as compared with murine plasma, contains additional potential cholesterol sources, i.e., LDL Abstract The contribution of HDL to adrenal steroidogen-

esis appears to be different between mice and humans. In the current study, we tested the hypothesis that a difference in lipoprotein profi le may be the underlying cause. Hereto, we determined the impact of HDL defi ciency on the adrenal glucocorticoid output in genetically modifi ed mice with a human-like lipoprotein profi le. Genetic deletion of APOA1 in LDL receptor (LDLR) knockout mice was associated with HDL defi ciency and a parallel increase in the level of cho- lesterol associated with nonHDL fractions . Despite a com- pensatory increase in the adrenal relative mRNA expression levels of the cholesterol synthesis gene, HMG-CoA reduc- tase, adrenals from APOA1/LDLR double knockout mice were severely depleted of neutral lipids, as compared with those of control LDLR knockout mice. However, basal cor- ticosterone levels and the adrenal glucocorticoid response to stress were not different between the two types of mice.

In conclusion, we have shown that HDL is not critical for proper adrenal glucocorticoid function when mice are pro- vided with a human-like lipoprotein profi le. Our fi ndings provide the fi rst experimental evidence that APOB-contain- ing lipoproteins may facilitate adrenal steroidogenesis, in an LDLR-independent manner, in vivo in mice. —Hoekstra, M., and M. Van Eck. HDL is redundant for adrenal steroido- genesis in LDLR knockout mice with a human-like lipopro- tein profi le. J. Lipid Res. 2016. 57: 631–637.

Supplementary key words steroid hormones • cholesterol • high den- sity lipoprotein • low density lipoprotein • gene expression • corticoste- rone • low density lipoprotein receptor

Glucocorticoids are produced from the common ste- roidogenic precursor, cholesterol. It is generally accepted that under basal conditions suffi cient amounts of choles- terol are acquired from endogenous synthesis by the adre- nals. However, it has become evident from studies in genetically modifi ed mice that lipoproteins, in particular HDLs, are important for delivering cholesterol substrate

This work was supported by Dutch Heart Foundation Grants 2008T070 and 2012T080 awarded to M.H. and VICI Grant 91813603 from The Netherlands Organization for Scientifi c Research awarded to M.V.E . M.V.E. is an Estab- lished Investigator of the Dutch Heart Foundation (Grant 2007T056 ).

Manuscript received 22 December 2015 and in revised form 16 February 2016.

Published, JLR Papers in Press, February 18, 2016 DOI 10.1194/jlr.M066019

HDL is redundant for adrenal steroidogenesis in LDLR knockout mice with a human-like lipoprotein profi le

Menno Hoekstra 1 and Miranda Van Eck

Division of Biopharmaceutics, Cluster BioTherapeutics, Leiden Academic Centre for Drug Research , Gorlaeus Laboratories, 2333CC Leiden, The Netherlands

Abbreviations: DKO, double knockout; LDLR, LDL receptor;

SKO, single knockout . 1

To whom correspondence should be addressed.

e-mail: Hoekstra@lacdr.leidenuniv.nl

at Walaeus Library / BIN 299, on February 1, 2017www.jlr.orgDownloaded from

(2)

stained with hematoxylin (Sigma) and Oil red O (Sigma) for lipid visualization.

Real-time quantitative PCR

Gene expression analysis was performed essentially as de- scribed ( 16 ). Equal amounts of RNA were reverse transcribed and real-time quantitative PCR analysis was subsequently exe- cuted on the cDNA using an ABI Prism 7500 apparatus (Applied Biosystems, Foster City, CA) according to the manufacturer’s in- structions. Acidic ribosomal phosphoprotein P0 (36B4), GAPDH, succinate dehydrogenase complex subunit A fl avoprotein (SDHA), peptidylprolyl isomerase A (PPIA), ␤ -2-microglobulin (B2M), transferrin receptor (TFRC), and ␤ -actin (ACTB) were used as housekeeping genes for normalization.

Data analysis

Statistical analysis was performed using GraphPad InStat soft- ware (San Diego, CA, http://www.graphpad.com). Normality of the experimental groups was confi rmed using the method of Kol- mogorov and Smirnov. The signifi cance of differences was calcu- lated using a two-tailed unpaired t -test or two-way ANOVA where appropriate. Probability values less than 0.05 were considered signifi cant.

RESULTS

Regular chow diet-fed LDLR knockout mice exhibit a highly similar lipoprotein profi le to that observed in nor- molipidemic humans ( 13 ). To investigate whether the contribution of HDL to adrenal glucocorticoid output is different in mice with a human-like lipoprotein profi le, we determined the impact of genetic HDL defi ciency in mice on a LDLR knockout genetic background. Hereto, HDL- defi cient APOA1 knockout mice were crossbred with LDLR SKO mice to generate the respective APOA1/LDLR DKO mice.

