You are on page 1of 11

BBA - Molecular and Cell Biology of Lipids 1866 (2021) 158909

Contents lists available at ScienceDirect

BBA - Molecular and Cell Biology of Lipids


journal homepage: www.elsevier.com/locate/bbalip

Lipoprotein receptor SR-B1 deficiency enhances adipose tissue


inflammation and reduces susceptibility to hepatic steatosis during
diet-induced obesity in mice
Katherine Rivera a, b, Verónica Quiñones a, Ludwig Amigo a, Nicolás Santander c,
Francisca Salas-Pérez d, Aline Xavier b, e, Marta Fernández-Galilea f, g, Gonzalo Carrasco h,
Daniel Cabrera i, j, Marco Arrese i, k, Dolores Busso l, Marcelo E. Andia b, e, Attilio Rigotti a, b, m, *
a
Department of Nutrition, Diabetes, and Metabolism, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile
b
Millennium Nucleus for Cardiovascular Magnetic Resonance, Santiago 7820436, Chile
c
Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94143, USA
d
Instituto de Ciencias de la Salud, Universidad de O’Higgins, Rancagua 2820000, Chile
e
Biomedical Imaging Center, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
f
Centre for Nutrition Research, School of Pharmacy and Nutrition, University of Navarra, Pamplona 31008, Spain
g
IDISNA, Navarra’s Health Research Institute, Pamplona 31008, Spain
h
Department of Pathology, Hospital Clínico Universidad de Chile, Santiago 8320000, Chile
i
Department of Gastroenterology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile
j
Faculty of Medical Sciences, School of Medicine, Universidad Bernardo O Higgins, Santiago 8370854, Chile
k
Centro de Envejecimiento y Regeneración (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
l
Biomedical Research and Innovation Center, Faculty of Medicine, Universidad de los Andes, Santiago 111711, Chile
m
Center of Molecular Nutrition and Chronic Diseases, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile

A R T I C L E I N F O A B S T R A C T

Keywords: Scavenger receptor class B type 1 (SR-B1) is a membrane lipoprotein receptor/lipid transporter involved in the
SR-B1 pathogenesis of atherosclerosis, but its role in obesity and fatty liver development is unclear. Here, we deter­
Lipid metabolism mined the effects of SR-B1 deficiency on plasma metabolic and inflammatory parameters as well as fat deposition
Obesity
in adipose tissue and liver during obesity. To induce obesity, we performed high-fat diet (HFD) exposure for 12
Fatty liver
Adipose tissue
weeks in male SR-B1 knock-out (SR-B1− /− , n = 14) and wild-type (WT, n = 12) mice. Compared to HFD-fed WT
mice, plasma from HFD-fed SR-B1− /− animals exhibited increased total cholesterol, triglycerides (TG) and tumor
necrosis factor-α (TNF-α) levels. In addition, hypertrophied adipocytes and macrophage-containing crown-like
structures (CLS) were observed in adipose tissue from HFD-fed SR-B1 deficient mice. Remarkably, liver from

Abbreviations: ALT, alanine transaminase; ApoA-I, apolipoprotein A-I; Apob, apolipoprotein B; AUC, area under the curve; BMI, body mass index; CLS, crown-like
structures; Cpt1, carnitine palmitoyltransferase 1; CVD, cardiovascular disease; Dgat2, diacylglycerol o-acyltransferase 2; ELISA, enzyme-linked immunosorbent
assay; eWAT, epididimal WAT; FA, fatty acids; Fasn, fatty acid synthase; FFA, free FA; FPLC, fast protein liquid chromatography; Gapdh, glyceraldehyde-3-phosphate
dehydrogenase; GC–MS, gas chromatography with mass spectrometer; GGT, gamma-glutamyltransferase; H&E, hematoxylin-eosin; HDL, high-density lipoprotein;
HDL-C, HDL cholesterol; HFD, high fat diet; HOMA-IR, homeostatic model of assessment for IR; IL-10, interleukin 10; IR, insulin resistance; LDL, low-density li­
poprotein; Ldlr, LDL receptor; LPL, lipoprotein lipase; Lrp1, low-density lipoprotein receptor-related protein 1; Mgat1, monoacylglycerol acyltransferase 1; Mttp,
microsomal triglyceride transfer protein; MAFLD, metabolic (dysfunction) associated fatty liver disease; MUFA, monounsaturated FA; NAFLD, non-alcoholic fatty
liver disease; NASH, non-alcoholic steatohepatitis; PPARα, Peroxisome proliferator-activated receptor α; PPARγ, peroxisome proliferator-activated receptor γ; PUFA,
polyunsaturated FA; RCT, reverse cholesterol transport; RT-PCR, reverse transcription & polymerase chain reaction; S1P, sphingosine 1 phosphate; Scd1, stearoyl-
CoA desaturase 1; SFA, saturated FA; SR-B1, scavenger receptor class B type 1; Srebp1c, sterol regulatory element binding protein 1c; TG, triglycerides; TNF-α, tumor
necrosis factor-α; VLDL, very-low-density lipoproteins; WAT, white adipose tissue; WT, wild-type.
* Corresponding author at: Departamento de Nutrición, Diabetes y Metabolismo, Escuela de Medicina, Pontificia Universidad Católica de Chile, Marcoleta 367,
interior, 2do Piso, Santiago CP 83300024, Chile.
E-mail addresses: ksrivera@uc.cl (K. Rivera), veroqui.quinones@gmail.com (V. Quiñones), ludwig@uc.cl (L. Amigo), nicolas.santandergrez@ucsf.edu
(N. Santander), francisca.salas@uoh.cl (F. Salas-Pérez), acarvalhodasilva@uc.cl (A. Xavier), mfgalilea@unav.es (M. Fernández-Galilea), gcarrasa@me.com
(G. Carrasco), dacabrer@uc.cl (D. Cabrera), marrese@uc.cl (M. Arrese), dbusso@uandes.cl (D. Busso), meandia@uc.cl (M.E. Andia), arigotti@med.puc.cl
(A. Rigotti).

https://doi.org/10.1016/j.bbalip.2021.158909
Received 3 December 2020; Received in revised form 5 February 2021; Accepted 17 February 2021
Available online 22 February 2021
1388-1981/© 2021 Elsevier B.V. All rights reserved.
K. Rivera et al. BBA - Molecular and Cell Biology of Lipids 1866 (2021) 158909

obese SR-B1− /− mice showed attenuated TG content, dysregulation in hepatic peroxisome proliferator-activated
receptors (PPARs) expression, increased hepatic TG secretion, and altered hepatic fatty acid (FA) composition. In
conclusion, we show that SR-B1 deficiency alters the metabolic environment of obese mice through modulation
of liver and adipose tissue lipid accumulation. Our findings provide the basis for further elucidation of SR-B1’s
role in obesity and fatty liver, two major public health issues that increase the risk of advanced chronic diseases
and overall mortality.

1. Introduction 2. Methods

Obesity is a multifactorial syndrome and major risk factor for a va­ 2.1. Animals and diet
riety of cardiometabolic diseases [1]. This condition, characterized by
abnormal or excess fat accumulation, promotes a chronic inflammatory Mice with a targeted mutation in the SR-B1 gene locus on a C57BL/6
state and atherogenic dyslipidemia, which are strongly associated with J:129 mixed background (B6.129S2-Scarb1tm1Kri) were originally obtained
insulin resistance (IR) and diabetes, increased cardiovascular disease from Dr. Monty Krieger (Massachusetts Institute of Technology, Cambridge,
(CVD) incidence and mortality risk in the general population [2]. MA, USA) and maintained in the animal facility of the School of Medicine,
During obesity, alterations in white adipose tissue (WAT) can pro­ Pontificia Universidad Católica de Chile at 25 ◦ C and 12h light:dark cycling.
mote lipid accumulation in the liver, leading to intracellular lipid Because homozygous females are infertile in this mixed genetic background
droplet formation, a condition known as hepatic steatosis [3]. In several and their cyclic changes in sexual hormone levels may interfere with bio­
pathological conditions, these lipid droplets may expand and potentially markers, 8-week-old homozygous SR-B1 knock-out (SR-B1− /− , n = 14) and
interfere with essential hepatocellular functions [4]. This alteration is a wild-type (WT, n = 12) male mice obtained from heterozygous (SR-B1+/− )
hallmark feature of non-alcoholic fatty liver disease (NAFLD), the most intercrosses were fed a HFD (60% kcal from fat; 58Y1 TestDiet, St. Louis, MO,
prevalent liver disease worldwide that results in a major impact on USA) in three independent sets. This diet induces obesity and hepatic stea­
health and economic burden [5]. tosis when administered for 12 weeks. A normal chow diet control group was
Although obesity is intimately associated with liver fat accumulation not included because the aim of this study was to evaluate specifically the
and its comorbidities, some observations have documented that not all impact of SR-B1 deficiency within an obesogenic context. Body weight and
patients with obesity develop metabolic complications, supporting the food intake were weekly assessed during the duration of the study and no
idea that genetic variants may predispose a subset of obese subjects to important adverse events were observed. All procedures were performed
increased risk of chronic complications and death [5–7]. Indeed, special following the Guide for the Care and Use of Laboratory Animals (copyright
attention in lipid transport-related proteins on development of meta­ 1996, National Academy of Science) published by National Research Council
bolic abnormalities induced by obesity could help to understand this (NRC) and approved by the Ethics and Animal Welfare Committee from the
wide variety of obesity-related phenotypes [8]. Interestingly, circulating School of Medicine of Pontificia Universidad Católica de Chile (Protocol
high-density lipoprotein cholesterol (HDL-C) and triglyceride (TG) #14-036).
levels are altered in obese subjects with fatty liver disease [9,10]. In
addition, recent observations have shown that impaired HDL-C efflux 2.2. Tissue sampling
capacity is associated with subclinical atherosclerosis in NAFLD pa­
tients, further contributing to increased CVD risk [11]. After 12 weeks of dietary intervention, WT and SR-B1− /− obese animals
Scavenger receptor class B type 1 (SR-B1) is a key membrane re­ were euthanized with a mixture of ketamine:xylazine (150:10 mg/kg,
ceptor that modulates the bidirectional flux of lipids between lipopro­ intraperitoneally) after overnight fasting. Blood was collected by cardiac
teins and cells in different tissues [12]. As a feature of lipoprotein puncture. Plasma was obtained by low-speed centrifugation of blood and
metabolism, hepatic SR-B1 mediates HDL lipid delivery through reverse kept at − 80 ◦ C until further analysis. Epididymal WAT (eWAT) and liver
cholesterol transport (RCT), playing a critical role against atheroscle­ samples were removed, weighed, and flash frozen in liquid nitrogen or
rosis [13,14]. processed for histological analyses by light microscopy and
Studies using 3T3-L1 adipocytes have shown that lipogenic stimu­ immunohistochemistry.
lation promoted mobilization of SR-B1 to the plasma membrane
[15,16]. As well, SR-B1 protein expression was down-regulated by in­
flammatory tumor necrosis factor-α (TNF-α) exposure in partially 2.3. Biochemical analyses
differentiated adipocytes [17]. In addition, the expression of SR-B1 in
hepatocytes was modulated by peroxisome proliferator-activated re­ Plasma or frozen tissues were used for biochemical analyses. Plasma
ceptor γ (PPARγ), a crucial regulator of lipogenic pathways [18]. total cholesterol levels were quantified through an enzymatic method
Remarkably, a recent study showed that SR-B1 modulation by over­ described previously [20]. Plasma TG and free FA (FFA) concentrations
expression or siRNA-mediated knock− down regulates fatty acid (FA) were measured by enzymatic assays using commercial kits (Sigma-
uptake in cells [19]. Taken together, these findings strongly suggest that Aldrich, St. Louis, MO, USA). Plasma concentrations of TNF-α and
SR-B1 may play a role in metabolic environment maintenance and lipid interleukin 10 (IL-10) were determined using enzyme-linked immuno­
deposition during obesity. However, the specific function of SR-B1 sorbent assay (ELISA) kits (R&D Systems, Minneapolis, MN, USA).
during obesity and progression of its comorbidities is still not fully Plasma levels of leptin and adiponectin were also determined by ELISA
understood. kits (Sigma-Aldrich, St. Louis, MO, USA). Total lipids from liver samples
In the current study, we used a SR-B1 deficient mouse model fed with were extracted using a chloroform/methanol solution (2:1, vol/vol), as
high-fat diet (HFD) that recapitulates obesity and initiation of NAFLD to previously described [21], and total TG and cholesterol content were
evaluate if SR-B1 expression modulates the metabolic and inflammatory analyzed by enzymatic tests indicated above. Lipoprotein cholesterol
phenotype of obese mice through the regulation of lipid deposition levels were quantified in fast protein liquid chromatography (FPLC)-
within adipocytes and hepatocytes. Our findings suggest that variations fractionated plasma samples as described previously [20]. Blood glucose
in human obesity phenotypes and comorbidities may be modulated and levels were measured by Accu-Chek Active glucometer (Roche Di­
explained through changes in expression and/or function of SR-B1. agnostics, Mannheim, Germany). Plasma insulin concentrations were
measured with a commercial kit (Sigma-Aldrich, St. Louis, MO, USA).
Plasma glucose-to-insulin ratio and homeostatic model of assessment for