As can be appreciated from Fig. 1A , plasma free and to- tal cholesterol levels did not signifi cantly differ between regular chow diet-fed male DKO and SKO mice. How- ever, lipoprotein distribution analysis on pooled plasma ( Fig. 1B ) revealed that DKO mice exhibited a highly simi- lar reduction in plasma HDL-cholesterol levels ( ⫺ 65%), as previously noted in APOA1 knockout mice on a wild- type background ( 3 ). DKO mice showed a parallel 89%

increase in levels of cholesterol associated with VLDL particles, as compared with their respective APOA1-con- taining SKO controls ( Fig. 1B ). As a result, the plasma nonHDL-cholesterol over HDL-cholesterol ratio was, thus, markedly higher in DKO mice as compared with SKO mice ( Fig. 1C ).

Previous studies by the group of Dr. Mary Sorci-Thomas have suggested that, after a short-term (4 h) fasting pe- riod, the adrenals of DKO mice are severely depleted of cholesterol esters, despite the fact that DKO mice still carry ⵑ 30% of the normal amount of HDL-associated choles- terol in APOE-enriched HDL particles ( 17 ). Quantifi cation of the adrenal lipid stores revealed that both free choles- terol ( ⫺ 21%; P = 0.002) and cholesterol ester ( ⫺ 51%; P = 0.005) levels were also markedly lower in the adrenals of our DKO mice, as compared their SKO controls, after an and VLDL. To provide experimental proof for this hy-

pothesis in the current study, we determined the impact of HDL defi ciency on the adrenal glucocorticoid output in genetically modifi ed mice that contain a human-like lipo- protein profi le.

MATERIALS AND METHODS

Animals

APOA1 knockout mice lacking a functional APOA1 protein ( 12 ) were provided on a hyperlipidemic LDL receptor (LDLR) knockout background ( 13 ) by Dr. J. A. Kuivenhoven from the Amsterdam Medical Center (Amsterdam, The Netherlands).

These APOA1/LDLR double knockout (DKO) mice were subse- quently inbred to maintain an in-house colony.

Male APOA1/LDLR DKO (N = 14) and control LDLR single knockout (SKO) mice (N = 14) were maintained on a regular chow diet. Throughout the experiment, both types of mice were housed in the same climate-controlled stable with a 12 h/12 h dark-light cycle and handled identically. At 4 months of age, ad libitum-fed age-matched mice (N = 6 per genotype) were bled at 9:00 AM from the tail to obtain a basal plasma corticosterone measurement. Subsequently, these six mice per genotype were injected intraperitoneally with a sublethal dose (50 ␮ g/kg) of li- popolysaccharide from Salmonella minnesota R595 and euthanized 2 h later to measure the maximum endotoxemia-related plasma corticosterone response ( 1, 14 ). The remaining mice (N = 8 per genotype) were subjected to overnight fasting by food depriva- tion from 5:00 PM onwards. At 9:00 AM the next morning, mice were bled via the tail for fasting plasma corticosterone and blood glucose measurements. After anesthesia by subcutaneous injec- tion with a mix of 70 mg/kg bodyweight xylazine, 1.8 mg/kg bodyweight atropine, and 350 mg/kg bodyweight ketamine, mice were bled via retro-orbital bleeding, euthanized, and sub- jected to whole body perfusion with ice-cold PBS. Adrenals were collected free of surrounding fat, weighed, and stored at ⫺ 20°C or fi xed overnight in 3.7% neutral-buffered formalin solution (Formalfi xx; Shandon Scientifi c Ltd, UK). All animal work was approved by the Leiden University Animal Ethics Committee and performed in compliance with the Dutch government guidelines and Directive 2010/63/EU of the European Parliament.

Blood and plasma analyses

Corticosterone levels in tail blood plasma were determined us- ing the corticosterone 3 H RIA kit from ICN Biomedicals accord- ing to the protocol from the supplier. Plasma concentrations of free cholesterol and cholesterol esters were determined using enzymatic colorimetric assays. The cholesterol distribution over the different lipoproteins in plasma was analyzed by fraction- ation of 30 ␮ l pooled plasma of each mouse genotype using a Superose 6 column (3.2 × 30 mm, Smart-system; Pharmacia). Total cholesterol content of the effl uent was determined using enzymatic colorimetric assays. Blood glucose levels were routinely measured using a calibrated Accu-Check glucometer (Roche Diagnostics, Almere, The Netherlands)