2
K. Rivera et al. BBA - Molecular and Cell Biology of Lipids 1866 (2021) 158909

insulin resistance (IR) (HOMA-IR; fasting plasma insulin (μU/mL) × Table 1


fasting glucose (mmol/L)/22.5) were also calculated. Plasma levels of RT-PCR primers sequences for mRNA analysis.
liver enzymes [alanine transaminase (ALT) and gamma- Gene Forward primer Reverse primer
glutamyltransferase (GGT)] were measured with a commercial kit
Gapdh TGCGACTTCAACAGCAACTCCCAC TTGCTCAGTGTCCTTGCTGGGG
(HUMAN Diagnostics Worldwide, Wiesbaden, Denmark). Lrp1 TGGGTCTCCCGAAATCTGTT ACCACCGCATTCTTGAAGGA
Ldlr AGGCTGTGGGCTCCATAGG TGCGGTCCAGGGTCATCT
2.4. Histological analyses Srebp1c GGAGCCATGGATTGCACATT GGCCCGGGAAGTCACTGT
Scd1 CCGGAGACCCCTTAGATCGA TAGCCTGTAAAAGATTTCTGCAAACC
Fasn GCTGCGGAAACTTCAGGAAAT AGAGACGTGTCACTCCTGGACTT
For standard histology, eWAT and liver samples were fixed overnight Mgat1 CTGGTTCTGTTTCCCGTTGT TGGGTCAAGGCCATCTTAAC
in 10% formalin at 4 ◦ C, then dehydrated, cleared, and paraffin- Dgat2 CCGCAAAGGCTTTGTGAAG GGAATAAGTGGGAACCAGATCA
embedded. Ten-micrometer sections from formalin-fixed paraffin- Pparα ACAAGGCCTCAGGGTACCA GCCGAAAGAAGCCCTTACAG
embedded eWAT and liver samples were mounted and stained with Cpt1 CACCAACGGGCTCATCTTCTA CAAAATGACCTAGCCTTCTATCGAA
Pparγ GCAGAGACAAATGTGCTTCG TCTGGGGTCAGAGGAAGAGA
hematoxylin and eosin (H&E) solution. All stained slides were examined
Apob CGTGGGCTCCAGCATTCTA TCACCAGTCATTTCTGCCTTTG
with a Nikon Eclipse E200 light microscope and photographed with a Mttp CCTACCAGGCCCAACAAGAC CGCTCAATTTTGCATGTATCC
Nikon DS-Fi1 camera. Representative images (4–5 per animal) from
Gapdh, Glyceraldehyde-3-phosphate dehydrogenase; Lrp1, Low-density lipo­
each tissue were taken and adipocyte and hepatocyte lipid droplet areas
protein receptor related protein; Ldlr, Low-density lipoprotein receptor; Srebp1c,
were measured blindly for at least 100 individual cells per mouse using
Sterol regulatory element binding protein 1c; Scd1, Stearoyl-coA desaturase
Image J software (National Institutes of Health, Bethesda, MD, USA). 1rp1; Fasn, Fatty acid synthase; Mgat1, Monoacylglycerol acyltransferase 1;
For immunohistochemistry, dewaxed 3 μm serial sections of paraffin- Dgat2, Diacylglycerol o-acyltransferase 2; Pparα, Peroxisome proliferator-
embedded eWAT were treated with 3% hydrogen peroxide to inactivate activated receptor α; Cpt1, Carnitine palmitoyltransferase 1; Pparγ, Peroxisome
endogenous peroxidases and then macrophage infiltration of the adipose proliferator-activated receptor γ; Apob, Apolipoprotein b; Mttp, Microsomal
tissue was characterized using anti-MAC-2 (1:100; Cedarlane Labora­ triglyceride transfer protein.
tories, Paletta Court, Burlington, Ontario, Canada), as previously
described [22]. Biotinylated HRP-conjugated secondary antibodies were 2.7. Hepatic triglyceride secretion
goat anti-mouse IgG (H + L) (Molecular Probes, Life Technologies,
Thermo Fisher Scientific, Waltham, MA, USA). The histochemical re­ To measure hepatic TG production, mice fed with HFD for 12 weeks
action was performed using 3,3′ -diaminobenzidine as a substrate were fasted for 4 h. Animals were injected with Tyloxapol (Sigma-
(Sigma-Aldrich, St. Louis, MO). Sections were counterstained with he­ Aldrich, St. Louis, MO), a lipoprotein lipase inhibitor, at 500 mg/kg
matoxylin. Tissue sections were observed and macrophage-containing intravenously in saline, as previously shown to be optimal in mice [24].
crown-like structure (CLS) density was obtained by counting the total Immediately before injection and at 1 h post-injection, blood samples
number of CLS in each section compared with the total number of adi­ were drawn in heparinized capillary tubes and plasma was prepared and
pocytes and expressed as CLS number/1000 adipocytes. fractionated by FPLC using a Superose-6 10/300 GL column (GE
Healthcare Life Sciences, Marlborough, MA, USA), as previously
2.5. Quantitative real-time PCR assay described [20]. Lipoprotein fractions eluted according to their particle
size were recovered and TG concentrations were determined.
Total RNA from livers was extracted using Trizol Reagent (Invitrogen
Corporation, Carlsbad, CA, USA). Aliquots of 1–2 μg of RNA from each 2.8. Lipid FA profile analyses
tissue sample were reverse-transcribed to cDNA by a high-capacity
cDNA reverse transcription kit (Applied Biosystems, Foster City, CA, Lipids were extracted from livers using a chloroform/methanol so­
USA). The reverse transcription polymerase chain reaction (RT-PCR) lution (2:1, vol/vol), as previously described [21], and subjected to
was performed by QuantiTest SYBR Green PCR kit (Applied Biosystems, esterification for analytical stability. FA were analyzed using gas chro­
Foster City, CA, USA) using the ABI 7300 sequence detection system matography–mass spectrometry (GC–MS) (PerkinElmer, Clarus 680,
(Applied Biosystems, Foster City, CA, USA). Specific mRNA levels were Waltham, MA, USA) equipped with HP-Innowax capillary column
quantified and normalized to those of glyceraldehyde-3-phosphate de­ (length 25 m, 0.2 mm internal diameter, 0.2 mm film). Data register was
hydrogenase (Gapdh) using the mathematical model previously in SCAN mode and peak integration was obtained by the TurboMass
described [23]. For primer list and sequences, see Table 1. Training 2016 PRO software. FA peaks were identified based on
comparing retention times with external standards, and mass was
2.6. Western blot calculated according to the integration area of corresponding peaks. FA
composition was defined as the percentage of individual fatty acids
Liver samples were disrupted in homogenization buffer with prote­ relative to total content, as previously detailed [25].
ase inhibitors and homogenates were used for protein quantification by
Bio-Rad Protein Assay (Bio-Rad Laboratories, Hercules, CA, USA). 2.9. Statistical analyses
Western blot analysis of PPARγ protein in the liver was performed using
50 μg of protein/sample. Proteins were size-fractionated in 12% SDS- Statistical analyses were performed using GraphPad Prism 8.0.1
PAGE gels and transferred onto PVDF membranes. For protein detec­ (GraphPad Software, San Diego, CA, USA). The normality of data was
tion, specific primary and secondary antibodies were used as follows. determined with the Kolmogorov–Smirnov test. Unpaired Student’s t-
The membrane was incubated with rabbit polyclonal antibody to PPARγ test was used to assess whether the means of normally distributed data
(Cell Signalling Technology, Inc., London, United Kingdom) overnight at differed significantly, and Mann Whitney U test as its non-parametric
4 ◦ C. PPARγ protein expression was normalized to ԑ-COP levels using a counterpart. Two-way ANOVA analysis with repeated measures was
rabbit polyclonal antibody obtained from Dr. Monty Krieger (Massa­ also used in some data evaluation with Bonferroni’s multiple compari­
chusetts Institute of Technology, Cambridge, MA, USA). After incuba­ sons post-hoc test. Tests were two-tailed and differences were consid­
tion with the primary antibody, the membrane was washed and ered significant when p < 0.05, where n indicates the sample size.
incubated with peroxidase-conjugated secondary antibodies (Invi­ RStudio package was used for principal component analysis.
trogen, Carlsbad, CA, USA), and the bound antibody was visualized with
the ECL kit (Amersham, Buckinghamshire, United Kingdom) and X-
Omat S film (Eastman Kodak Co., Rochester, NY, USA).

3
K. Rivera et al. BBA - Molecular and Cell Biology of Lipids 1866 (2021) 158909

3. Results B1− /− mice, we performed immunostaining analysis of macrophages


using the anti-MAC-2 (Galectin-3) antibody [22]. In association with
3.1. SR-B1 expression influences plasma lipid and cytokine levels in HFD- adipocyte hypertrophy, HFD-fed SR-B1− /− mice presented changes in
fed mice macrophage-containing CLS (Fig. 4D), exhibiting a significant increase
in the density of CLS within adipose tissue in comparison with HFD-fed
We used a high-calorie fat-based diet to evaluate a putative role of WT mice (+94%, p < 0.01) (Fig. 4E).
SR-B1 deficiency on the metabolic phenotype and the potential mech­ Adipocytes are the source of several hormones, including leptin and
anisms by which this lipoprotein receptor may be involved during diet- adiponectin that regulate whole-body metabolism [29]. However, the
induced obesity in mice. As expected, animals fed a HFD for 12 weeks expression of SR-B1 did not significantly modify plasma adiponectin
turned significantly obese as indicated by higher body weights, (Fig. 4F) or leptin (Fig. 4G) levels compared to WT mice after con­
compared to baseline (p < 0.0001) (Fig. 1A). However, body weight gain sumption of the HFD diet (p > 0.05).
and the average food intake were similar between SR-B1− /− and WT
groups (Fig. 1B–C). 3.3. Obese SR-B1-deficient exhibited marked attenuation in hepatic
Next, we investigated whether SR-B1 deficiency affected key plasma steatosis
parameters of obesity-associated metabolic dysfunction. Higher levels of
total plasma cholesterol (+107%, p < 0.0001) were found in HFD-fed During obesity, pathological adipocyte enlargement in association
SR-B1− /− mice than in obese WT mice (Fig. 2A). Consistently, the li­ with fat tissue immune cell infiltration leads to lipid accumulation in
poprotein profile showed that plasma cholesterol levels were associated non-adipose tissues [27]. To explore the potential role of SR-B1
with HDL-C accumulation in SR-B1− /− mice (Fig. 2B), most likely due to expression on the pathogenesis of NAFLD, we investigated its involve­
decreased selective HDL cholesterol uptake previously reported in ment in liver steatosis development in response to HFD intake in WT and
chow-fed SR-B1 knock-out animals [26]. SR-B1− /− mice. Livers from SR-B1 deficient animals showed similar size
Surprisingly, the absence of SR-B1 in obese mice also caused a sig­ and weight, but were less yellowish, suggesting attenuated fatty liver,
nificant increase in circulating TG levels (+50%, p < 0.01) (Fig. 2C), compared to WT mice (Fig. 5A). Indeed, intrahepatic lipid droplet for­
while plasma FFA levels were similar (p > 0.05) (Fig. 2D), in comparison mation were evaluated by H&E staining supporting that obese SR-B1-
with WT mice. Furthermore, we found that TNF-α levels were also deficient mice exhibited fewer lipid deposits (Fig. 5B) as well as
significantly higher in obese SR-B1− /− mice than in WT (+214%, p < decreased size in hepatocyte lipid droplets, compared with WT mice
0.0001) (Fig. 2E). A trend to lower, but not statistically significant, (− 43%, p < 0.0001) (Fig. 5C).
plasma IL-10 levels was observed in obese SR-B1-deficient mice vs. obese As expected, biochemical measurements of total TG and cholesterol
WT (p = 0.0749) (Fig. 2F). These data suggest that SR-B1 deficiency in hepatic content showed that liver steatosis observed in WT obese mice
HFD-fed mice associated with inflammatory dyslipidemia. was due to excess accumulation of TG rather than cholesterol
The SR-B1− /− deficiency did not significantly modify plasma glucose (Fig. 5D–E). Specifically, HFD-fed SR-B1− /− mice had a significantly
levels after HFD intake (p > 0.05) (Fig. 3A). Interestingly, obese SR-B1- lower TG content (− 47%, p < 0.0001) but similar levels of cholesterol (p
deficient mice showed increased plasma insulin levels (+69%, p < 0.05) > 0.05) compared to WT mice (Fig. 5D–E).
and impaired insulin sensitivity as indicated by higher HOMA-IR Despite the marked dependence of liver fat accumulation on SR-B1
compared to WT (+79%, p < 0.05) (Fig. 3B-C), suggesting that SR-B1 expression, NAFLD-associated injury ALT and GGT enzymes in plasma
may be involved directly or indirectly in obesity-induced IR. samples showed no changes comparing SR-B1− /− and WT mice after
HFD-intake for 12 weeks (p > 0.05) (Fig. 5F–G).
3.2. Absence of SR-B1 promotes inflammatory hypertrophy of white
adipocytes in obese mice 3.4. Effect of SR-B1 deficiency on hepatic protein and gene expression
during obesity
Adipose tissue is the main organ that stores the excess of energy
intake as TG. It is well known that HFD consumption increases adipocyte A delicate balance between anabolic and catabolic processes regu­
size and adipose tissue mass, contributing to adipose tissue inflamma­ lates the hepatic lipid metabolism [30]. Indeed, PPARs and their target
tion and its associated metabolic abnormalities [27,28]. genes modulate lipogenesis, lipid oxidation, and lipid accumulation in
To investigate whether adipose morphology was changed in animals steatotic hepatocytes [31,32]. To delineate the mechanisms by which
lacking SR-B1 under obesity, we examined the average adipocyte size in the absence of SR-B1 attenuated liver steatosis during obesity, we next
obese mice. SR-B1− /− obese animals had a shift towards bigger fat cells, examined the expression of key hepatic protein/genes involved in lipid
compared to WT mice (+75%, p < 0.0001) (Fig. 4A–C). Furthermore, in uptake, biosynthesis, and catabolism.
order to assess the inflammatory status of the adipose tissue in obese SR- Interestingly, in livers from obese mice, the absence of SR-B1

Fig. 1. Body weight, body weight gain, and food intake in SR-B1− /− mice fed on a HFD for 12 weeks. (A) Body weight (B) body weight gain and (C) food intake for
WT (n = 12) and SR-B1− /− (n = 14) mice. Data are presented as mean ± SD or shown in box plots.

4
K. Rivera et al. BBA - Molecular and Cell Biology of Lipids 1866 (2021) 158909

Fig. 2. Plasma metabolic/inflammatory markers in SR-B1− /− mice fed on a HFD for 12 weeks. Plasma (A) total cholesterol, (B) lipoprotein cholesterol, (C) TG, (D)
FFA, (E) TNF-α, and (F) IL-10 levels for WT (n = 12) and SR-B1− /− (n = 14) mice. Data are presented as mean ± SD or shown in box plots. **p < 0.01; ****p < 0.001
vs. WT mice.

Fig. 3. Glucose and insulin production analysis in SR-B1− /− mice fed on a HFD for 12 weeks. Plasma concentrations of (A) glucose, (B) insulin, and (C) HOMA-IR for
WT (n = 12) and SR-B1− /− (n = 14) mice. Data are shown in box plots. *p < 0.05 vs. WT mice.

-compared with WT animals- was associated with a decrease in PPARγ genes.


protein levels (− 57%, p < 0.001) (Fig. 6A). In addition, consistent with Remarkably, SR-B1-deficiency was also associated with an approxi­
western blot analysis, Pparg mRNA levels were down-regulated in HFD- mate 2-fold increase in hepatic levels of apolipoprotein b (Apob) mRNA
fed SR-B1-deficient mice in comparison to WT mice (p < 0.05) (Fig. 6B). after HFD intake compared to the WT group (p < 0.05) (Fig. 6B), while
We did not observe any significant differences in genes regulating the no differences were observed in the expression of microsomal triglyc­
hepatic uptake of lipoproteins, such as low-density lipoprotein receptor- eride transfer protein (Mttp) (p > 0.05) (Fig. 6B), both key determinants
related protein (Lrp1) and low-density lipoprotein receptor (Ldlr) (p > of hepatic very-low-density lipoproteins (VLDL) assembly and secretion.
0.05) (Fig. 6B). Also, mRNA levels of genes regulating FA and TG syn­
thesis, such as sterol regulatory element-binding protein 1c (Srebp1c), 3.5. SR-B1 deficiency increases VLDL-TG secretion in obese mice
stearoyl-CoA desaturase 1 (Scd1), fatty acid synthase (Fasn), mono­
acylglycerol acyltransferase 1 (Mgat1), and diacylglycerol o-acyl­ During post-absorptive fasting conditions, VLDL particles produced
transferase 2 (Dgat2), did not show changes associated with SR-B1 by the liver are the primary carriers of TG in plasma, which require apoB
expression (p > 0.05) (Fig. 6B). Interestingly, obese SR-B1-deficient synthesis in the liver [33]. In order to determine if SR-B1 expression
mice expressed significantly higher mRNA levels of hepatic genes modulated hepatic and plasma TG levels after HFD feeding through
associated with FA β-oxidation, including peroxisome proliferator- changes in VLDL-TG secretion, we measured plasma accumulation of
activated receptor a (Ppara) and carnitine palmitoyltransferase 1 VLDL-TG after treatment with Tyloxapol, which acutely inhibits pe­
(Cpt1), compared to obese WT mice (p < 0.05) (Fig. 6B). Taken together, ripheral lipolysis of TG-rich lipoproteins.
these data suggest that the impact of SR-B1 expression on fatty liver was As shown in Fig. 7A, HFD-fed SR-B1− /− mice pretreated with
due in part to modulated expression of PPARs and some of their target tyloxapol had significantly increased plasma TG levels and VLDL-TG

5
K. Rivera et al. BBA - Molecular and Cell Biology of Lipids 1866 (2021) 158909

secretion compared to WT obese mice. These results indicate that the

Fig. 4. Adipose tissue histological analysis and plasma adipokine levels in SR-B1− /− mice fed on a HFD for 12 weeks. (A) Representative H&E-stained sections of eWAT. (B) Mean adipocyte cross-sectional area for WT
(n = 11) and SR-B1− /− (n = 14) mice. (C) The frequency distribution of adipocyte sizes for WT (n = 12) and SR-B1− /− (n = 14) mice. (D) Representative MAC-2 immunostained cross sections of eWAT showing CLS. (E)
Quantitative assessment of CLS per 1000 adipocytes for WT (n = 12) and SR-B1− /− (n = 14) mice. Plasma (F) adiponectin and (G) leptin levels for WT (n = 12) and SR-B1− /− (n = 14) mice. Data are shown in box plots.
absence of SR-B1 during obesity stimulates hepatic VLDL-TG secretion
(p < 0.0001). In addition, analysis of the area under the curve (AUC) of
TG levels in lipoprotein fractions revealed a significant increase in obese
SR-B1-deficient vs. WT animals (+166%, p < 0.01) (Fig. 7B), which is
consistent with high baseline total TG levels observed above in this gene-
manipulated mouse strain.