Adrenal lipid composition and histology

Lipids from adrenals were extracted using the method of Bligh and Dyer ( 15 ). After dissolving the lipids in 1% Triton X-100, the contents of free cholesterol and cholesterol esters were determined using enzymatic colorimetric assays and expressed as micrograms per milligram of protein. Seven micrometer cryosections were prepared on a Leica CM3050-S cryostat. Cryosections were routinely

at Walaeus Library / BIN 299, on February 1, 2017www.jlr.orgDownloaded from

(3)

neutral lipids in their adrenal cortex. In contrast, an equally low extent of lipid accumulation was microscopically de- tected within cortical cells of DKO adrenals ( Fig. 2B ), as previously noted in glucocorticoid insuffi cient LCAT knock- out mice and probucol-treated C57BL/6 mice ( 1, 2 ).

overnight fast ( Fig. 2A ), suggesting that the adrenal lipid depletion effect associated with APOA1 defi ciency is independent of the metabolic/stress state. Oil red O stain- ing of adrenal cryostat sections further verifi ed adrenal lipid depletion. Adrenals from SKO mice had abundant

Fig. 1. Plasma free and total cholesterol levels (A), the cholesterol distribution over the different lipoprotein fractions (B), and the plasma nonHDL- to HDL-cholesterol ratio (C) in age-matched male APOA1/LDLR DKO and LDLR SKO mice.

Fig. 2. A: Adrenal free cholesterol and cholesterol ester levels in APOA1/LDLR DKO and LDLR SKO mice. B: Representative images of Oil red O-stained adrenal sections showing neutral lipid depletion in the cortex of DKO mice. C: Adrenal relative gene expression levels as measured by quantitative PCR. ** P < 0.01, *** P < 0.001 versus SKO. HMGCR, HMG-CoA reductase .

at Walaeus Library / BIN 299, on February 1, 2017www.jlr.orgDownloaded from

(4)

signifi cant 6.6-fold increase ( P < 0.001 vs. basal) in circulat- ing corticosterone levels in SKO mice, as anticipated.

Strikingly, corticosterone levels were virtually identical in both groups of fasted mice (256 ± 21 ng/ml for DKO vs.

269 ± 15 ng/ml for SKO; P > 0.05). HDL defi ciency thus does not seem to be associated with glucocorticoid insuf- fi ciency in mice with a human-like lipoprotein. In agree- ment with a normal metabolic glucocorticoid action in HDL-defi cient mice, DKO mice did not display hypoglyce- mia, as compared with SKO mice under fasting conditions ( Fig. 3B ). The induction of endotoxemia is associated with a concomitant rise in the plasma level of glucocorticoids ( 21, 22 ). In further support of a similar maximal steroido- genic capacity of the adrenals in the two types of mice, equally high levels of corticosterone ( ⵑ 250 ng/ml; Fig.

3A ) were detected in the plasma of SKO and DKO mice after induction of endotoxemia through injection of a sub- lethal dose of lipopolysaccharide.

DISCUSSION

In the current study, we tested the hypothesis that a dif- ference in lipoprotein profi le between mice and humans can explain the relative importance of HDL-cholesterol as substrate for adrenal steroidogenesis.

A 70% reduction in plasma HDL-cholesterol levels in APOA1 SKO mice is associated with a severe depletion of adrenal cholesterol ester stores and a concomitant impair- ment of the adrenal glucocorticoid response to stress ( 3 ).

The APOE-rich HDL particles remaining in these mice ( 23 ) are apparently not able to compensate for the lack of An effi cient feedback system exists that modulates the

expression of genes involved in cholesterol synthesis and uptake in response to changes in intracellular cholesterol levels [reviewed by Sato and Takano ( 18 )]. Quantitative real-time PCR was employed to uncover possible compen- satory gene regulation. No change was noted, as compared with SKO adrenals, in the relative mRNA expression level of the HDL receptor, SR-BI, in DKO adrenals ( Fig. 2C ). In addition, genetic APOA1 defi ciency was not associated with a difference in relative mRNA expression levels of hormone-sensitive lipase (HSL), ACAT-1, and steroido- genic acute regulatory protein (STAR) that are respec- tively involved in the de- and re-esterifi cation of cholesterol and intracellular mobilization of cholesterol to the ste- roidogenic pathway ( Fig. 2C ). However, we did observe a marked increase (425%; P < 0.001; Fig. 2C ) in the mRNA expression of the enzyme, HMG-CoA reductase, in DKO adrenals. It thus appears that, in a human-like lipoprotein context, HDL defi ciency in mice is associated with deple- tion of adrenal cholesterol stores despite a compensatory increase in intra-adrenal cholesterol synthesis.