3.6. SR-B1-deficiency differentially affects hepatic FA composition in


HFD-fed mice

Liver FA composition is closely related to the severity and progres­


sion of NAFLD [34]. Previous studies strongly suggest that β-oxidation
plays a crucial role in regulating the FA profile [35].
In order to evaluate if SR-B1 expression modifies hepatic FA content
during obesity, we compared liver FA composition in HFD-fed WT and
SR-B1− /− mice. We identified eight main FA (C16:0, C16:1, C18:0,
C18:1, C18:2, C20:4, C22:5, C22:6) that were considered in this analysis.
Principal component analysis using all liver FA showed that WT and SR-
B1− /− mice clustered in two independent groups (Fig. 8A), highlighting
the role of the SR-B1 expression in modulating hepatic FA composition.
In more detail, SR-B1-deficient mice fed a HFD showed lower C18:1
(oleic acid) content compared to HFD-fed WT mice (− 12%, p < 0.01)
(Fig. 8B). Furthermore, FA were grouped and analyzed based on three
main standard categories: saturated (SFA), monounsaturated (MUFA),
and polyunsaturated FA (PUFA). Fig. 8C depicts that the absence of SR-
B1 in obese mice was associated with overall reduced liver MUFA con­
tent (− 12%, p < 0.01), compared with WT obese mice. SFA and PUFA
liver contents remained unchanged between groups (p > 0.05).

4. Discussion

Chronic overnutrition and sedentary lifestyle drive the epidemic of


obesity in modern societies. Abnormalities in lipid and lipoprotein
metabolism accompanied by chronic inflammation usually present in
obese patients play an essential role in the development of several
obesity-related comorbidities [36,37].
Even though SR-B1 is classically known for its role in HDL meta­
bolism and CVD, additional functions in physiology and disease have
been described for this receptor [38]. Here, we evaluated the impact of
SR-B1 deficiency on adipocyte morphology, adipose tissue inflamma­
tion, and hepatic steatosis under diet-induced obesity. In the present
study, we demonstrated that the global deficiency of SR-B1 modifies the
metabolic environment promoting an obesity phenotype characterized
by inflammatory dyslipidemia associated with white adipocyte hyper­
trophy, increased adipose tissue macrophage infiltration, and attenu­
ated hepatic steatosis in mice under HFD.
Obesity is intimately associated with dyslipidemia and atheroscle­
rotic CVD. A hallmark of dyslipidemia in obesity is low HDL-C levels
[39]. However, recent evidence has demonstrated that cholesterol efflux
capacity, more than HDL-C levels, is impaired in obese patients or mice,
and is inversely associated with the incidence of cardiovascular events in
a population-based cohort [40,41]. Indeed, SR-B1 knock-out mice are
atherosclerosis prone despite exhibiting high HDL cholesterol levels
**p < 0.01; ****p < 0.001 vs. WT mice.

[12–14]. Interestingly, SR-B1-deficient HDL particles are abnormal in


composition and function, which most likely lead to decreased anti-
atherogenic activity [12–14]. This HDL dysfunctionality together with
the SR-B1-dependent pro-inflammatory profile observed in obese SR-
B1− /− mice may significantly increase the risk of atherosclerosis. Thus,
the mechanisms underlying obesity-associated compositional and
functional HDL alterations are of large importance.
Interestingly, previous studies reported an association between some
genetic variants in the human SR-B1 gene locus and body mass index
(BMI) [42,43]. Despite these human data, we did not observe any dif­
ference in murine body weight gain dependent on the SR-B1 genotype.
However, despite that the major cause of obesity and its co-morbidities

6
K. Rivera et al. BBA - Molecular and Cell Biology of Lipids 1866 (2021) 158909

Fig. 5. Liver tissue analysis and plasma enzyme levels in SR-B1− /− mice fed on a HFD for 12 weeks. Representative images of (A) livers and (B) H&E-stained liver
sections obtained from HFD-fed mice. (C) Mean liver lipid droplet area for WT (n = 12) and SR-B1− /− (n = 14) mice. (D) Hepatic TG content for WT (n = 12) and SR-
B1− /− (n = 14) mice. (E) Hepatic cholesterol content for WT (n = 7) and SR-B1− /− (n = 8) mice. Plasma (F) ALT and (F) GGT levels for WT (n = 7) and SR-B1− /− (n
= 9) mice. Data are shown in box plots. ****p < 0.001 vs. WT.

are related to increased food intake and body weight gain, not all in­ been reported in body weight gains and plasma TG levels in SR-B1-
dividuals become obese when consuming high-calorie diets [44]. Even deficient animals compared to WT mice [50–52], suggesting that
in some studies, overweight and obese patients have shown similar hypertriglyceridemia observed our study as well as in [45] is dependent
prognosis compared to normal-weight subjects, suggesting that BMI is on the exposure to the obesogenic high fat diet. It was previously shown
not always the best indicator of metabolic health [6]. that SR-B1 intestinal overexpression increases intestinal absorption of
Surprisingly, we found marked hypertriglyceridemia in SR-B1- TG, suggesting that high levels of plasma TG may also be explained in
deficient obese mice, compared to WT. This finding is consistent with part by differences in the uptake of intestinal lipids between both ge­
a very recent study that evaluated the potential relevance of SR-B1 notypes [53].
function in the development of obesity and associated metabolic com­ An additional critical event in obesity is macrophage infiltration of
plications in wild-type and SR-B1 deficient mice fed an obesogenic diet adipose tissue and macrophages resident within CLS in adipose tissue
enriched in fat [45], supporting previous observations that SR-B1 can are lipid-laden and pro-inflammatory [54]. A previous study investi­
participate in the clearance of circulating TG-rich lipoproteins [45–47]. gating the effect of SR-B1 and LDLR deletions on adipose tissue in mice
Based on combined data from prospective studies, TG levels are a risk fed with 20% HFD showed severe macrophage infiltration with evident
factor for CVD, independent of HDL-C levels [48]. Adipose tissue has a apoptosis in WAT [55]. Considering that SR-B1 mRNA is highly
central role in TG metabolism and proper WAT functionality is crucial expressed in differentiated 3T3 adipocytes and increases during differ­
for systemic metabolic homeostasis [27]. Recent observations indicate entiation [56], our data showing adipocyte hypertrophy and immune
that cholesterol imbalance may contribute to adipocyte dysfunction and cell recruitment by CLS presence suggest that SR-B1 deficiency may
elevated blood TG levels in obese states [49]. Interestingly, abnormal contribute to the obesogenic environment promoting higher circulating
cholesterol metabolism profoundly influences adipocyte homeostasis pro-inflammatory TNF-α and possibly by lowering anti-inflammatory IL-
mimicking that found in hypertrophied adipocytes in obesity [49]. 10 levels. These fidings are consistent with a recent report showing a
Previous studies have shown that HDL cholesterol levels are elevated in trend towards increased adipocyte size in chow-fed SR-B1− /− animals
SR-B1 deficient mice fed a normal diet most likely due to blocked RCT [45]. Further, systemic inflammatory response reported in obese SR-
[26]. However, under low-fat chow diet, no significant changes have B1− /− mice is similar to that previously observed in animals deficient in

7
K. Rivera et al. BBA - Molecular and Cell Biology of Lipids 1866 (2021) 158909

Fig. 6. Hepatic protein and gene expression analysis in SR-B1− /− mice fed on a HFD for 12 weeks. (A) Immunoblotting analysis and quantitation of PPARγ protein
level in total liver lysates for WT (n = 7) and SR-B1− /− (n = 7) mice. (B) Quantitative RT-PCR gene expression for WT (n = 6) and SR-B1− /− (n = 6) mice. Data are
presented as mean ± SD or shown in box plots. *p < 0.05; ***p < 0.001 vs. WT mice.