Levels of the primary glucocorticoid, corticosterone, were measured in plasma under basal and stressed condi- tions to verify whether the depletion of adrenal choles- terol esters also executed a negative impact on the overall steroid output. In line with the general notion that lipo- protein-derived cholesterol is not required for the synthe- sis of glucocorticoids under low steroidogenic conditions, plasma corticosterone levels were similar in nonstressed ad libitum-fed SKO and DKO mice ( Fig. 3A ). Food depri- vation is a powerful inducer of an adrenal glucocorticoid response in mice ( 19, 20 ). Overnight fasting resulted in a

Fig. 3. A: Plasma corticosterone levels in APOA1/LDLR DKO and LDLR SKO mice measured in the basal state, in response to overnight food deprivation (fasting), and after injection with lipopolysaccharide (endo- toxemia). B: Plasma glucose levels as measured in the fasted state. # P < 0.001 versus respective basal values.

at Walaeus Library / BIN 299, on February 1, 2017www.jlr.orgDownloaded from

(5)

by these circulating lipid/protein complexes. Consider- able evidence is, however, present that SR-BI is also able to facilitate the uptake of cholesterol from APOB-containing lipoproteins. Initial in vitro studies by Swarnakar et al.

( 29 ) and Stangl, Hyatt, and Hobbs ( 30 ) showed that mu- rine SR-BI is able to mediate the selective uptake of choles- terol esters from human LDL. Subsequent cell culture studies by Webb et al. ( 31 ) verifi ed a similar interaction of SR-BI with autologous mouse LDL. In support of a parallel role for SR-BI in APOB-containing lipoprotein cholesterol delivery in vivo, the removal from the blood circulation and tissue uptake of ␤ -migrating VLDL particles, LDL, and chylomicron-remnants has been shown to be signifi - cantly lower in mice lacking functional SR-BI expression ( 32–34 ). As a result, SR-BI knockout mice not only display increased plasma levels of HDL-cholesterol, but also ex- hibit an increase in the amount of cholesterol carried by APOB-containing lipoproteins ( 35 ), while plasma levels of APOB-containing lipoproteins are markedly lower in mice upon transgenic or adenoviral overexpression of SR-BI ( 36, 37 ). Adrenal glucocorticoid output is diminished in human subjects carrying a functional mutation in the SR- BI gene ( 38 ). Several heterozygote SR-BI P297S carriers actually show signs of adrenal dysfunction in spite of mark- edly increased plasma HDL-cholesterol levels ( 38 ). Given that, in the human situation, disruption of SR-BI function appears to be associated with a more extreme effect on the adrenal steroidogenic capacity than genetic lowering of HDL-cholesterol levels, it can be suggested that, in our current human-like lipoprotein setting, the impaired acquisi- tion of cholesterol from APOA1-containing HDL particles can be fully compensated by enhanced cholesterol synthe- sis combined with SR-BI-mediated delivery of cholesterol from APOB-containing lipoproteins to the adrenals.

Novel intervention strategies to reduce cardiovascular disease risk, such as proprotein convertase subtilisin/kexin type 9 (PCSK9) antibody treatment and statin/ezetimibe combination therapies, are aimed at reaching extremely low plasma LDL-cholesterol levels. No remarkable adre- nal-associated events have been reported in meta-analyses of anti-PCSK9 antibody ( 39 ) and statin/ezetimibe trials ( 40 ). This may, at fi rst sight, argue against our current working hypothesis that APOB-containing lipoproteins serve as primary cholesterol donors for steroidogenesis.

However, one should take into account that: 1 ) adrenal dysfunction may only become evident under stress condi- tions; and 2 ) in-depth adrenal function testing is not com- mon within these cardiovascular-oriented clinical trials.

As such, inclusion of the adrenocorticotropic hormone (ACTH) stimulation test, the standard method to assess the maximal adrenal cortisol response, in trial protocols may aid in validating our hypothesis in the human setting.

In light of our challenging concept, it is of interest to note that several case studies by Illingworth and colleagues ( 41–

43 ) have indicated that genetic LDL defi ciency (abetalipo- proteinemia) in humans is associated with subclinical adrenal insuffi ciency, as evident from an impaired ACTH- induced cortisol response and a lower urinary (free) corti- sol excretion rate.

cholesterol supplied by APOA1-containing particles for steroidogenesis. Although it cannot be excluded that the APOE-rich HDL is a poor substrate for adrenal cholesterol delivery, we anticipate that the adrenal cholesterol insuf- fi ciency observed in APOA1 SKO mice is primarily the re- sult of an overall too low amount of HDL particles being present in the circulation.