Fig. 7. Hepatic VLDL-TG secretion analysis in SR-B1− /− mice fed an HFD for 12 weeks. (A) TG content in lipoprotein fractions and (B) AUC of chromatogram after
Tyloxapol injection for WT (n = 6) and SR-B1− /− (n = 6) mice. Data are presented as mean ± SD or shown in box plots. **p < 0.01; ****p < 0.001 vs. WT mice.

this receptor during infection with bacterial pneumonia [57]. a positive effect of SR-B1 deficiency on glucose tolerance [45], our study
A previous study showed that supplementation with vitamin E, a showing baseline IR -as measured by HOMA-IR- in obese SR-B1− /− mice
potent lipid-soluble antioxidant, reduces adipose tissue inflammation suggests that SR-B1 expression may function as a protective mechanism
and improves the overall metabolic profile in a mouse model of diet- against risk of diabetes. However, this discrepancy in glucometabolic
induced obesity [58]. Moreover, vitamin E significantly improved findings can be explained by the fact that the aforementioned study [45]
liver function and histologic changes in patients with NAFLD [59]. These used pure C57BL/6 mice as control group compared most likely to SR-
data are relevant, considering that our group previously demonstrated B1− /− mice with mixed genetic background instead of WT littermates -as
that vitamin E metabolism is abnormal in SR-B1-deficient mice [60,61]. occurred in our study- and fed a different HFD, containing 45 and 35%
Interestingly, in an animal model of diet-induced IR, SR-B1 was upre­ kcal from fat and carbohydrates, relative to 60 and 20% kcal content,
gulated and redistributed from the cytoplasm to a predominantly peri­ respectively, in our experiments. In addition, Hoekstra and colleagues
nuclear localization in enterocytes, in association with an applied a glucose challenge and assessed glucose tolerance at 10 weeks
overproduction of intestinal apoB48-containing lipoproteins [62]. of feeding [45], whereas we only performed HOMA-IR evaluation in
Additionally, gamma-tocotrienol, a minor form of vitamin E, attenuated fasting blood samples after 12 weeks of HFD feeding. Thus, different
HFD-induced obesity and IR by inhibiting systemic and adipose control group comparators, potentially divergent SR-B1 KO murine
inflammation, as well as adipose tissue macrophage recruitment in mice strains maintained in different locations, dissimilar types of dietary in­
[63]. Thus, deficient SR-B1-dependent vitamin E delivery may have also terventions as well as the use different techniques to evaluate metabolic
contributed to the WAT phenotype of obese SR-B1− /− mice. and inflammatory changes may explain the significant diversity in body
Although a recent study reported a trend, but not significant, towards weight gain, adipokine levels, and glucose metabolism detected between

8
K. Rivera et al. BBA - Molecular and Cell Biology of Lipids 1866 (2021) 158909

Fig. 8. Liver FA composition in SR-B1− /− mice fed an HFD for 12 weeks. (A) Principal components analysis for WT and SR-B1− /− obese mice. Principal components 1
and 2 explained 66.5% of the liver FA composition variability. Percentage of (B) specific FA and (C) SFA, MUFA and PUFA content in livers of WT (n = 6) and SR-
B1− /− (n = 6) mice. Data are shown as mean ± SD. *p < 0.05; **p < 0.01 vs. WT mice.

these studies. Given the potentially opposite effects of SR-B1 expression on adipose
Subclinical low grade inflammation is induced by increased tissue pathobiology versus fatty liver development, additional research is
adiposity leading to significant co-morbidities in the liver, triggering a required to define more accurately the role of SR-B1 in different tissues
continuous vicious cycle [64]. The available evidence regarding the role under a variety of metabolic diseases.
of SR-B1 in the development of NAFLD is limited. A study showed that Among limitations of our study, we used an animal model with
adult C57BL/6 mice fed with high fat and cholesterol diet increased global SR-B1 deficiency, which makes it difficult to establish the tissue-
hepatic mRNA and protein levels of SR-B1 in mice with NAFLD [65]. On specific (e.g., fat versus liver) role of SR-B1 expression during obesity. In
the other hand, Pparg knockdown in mice improves fatty liver without addition, HFD intake for 12 weeks essentially promotes fatty liver, but it
alterations in Srebp1c mRNA levels [66], consistent with our results in is not a good model to evaluate the relevance of SR-B1 function in
mRNA levels of these genes in SR-B1-deficient obese animals. Further­ further NASH progression (e.g., inflammation and/or fibrosis develop­
more, SR-B1 was involved in hepatic VLDL remnant uptake in the ment). Future studies evaluating the role of SR-B1 in hepatic inflam­
absence of classical ApoE-recognizing receptors [67]. Also, less accu­ mation and fibrosis in more advanced murine models of NAFLD are
mulation of MUFA in obese animals lacking SR-B1 suggests that the needed.
quality of FA accumulated in the liver, not only total fat content, may be
critical to progression and severity of NAFLD. Indeed, a high hepatic 5. Conclusion
MUFA (palmitoleic acid)/SFA (palmitic acid) ratio appears to be an in­
dicator of more inflammatory NAFLD [34]. Thus, reduced hepatic MUFA We revealed liver and adipose tissue-specific changes underlying
content in obese SR-B1− /− mice may indicate attenuated risk of devel­ diet-induced obesity in SR-B1-deficient mice. As potential implications
oping more advanced liver disease. Remarkably, our data showed that of the current work, SR-B1 may be a therapeutic target not only in
SR-B1-deficient obese animals are less prone to overall hepatic TG atherosclerotic CVD settings, but also in obesity, which increases overall
deposition, which is consistent with previously published data [45,68]. chronic disease risk and mortality worldwide.
This may be explained by the dysregulation of transcriptional factors
(Pparα and Pparγ) as well as lipid transport proteins (Cpt1 and Apob) that Ethics approval and consent to participate
play a key role in FA catabolism and TG secretion, respectively.
Recently, an expert group suggested a new acronym (MAFLD, Protocols were conducted in agreement with the National Research
Metabolic Associated Fatty Liver Disease) for NAFLD, whichmore Council (NRC) publication Guide for Care and Use of Laboratory Ani­
accurately reflects current knowledge of fatty liver diseases associated mals. All the studies were approved by the Ethics and Animal Committee
with metabolic dysfunction [69]. Our data supports the hypothesis that for Animal Welfare from the School of Medicine of the Pontificia Uni­
variation in obesity phenotypes (e.g., systemic inflammation and liver versidad Católica de Chile (Protocol #14-036).
fat accumulation) may be modulated by SR-B1 expression and function.

9
K. Rivera et al. BBA - Molecular and Cell Biology of Lipids 1866 (2021) 158909

Consent for publication diseases, Eur. J. Intern. Med. 48 (2018) 6–17, https://doi.org/10.1016/j.
ejim.2017.10.020.
[7] S. Sookoian, C.J. Pirola, L. Valenti, N.O. Davidson, Genetic pathways in
Not applicable. nonalcoholic fatty liver disease: insights from systems biology, Hepatology (2020),
https://doi.org/10.1002/hep.31229.
[8] B. Choromanska, P. Mysliwiec, H.R. Hady, J. Dadan, H. Mysliwiec, T. Bonda, et al.,
Funding The implication of adipocyte ATP-binding cassette A1 and G1 transporters in
metabolic complications of obesity, J. Physiol. Pharmacol. 70 (2019), https://doi.
This work was funded, in part, by the National Agency for Research org/10.26402/jpp.2019.1.14.
[9] P.K. Rao, K. Merath, E. Drigalenko, A.Y.L. Jadhav, R.A. Komorowski, M.
and Development (ANID) program Fondo Nacional del Desarrollo I. Goldblatt, et al., Proteomic characterization of high-density lipoprotein particles
Científico y Tecnológico (FONDECYT) of Ministry of Science and in patients with non-alcoholic fatty liver disease, Clin. Proteomics 15 (2018) 10,
Technology, Government of Chile, through grants #1150399 (to A.R.), https://doi.org/10.1186/s12014-018-9186-0.
[10] N. Fan, L. Peng, Z. Xia, L. Zhang, Z. Song, Y. Wang, et al., Triglycerides to high-
#1180525 (to M.E.A.), #1180347 (to D.B), #11171001 (to D.C.), and
density lipoprotein cholesterol ratio as a surrogate for nonalcoholic fatty liver
#1191145 to (M.A.). This publication also has received funding from disease: a cross-sectional study, Lipids Health Dis. 18 (2019), https://doi.org/
grant Millennium Nucleus for Cardiovascular Magnetic Resonance 10.1186/s12944-019-0986-7.
supported by Millennium Science Initiative of the Ministry of Economy, [11] R. Fadaei, H. Poustchi, R. Meshkani, N. Moradi, T. Golmohammadi, S. Merat,
Impaired HDL cholesterol efflux capacity in patients with non-alcoholic fatty liver
Development and Tourism, Government of Chile and by Comisión disease is associated with subclinical atherosclerosis, Sci. Rep. 8 (2018), https://
Nacional de Investigación, Ciencia y Tecnología (CONICYT, doi.org/10.1038/s41598-018-29639-5.
AFB170005, CARE Chile UC). Ph.D. Fellowship ANID-PFCHA/ [12] A. Rigotti, H.E. Miettinen, M. Krieger, The role of the high-density lipoprotein
receptor SR-BI in the lipid metabolism of endocrine and other tissues, Endocr. Rev.
Doctorado Nacional/2020-21201346 (to K.R.); and ANID-PCHA/ 24 (2003) 357–387, https://doi.org/10.1210/er.2001-0037.
Doctorado Nacional/2016-21160835 (to A.X.). M.F-G was funded by [13] B.L. Trigatti, M. Krieger, A. Rigotti, Influence of the HDL receptor SR-BI on
Ministry of Economy, Industry and Competitiveness (MINECO-FEDER; lipoprotein metabolism and atherosclerosis, Arterioscler. Thromb. Vasc. Biol. 23
(2003) 1732–1738, https://doi.org/10.1161/01.ATV.0000091363.28501.84.
“Juan de la Cierva” Program IJCI-2016-30025) of Government of Spain. [14] A. Leiva, H. Verdejo, M.L. Benítez, A. Martínez, D. Busso, A. Rigotti, Mechanisms
The funding bodies had no role in the design of the study, the collection, regulating hepatic SR-BI expression and their impact on HDL metabolism,
analysis, and interpretation of data, or in writing the manuscript. Atherosclerosis 217 (2011) 299–307, https://doi.org/10.1016/j.
atherosclerosis.2011.05.036.
[15] G. Dagher, N. Donne, C. Klein, P. Ferré, I. Dugail, HDL-mediated cholesterol uptake
CRediT authorship contribution statement and targeting to lipid droplets in adipocytes, J. Lipid Res. 44 (2003) 1811–1820,
https://doi.org/10.1194/jlr.M300267-JLR200.
[16] L. Yvan-Charvet, A. Bobard, P. Bossard, F. Massiéra, X. Rousset, G. Ailhaud, et al.,
Katherine Rivera: Conceptualization, Methodology, Validation, In vivo evidence for a role of adipose tissue SR-BI in the nutritional and hormonal
Formal analysis, Investigation, Data curation, Writing – original draft, regulation of adiposity and cholesterol homeostasis, Arterioscler. Thromb. Vasc.
Writing – review & editing. Verónica Quiñones: Conceptualization, Biol. 27 (2007) 1340–1345, https://doi.org/10.1161/ATVBAHA.106.136382.
[17] Y. Zhang, F.C. McGillicuddy, C.C. Hinkle, S. O’Neill, J.M. Glick, G.H. Rothblat, et
Investigation, Formal analysis. Ludwig Amigo: Investigation. Nicolás al., Adipocyte modulation of high-density lipoprotein cholesterol, Circulation 121
Santander: Conceptualization, Writing – review & editing. Francisca (2010) 1347–1355, https://doi.org/10.1161/CIRCULATIONAHA.109.897330.
Salas-Pérez: Investigation, Writing – review & editing. Aline Xavier: [18] R.A.M. Ahmed, K. Murao, H. Imachi, X. Yu, J. Li, N.C.W. Wong, et al., Human
scavenger receptor class b type 1 is regulated by activators of peroxisome
Investigation, Writing – review & editing. Marta Fernández-Galilea:
proliferators-activated receptor-γ in hepatocytes, Endocrine 35 (2009) 233–242,
Conceptualization, Writing – review & editing. Gonzalo Carrasco: https://doi.org/10.1007/s12020-008-9142-2.
Investigation. Daniel Cabrera: Conceptualization, Writing – review & [19] W. Wang, Z. Yan, J. Hu, W.J. Shen, S. Azhar, F.B. Kraemer, Scavenger receptor
class B, type 1 facilitates cellular fatty acid uptake, Biochim. Biophys. Acta Mol.
editing. Marco Arrese: Conceptualization, Writing – review & editing.
Cell Biol. Lipids 2020 (1865) 158554, https://doi.org/10.1016/j.
Dolores Busso: Conceptualization, Resources, Writing – review & bbalip.2019.158554.
editing. Marcelo E. Andia: Conceptualization, Data curation, Re­ [20] K. Rivera, F. Salas-Pérz, G. Echeverría, I. Urquiaga, S. Dicenta, D. Pérez, et al., Red
sources, Writing – review & editing, Supervision. Attilio Rigotti: wine grape pomace attenuates atherosclerosis and myocardial damage and
increases survival in association with improved plasma antioxidant activity in a
Conceptualization, Resources, Writing – original draft, Writing – review murine model of lethal ischemic heart disease, Nutrients 11 (2019), https://doi.
& editing, Supervision, Funding acquisition. org/10.3390/nu11092135.
[21] J. Folch, M. Lees, G.H. Sloane Stanley, A simple method for the isolation and
purification of total lipides from animal tissues, J. Biol. Chem. 226 (1957)
Declaration of competing interest 497–509, https://doi.org/10.3989/scimar.2005.69n187.
[22] I. Murano, G. Barbatelli, V. Parisani, C. Latini, G. Muzzonigro, M. Castellucci, et al.,
Dead Adipocytes, Detected as Crown-like Structures, Are Prevalent in Visceral Fat
The authors declare that they have no competing interests. Depots of Genetically Obese Mice, 2008, https://doi.org/10.1194/jlr.M800019-
JLR200.
[23] M.W. Pfaffl, Relative expression software tool (REST(C)) for group-wise
Acknowledgments comparison and statistical analysis of relative expression results in real-time PCR,
Nucleic Acids Res. 30 (2002), 36e–36, https://doi.org/10.1093/nar/30.9.e36.
[24] X. Li, F. Catalina, S.M. Grundy, S. Patel, Method to measure apolipoprotein B-48
We thank Dr. Víctor Cortés for discussion at early stage of this study.
and B-100 secretion rates in an individual mouse: evidence for a very rapid
turnover of VLDL and preferential removal of B-48- relative to B-100-containing
References lipoproteins, J. Lipid Res. 37 (1996) 210–220.
[25] A. Xavier, F. Zacconi, C. Gainza, D. Cabrera, M. Arrese, S. Uribe, et al., Intrahepatic
fatty acids composition as a biomarker of NAFLD progression from steatosis to
[1] Obesidad y sobrepeso n.d. https://www.who.int/news-room/fact-sheets/detail/o
NASH by using 1H-MRS, RSC Adv. 9 (2019) 42132–42139, https://doi.org/
besity-and-overweight (accessed June 2, 2020).
10.1039/c9ra08914d.
[2] C. Koliaki, S. Liatis, A. Kokkinos, Obesity and cardiovascular disease: revisiting an
[26] A. Rigotti, B.L. Trigatti, M. Penman, H. Rayburn, J. Herz, M. Krieger, A targeted
old relationship, Metabolism 92 (2019) 98–107, https://doi.org/10.1016/j.
mutation in the murine gene encoding the high density lipoprotein (HDL) receptor
metabol.2018.10.011.
scavenger receptor class B type I reveals its key role in HDL metabolism, Proc. Natl.
[3] S.A. Polyzos, J. Kountouras, C.S. Mantzoros, Obesity and nonalcoholic fatty liver
Acad. Sci. U. S. A. 94 (1997) 12610–12615.
disease: from pathophysiology to therapeutics, Metabolism 92 (2019) 82–97,
[27] F. Haczeyni, K.S. Bell-Anderson, G.C. Farrell, Causes and mechanisms of adipocyte
https://doi.org/10.1016/j.metabol.2018.11.014.
enlargement and adipose expansion, Obes. Rev. 19 (2018) 406–420, https://doi.
[4] N.L. Gluchowski, M. Becuwe, T.C. Walther, R.V. Farese, Lipid droplets and liver
org/10.1111/obr.12646.
disease: from basic biology to clinical implications, Nat. Rev. Gastroenterol.
[28] M. Longo, F. Zatterale, J. Naderi, L. Parrillo, P. Formisano, G.A. Raciti, et al.,
Hepatol. 14 (2017) 343–355, https://doi.org/10.1038/nrgastro.2017.32.
Adipose tissue dysfunction as determinant of obesity-associated metabolic
[5] Z. Younossi, Q.M. Anstee, M. Marietti, T. Hardy, L. Henry, M. Eslam, et al., Global
complications, Int. J. Mol. Sci. 20 (2019), https://doi.org/10.3390/ijms20092358.
burden of NAFLD and NASH: trends, predictions, risk factors and prevention, Nat.
[29] K. Nimptsch, S. Konigorski, T. Pischon, Diagnosis of obesity and use of obesity
Rev. Gastroenterol. Hepatol. 15 (2018) 11–20, https://doi.org/10.1038/
biomarkers in science and clinical medicine, Metabolism 92 (2019) 61–70, https://
nrgastro.2017.109.
doi.org/10.1016/j.metabol.2018.12.006.
[6] A. Vecchié, F. Dallegri, F. Carbone, A. Bonaventura, L. Liberale, P. Portincasa, et
al., Obesity phenotypes and their paradoxical association with cardiovascular