Genetic variations in the APOA1 gene have also been associated with HDL defi ciency in humans ( 24–26 ). How- ever, due to the limited number of subjects with genetic APOA1 defi ciency, the specifi c contribution of APOA1- containing HDL particles to adrenal steroidogenesis remains to be determined in the human setting. In the current study, we observed that APOA1 defi ciency in mice with a human-like lipoprotein, i.e., on a LDLR knockout (hyper- lipidemic) background, is associated with a similar 65%

reduction in plasma HDL-cholesterol levels and adrenal cholesterol depletion, as observed in APOA1 knockout mice on a wild-type (normolipidemic) background. In contrast, APOA1/LDLR DKO mice do not suffer from glucocorti- coid insuffi ciency, as their maximal glucocorticoid output is similar to that of HDL-containing single LDLR knockout controls. It thus appears that the presence of a human-like lipoprotein profi le alleviates the glucocorticoid insuffi - ciency associated with APOA1 defi ciency in mice.

Studies by Plump et al. ( 3 ) have suggested that adrenals from APOA1 knockout mice are still able to respond to stress, although to a minor extent as compared with those of wild-type mice, due to compensatory upregulation of pathways that are normally of minor importance, such as cholesterol uptake by the LDLR and de novo cholesterol synthesis. A 5-fold increase in the gene expression of HMG-CoA reductase in the adrenals of DKO mice was de- tected under fasting stress conditions, which suggests that de novo cholesterol synthesis is stimulated to compensate for the loss of HDL-cholesterol. In contrast to our DKO mice, HDL-defi cient LCAT knockout mice do display a di- minished adrenal glucocorticoid function despite a marked 6-fold increase in adrenal HMG-CoA reductase expression ( 2 ). From these combined fi ndings, it can be concluded that such a 5- to -6-fold increase in adrenal HMG-CoA reductase expression is, by itself, not suffi cient to overcome adrenal glucocorticoid insuffi ciency. All mice used in the current study did not express a functional LDLR, which excludes a compensatory role for LDLR-me- diated cholesterol acquisition by adrenals in DKO mice. In vitro studies by Kraemer et al. ( 27 ) have suggested that the LDLR is of negligible importance for acute steroidogene- sis by adrenocortical cells. Furthermore, the levels of corti- costerone measured in the fasting state in male LDLR knockout mice in the current experiment are almost identical to those found in male wild-type mice in our pre- vious studies ( 28 ). We therefore consider it highly unlikely that the impact of APOA1/HDL defi ciency on glucocorti- coid output in vivo is dependent on the adrenal LDLR genotype.

In our experimental setup, APOB-containing lipopro- teins could not be cleared by the LDLR, which is normally suggested to be the primary route of cholesterol delivery

at Walaeus Library / BIN 299, on February 1, 2017www.jlr.orgDownloaded from

(6)

through its roles in glucocorticoid production and hepatic clear- ance. J. Clin. Invest. 118 : 364 – 375 .

15 . Bligh , E. G. , and W. J. Dyer . 1959 . A rapid method of total lipid extraction and purifi cation. Can. J. Biochem. Physiol. 37 : 911 – 917 . 16 . Hoekstra , M. , J. K. Kruijt , M. Van Eck , and T. J. Van Berkel . 2003 .

Specifi c gene expression of ATP-binding cassette transporters and nuclear hormone receptors in rat liver parenchymal, endothelial, and Kupffer cells. J. Biol. Chem. 278 : 25448 – 25453 .

17 . Zabalawi , M. , S. Bhat , T. Loughlin , M. J. Thomas , E. Alexander , M. Cline , B. Bullock , M. Willingham , and M. G. Sorci-Thomas . 2003 . Induction of fatal infl ammation in LDL receptor and ApoA-I double-knockout mice fed dietary fat and cholesterol. Am. J. Pathol.

163 : 1201 – 1213 .

18 . Sato , R. , and T. Takano . 1995 . Regulation of intracellular choles- terol metabolism. Cell Struct. Funct. 20 : 421 – 427 .

19 . Bates , H. E. , J. E. Campbell , J. R. Ussher , L. L. Baggio , A. Maida , Y.

Seino , and D. J. Drucker . 2012 . Gipr is essential for adrenocortical steroidogenesis; however, corticosterone defi ciency does not medi- ate the favorable metabolic phenotype of Gipr(-/-) mice. Diabetes . 61 : 40 – 48 .

20 . Petersen , H. H. , T. K. Andreassen , T. Breiderhoff , J. H. Bräsen , H. Schulz , V. Gross , H. J. Gröne , A. Nykjaer , and T. E. Willnow . 2006 . Hyporesponsiveness to glucocorticoids in mice genetically defi cient for the corticosteroid binding globulin. Mol. Cell. Biol. 26 : 7236 – 7245 .

21 . Suzuki , S. , and K. Nakano . 1986 . LPS-caused secretion of corticos- terone is mediated by histamine through histidine decarboxylase.

Am. J. Physiol. 250 : E243 – E247 .

22 . Hoekstra , M. , V. Frodermann , T. van den Aardweg , R. J. van der Sluis , and J. Kuiper . 2013 . Leukocytosis and enhanced susceptibil- ity to endotoxemia but not atherosclerosis in adrenalectomized APOE knockout mice. PLoS One . 8 : e80441 .

23 . Plump , A. S. , N. Azrolan , H. Odaka , L. Wu , X. Jiang , A. Tall , S.

Eisenberg , and J. L. Breslow . 1997 . ApoA-I knockout mice: charac- terization of HDL metabolism in homozygotes and identifi cation of a post-RNA mechanism of apoA-I up-regulation in heterozy- gotes. J. Lipid Res. 38 : 1033 – 1047 .

24 . Wada , M. , T. Iso , B. F. Asztalos , N. Takama , T. Nakajima , Y. Seta , K. Kaneko , Y. Taniguchi , H. Kobayashi , K. Nakajima , et al . 2009 . Marked high density lipoprotein defi ciency due to apolipo- protein A-I Tomioka (codon 138 deletion). Atherosclerosis . 207 : 157 – 161 .

25 . Tilly-Kiesi , M. , Q. Zhang , S. Ehnholm , J. Kahri , S. Lahdenperä , C.

Ehnholm , and M. R. Taskinen . 1995 . ApoA-IHelsinki (Lys107 → 0) associated with reduced HDL cholesterol and LpA-I:A-II defi ciency.

Arterioscler. Thromb. Vasc. Biol. 15 : 1294 – 1306 .

26 . Matsunaga , T. , Y. Hiasa , H. Yanagi , T. Maeda , N. Hattori , K.

Yamakawa , Y. Yamanouchi , I. Tanaka , T. Obara , and H. Hamaguchi . 1991 . Apolipoprotein A-I defi ciency due to a codon 84 nonsense mutation of the apolipoprotein A-I gene. Proc. Natl. Acad. Sci. USA . 88 : 2793 – 2797 .

27 . Kraemer , F. B. , W. J. Shen , S. Patel , J. Osuga , S. Ishibashi , and S.

Azhar . 2007 . The LDL receptor is not necessary for acute adre- nal steroidogenesis in mouse adrenocortical cells. Am. J. Physiol.

Endocrinol. Metab. 292 : E408 – E412 .

28 . Hoekstra , M. , I. Meurs , M. Koenders , R. Out , R. B. Hildebrand , J.

K. Kruijt , M. Van Eck , and T. J. Van Berkel . 2008 . Absence of HDL cholesteryl ester uptake in mice via SR-BI impairs an adequate ad- renal glucocorticoid-mediated stress response to fasting. J. Lipid Res. 49 : 738 – 745 .

29 . Swarnakar , S. , R. E. Temel , M. A. Connelly , S. Azhar , and D. L.

Williams . 1999 . Scavenger receptor class B, type I, mediates selec- tive uptake of low density lipoprotein cholesteryl ester. J. Biol. Chem.

274 : 29733 – 29739 .

30 . Stangl , H. , M. Hyatt , and H. H. Hobbs . 1999 . Transport of lipids from high and low density lipoproteins via scavenger receptor-BI. J.

Biol. Chem. 274 : 32692 – 32698 .

31 . Webb , N. R. , M. C. de Beer , J. Yu , M. S. Kindy , A. Daugherty , D. R.

van der Westhuyzen , and F. C. de Beer . 2002 . Overexpression of SR-BI by adenoviral vector promotes clearance of apoA-I, but not apoB, in human apoB transgenic mice. J. Lipid Res. 43 : 1421 – 1428 . 32 . Van Eck , M. , M. Hoekstra , R. Out , I. S. Bos , J. K. Kruijt , R. B.

Hildebrand , and T. J. Van Berkel . 2008 . Scavenger receptor BI fa- cilitates the metabolism of VLDL lipoproteins in vivo. J. Lipid Res.

49 : 136 – 146 .

33 . Brodeur , M. R. , V. Luangrath , G. Bourret , L. Falstrault , and L.

Brissette . 2005 . Physiological importance of SR-BI in the in vivo In conclusion, we have shown that HDL is not critical

for proper adrenal glucocorticoid function in mice with a human-like lipoprotein profi le. Our fi ndings contribute to a better understanding of the adrenal glucocorticoid func- tion under human-like lipoprotein conditions and provide the fi rst experimental evidence that APOB-containing li- poprotein fractions may facilitate adrenal steroidogenesis, in an LDLR-independent manner, in vivo.

REFERENCES

1 . Hoekstra , M. , S. J. Korporaal , Z. Li , Y. Zhao , M. Van Eck , and T. J.

Van Berkel . 2010 . Plasma lipoproteins are required for both basal and stress-induced adrenal glucocorticoid synthesis and protection against endotoxemia in mice. Am. J. Physiol. Endocrinol. Metab. 299 : E1038 – E1043 .

2 . Hoekstra , M. , S. J. Korporaal , R. J. van der Sluis , V. Hirsch- Reinshagen , A. E. Bochem , C. L. Wellington , T. J. Van Berkel , J.

A. Kuivenhoven , and M. Van Eck . 2013 . LCAT defi ciency in mice is associated with a diminished adrenal glucocorticoid function. J.

Lipid Res. 54 : 358 – 364 .

3 . Plump , A. S. , S. K. Erickson , W. Weng , J. S. Partin , J. L. Breslow , and D. L. Williams . 1996 . Apolipoprotein A-I is required for cholesteryl ester accumulation in steroidogenic cells and for normal adrenal steroid production. J. Clin. Invest. 97 : 2660 – 2671 .

4 . Hoekstra , M. , D. Ye , R. B. Hildebrand , Y. Zhao , B. Lammers , M.

Stitzinger , J. Kuiper , T. J. Van Berkel , and M. Van Eck . 2009 . Scavenger receptor class B type I-mediated uptake of serum choles- terol is essential for optimal adrenal glucocorticoid production. J.

Lipid Res. 50 : 1039 – 1046 .

5 . Hoekstra , M. , R. J. van der Sluis , M. Van Eck , and T. J. Van Berkel . 2013 . Adrenal-specifi c scavenger receptor BI defi ciency induces glucocorticoid insuffi ciency and lowers plasma very-low-density and low-density lipoprotein levels in mice. Arterioscler. Thromb. Vasc. Biol.

33 : e39 – e46 .

6 . Bochem , A. E. , A. G. Holleboom , J. A. Romijn , M. Hoekstra , G. M.

Dallinga-Thie , M. M. Motazacker , G. K. Hovingh , J. A. Kuivenhoven , and E. S. Stroes . 2013 . High density lipoprotein as a source of cho- lesterol for adrenal steroidogenesis: a study in individuals with low plasma HDL-C. J. Lipid Res. 54 : 1698 – 1704 .

7 . Bochem , A. E. , A. G. Holleboom , J. A. Romijn , M. Hoekstra , G.

M. Dallinga , M. M. Motazacker , G. K. Hovingh , J. A. Kuivenhoven , and E. S. Stroes . 2014 . Adrenal function in females with low plasma HDL-C due to mutations in ABCA1 and LCAT. PLoS One . 9 : e90967 . 8 . Xing , Y. , W. E. Rainey , J. W. Apolzan , O. L. Francone , R. B. Harris , and W. B. Bollag . 2012 . Adrenal cell aldosterone production is stimulated by very-low-density lipoprotein (VLDL). Endocrinology . 153 : 721 – 731 .

9 . Higashijima , M. , H. Nawata , K. Kato , and H. Ibayashi . 1987 . Studies on lipoprotein and adrenal steroidogenesis: I. Roles of low den- sity lipoprotein- and high density lipoprotein-cholesterol in steroid production in cultured human adrenocortical cells. Endocrinol. Jpn.

34 : 635 – 645 .

10 . Higashijima , M. , K. Kato , H. Nawata , and H. Ibayashi . 1987 . Studies on lipoprotein and adrenal steroidogenesis: II. Utilization of low density lipoprotein- and high density lipoprotein-cholesterol for steroid production in functioning human adrenocortical adenoma cells in culture. Endocrinol. Jpn. 34 : 647 – 657 .

11 . Rainey , W. E. , R. J. Rodgers , and J. I. Mason . 1992 . The role of bovine lipoproteins in the regulation of steroidogenesis and HMG-CoA reductase in bovine adrenocortical cells. Steroids . 57 : 167 – 173 .

12 . Vaessen , S. F. , R. J. Veldman , E. M. Comijn , J. Snapper , J. A. Sierts , K. van den Oever , S. G. Beattie , J. Twisk , and J. A. Kuivenhoven . 2009 . AAV gene therapy as a means to increase apolipoprotein (Apo) A-I and high-density lipoprotein-cholesterol levels: correc- tion of murine ApoA-I defi ciency. J. Gene Med. 11 : 697 – 707 . 13 . Ishibashi , S. , M. S. Brown , J. L. Goldstein , R. D. Gerard , R. E.

Hammer , and J. Herz . 1993 . Hypercholesterolemia in low density lipoprotein receptor knockout mice and its reversal by adenovirus- mediated gene delivery. J. Clin. Invest. 92 : 883 – 893 .

14 . Cai , L. , A. Ji , F. C. de Beer , L. R. Tannock , and D. R. van der Westhuyzen . 2008 . SR-BI protects against endotoxemia in mice

at Walaeus Library / BIN 299, on February 1, 2017www.jlr.orgDownloaded from

(7)

metabolism of human HDL and LDL in male and female mice. J.

Lipid Res. 46 : 687 – 696 .

34 . Out , R. , J. K. Kruijt , P. C. Rensen , R. B. Hildebrand , P. de Vos , M.

Van Eck , and T. J. Van Berkel . 2004 . Scavenger receptor BI plays a role in facilitating chylomicron metabolism. J. Biol. Chem. 279 : 18401 – 18406 .

35 . Van Eck , M. , J. Twisk , M. Hoekstra , B. T. Van Rij , C. A. Van der Lans , I. S. Bos , J. K. Kruijt , F. Kuipers , and T. J. Van Berkel . 2003 . Differential effects of scavenger receptor BI defi ciency on lipid me- tabolism in cells of the arterial wall and in the liver. J. Biol. Chem.

278 : 23699 – 23705 .

36 . Wang , N. , T. Arai , Y. Ji , F. Rinninger , and A. R. Tall . 1998 . Liver- specifi c overexpression of scavenger receptor BI decreases levels of very low density lipoprotein ApoB, low density lipoprotein ApoB, and high density lipoprotein in transgenic mice. J. Biol. Chem. 273 : 32920 – 32926 .

37 . Out , R. , M. Hoekstra , S. C. de Jager , P. de Vos , D. R. van der Westhuyzen , N. R. Webb , M. Van Eck , E. A. Biessen , and T. J. Van Berkel . 2005 . Adenovirus-mediated hepatic overexpression of scav- enger receptor class B type I accelerates chylomicron metabolism in C57BL/6J mice. J. Lipid Res. 46 : 1172 – 1181 .

38 . Vergeer , M. , S. J. Korporaal , R. Franssen , I. Meurs , R. Out , G. K.

Hovingh , M. Hoekstra , J. A. Sierts , G. M. Dallinga-Thie , M. M.

Motazacker , et al . 2011 . Genetic variant of the scavenger receptor BI in humans. N. Engl. J. Med. 364 : 136 – 145 .

39 . Zhang , X. L. , Q. Q. Zhu , L. Zhu , J. Z. Chen , Q. H. Chen , G. N. Li , J.

Xie , L. N. Kang , and B. Xu . 2015 . Safety and effi cacy of anti-PCSK9 antibodies: a meta-analysis of 25 randomized, controlled trials.

BMC Med. 13 : 123 .

40 . Luo , L. , X. Yuan , W. Huang , F. Ren , H. Zhu , Y. Zheng , and L.

Tang . 2015 . Safety of coadministration of ezetimibe and statins in patients with hypercholesterolaemia: a meta-analysis. Intern. Med. J.

45 : 546 – 557 .

41 . Illingworth , D. R. , T. A. Kenny , W. E. Connor , and E. S. Orwoll . 1982 . Corticosteroid production in abetalipoproteinemia: evidence for an impaired response ACTH. J. Lab. Clin. Med. 100 : 115 – 126 . 42 . Illingworth , D. R. , T. A. Kenny , and E. S. Orwoll . 1982 . Adrenal

function in heterozygous and homozygous hypobetalipoprotein- emia. J. Clin. Endocrinol. Metab. 54 : 27 – 33 .

43 . Illingworth , D. R. , E. S. Orwoll , and W. E. Connor . 1980 . Impaired cortisol secretion in abetalipoproteinemia. J. Clin. Endocrinol.

Metab. 50 : 977 – 979 .

at Walaeus Library / BIN 299, on February 1, 2017www.jlr.orgDownloaded from

Referenties

GERELATEERDE DOCUMENTEN

The selective uptake of high density lipoprotein cholesterol esters by the liver : the role of scavenger receptor BI in reverse cholesterol transport1. Retrieved

In order to test whether the changes in SR-BI expression induced by either or a high cholesterol diet also affected the increased selective uptake of HDL CEOH,

By understanding Chile as a state of exception I have been able to expose the in Agamben's terms ‘homo sacer’ and ‘bare life’, and also the relation of ban and zone of

Compared to family controls and relative to the concentrations of total 27OHC and cholesterol, lower 27OHC-ester but normal cholesterylester levels were found in HDL of

We recommend using the view based approach if a it is possible to exactly match the pose, illumination condition and camera of the suspect reference set to that of the probe image

plasma triglycerides, non-high density lipoprotein (non-HDL) cholesterol, apolipoprotein B (apoB) and apolipoprotein E (apoE) with thyroid-stimulating hormone (TSH) levels in

Van  Wiechenonderzoek  bij negatieve (-) score:  verwijzen​ B​ naar een audiologisch centrum voor multidisciplinaire diagnostiek  A​ : Door jeugdverpleegkundige,

Dat betekent bijvoorbeeld dat wanneer iemand nog 1 jaar te leven heeft zonder behandeling en 2 jaar als hij de aandoening niet had gehad, dat zijn proportional shortfall nog