10
K. Rivera et al. BBA - Molecular and Cell Biology of Lipids 1866 (2021) 158909

[30] L. Gan, W. Xiang, B. Xie, L. Yu, Molecular mechanisms of fatty liver in obesity, [51] A.C. Provost, L. Vede, K. Bigot, N. Keller, A. Tailleux, J.P. Jaïs, et al., Morphologic
Front. Med. 9 (2015) 275–287, https://doi.org/10.1007/s11684-015-0410-2. and electroretinographic phenotype of SR-BI knockout mice after a long-term
[31] Y.L. Zhang, A. Hernandez-Ono, P. Siri, S. Weisberg, D. Conlon, M.J. Graham, et al., atherogenic diet, Investig. Ophthalmol. Vis. Sci. 50 (2009) 3931–3942, https://doi.
Aberrant hepatic expression of PPARγ2 stimulates hepatic lipogenesis in a mouse org/10.1167/iovs.08-2527.
model of obesity, insulin resistance, dyslipidemia, and hepatic steatosis, J. Biol. [52] H. Wiersma, N. Nijstad, T. Gautier, J. Iqbal, F. Kuipers, M.M. Hussain, et al.,
Chem. 281 (2006) 37603–37615, https://doi.org/10.1074/jbc.M604709200. Scavenger receptor BI facilitates hepatic very low density lipoprotein production in
[32] M.A. de la Rosa Rodriguez, S. Kersten, Regulation of lipid droplet-associated mice, J. Lipid Res. 51 (2010) 544–553, https://doi.org/10.1194/jlr.M000844.
proteins by peroxisome proliferator-activated receptors, Biochim. Biophys. Acta [53] F. Bietrix, D. Yan, M. Nauze, C. Rolland, J. Bertrand-Michel, C. Coméra, et al.,
Mol. Cell Biol. Lipids 2017 (1862) 1212–1220, https://doi.org/10.1016/j. Accelerated lipid absorption in mice overexpressing intestinal SR-BI, J. Biol. Chem.
bbalip.2017.07.007. 281 (2006) 7214–7219, https://doi.org/10.1074/jbc.M508868200.
[33] B. Mittendorfer, M. Yoshino, B.W. Patterson, S. Klein, VLDL triglyceride kinetics in [54] Y. Ni, L. Ni, F. Zhuge, L. Xu, Z. Fu, T. Ota, Adipose tissue macrophage phenotypes
lean, overweight, and obese men and women, J. Clin. Endocrinol. Metab. 101 and characteristics: the key to insulin resistance in obesity and metabolic disorders,
(2016) 4151–4160, https://doi.org/10.1210/jc.2016-1500. Obesity 28 (2020) 225–234, https://doi.org/10.1002/oby.22674.
[34] K. Yamada, E. Mizukoshi, H. Sunagozaka, K. Arai, T. Yamashita, Y. Takeshita, et [55] X. Guo, J. Liao, X. Huang, Y. Wang, W. Huang, G. Liu, Reversal of adipose tissue
al., Characteristics of hepatic fatty acid compositions in patients with nonalcoholic loss by probucol in mice with deficiency of both scavenger receptor class B type 1
steatohepatitis, Liver Int. 35 (2015) 582–590, https://doi.org/10.1111/liv.12685. and LDL receptor on high fat diet, Biochem. Biophys. Res. Commun. 497 (2018)
[35] C.O. de Souza, A.A.S. Teixeira, L.A. Biondo, E.A. Lima Junior, H.A.P. Batatinha, J. 930–936, https://doi.org/10.1016/j.bbrc.2017.05.017.
C. Rosa Neto, Palmitoleic acid improves metabolic functions in fatty liver by [56] S.L. Acton, P.E. Scherer, H.F. Lodish, M. Krieger, Expression cloning of SR-BI, a
PPARα-dependent AMPK activation, J. Cell. Physiol. 232 (2017) 2168–2177, CD36-related class B scavenger receptor, J. Biol. Chem. 269 (1994) 21003–21009.
https://doi.org/10.1002/jcp.25715. [57] K.M. Gowdy, J.H. Madenspacher, K.M. Azzam, K.A. Gabor, K.S. Janardhan, J.
[36] J. Vekic, A. Zeljkovic, A. Stefanovic, Z. Jelic-Ivanovic, V. Spasojevic-Kalimanovska, J. Aloor, et al., Key role for scavenger receptor B-I in the integrative physiology of
Obesity and dyslipidemia, Metabolism 92 (2019) 71–81, https://doi.org/10.1016/ host defense during bacterial pneumonia, Mucosal Immunol. 8 (2015) 559–571,
j.metabol.2018.11.005. https://doi.org/10.1038/mi.2014.88.
[37] T. Zhang, J. Chen, X. Tang, Q. Luo, D. Xu, B. Yu, Interaction between adipocytes [58] M. Alcalá, I. Sánchez-Vera, J. Sevillano, L. Herrero, D. Serra, M.P. Ramos, et al.,
and high-density lipoprotein:new insights into the mechanism of obesity-induced Vitamin E reduces adipose tissue fibrosis, inflammation, and oxidative stress and
dyslipidemia and atherosclerosis, Lipids Health Dis. 18 (2019), https://doi.org/ improves metabolic profile in obesity, Obesity 23 (2015) 1598–1606, https://doi.
10.1186/s12944-019-1170-9. org/10.1002/oby.21135.
[38] M. Hoekstra, M. Sorci-Thomas, Rediscovering scavenger receptor type BI: [59] K. Sato, M. Gosho, T. Yamamoto, Y. Kobayashi, N. Ishii, T. Ohashi, et al., Vitamin E
surprising new roles for the HDL receptor, Curr. Opin. Lipidol. 28 (2017) 255–260, has a beneficial effect on nonalcoholic fatty liver disease: a meta-analysis of
https://doi.org/10.1097/MOL.0000000000000413. randomized controlled trials, Nutrition 31 (2015) 923–930, https://doi.org/
[39] S. Lamon-Fava, P.W.F. Wilson, E.J. Schaefer, Impact of body mass index on 10.1016/j.nut.2014.11.018.
coronary heart disease risk factors in men and women: the Framingham Offspring [60] P. Mardones, P. Strobel, S. Miranda, F. Leighton, V. Quiñones, L. Amigo, et al.,
Study, Arterioscler. Thromb. Vasc. Biol. 16 (1996) 1509–1515, https://doi.org/ α-Tocopherol metabolism is abnormal in scavenger receptor class B type I (SR-BI)-
10.1161/01.ATV.16.12.1509. deficient mice, J. Nutr. 132 (2002) 443–449, https://doi.org/10.1093/jn/
[40] A. Rohatgi, A. Khera, J.D. Berry, E.G. Givens, C.R. Ayers, K.E. Wedin, et al., HDL 132.3.443.
cholesterol efflux capacity and incident cardiovascular events, N. Engl. J. Med. 371 [61] L.E. Wright, D. Vecellio Reane, G. Milan, A. Terrin, G. Di Bello, A. Belligoli, et al.,
(2014) 2383–2393, https://doi.org/10.1056/NEJMoa1409065. Increased mitochondrial calcium uniporter in adipocytes underlies mitochondrial
[41] M.N. Duong, K. Uno, V. Nankivell, C. Bursill, S.J. Nicholls, Induction of obesity alterations associated with insulin resistance, Am. J. Physiol. Endocrinol. Metab.
impairs reverse cholesterol transport in ob/ob mice, PLoS One 13 (2018), https:// 313 (2017) E641–E650, https://doi.org/10.1152/ajpendo.00143.2016.
doi.org/10.1371/journal.pone.0202102. [62] A.A. Hayashi, J. Webb, J. Choi, C. Baker, M. Lino, B. Trigatti, et al., Intestinal SR-BI
[42] S. Acton, D. Osgood, M. Donoghue, D. Corella, M. Pocovi, A. Cenarro, et al., is upregulated in insulin-resistant states and is associated with overproduction of
Association of polymorphisms at the SR-BI gene locus with plasma lipid levels and intestinal apoB48-containing lipoproteins, Am. J. Physiol. Gastrointest. Liver
body mass index in a white population, Arterioscler. Thromb. Vasc. Biol. 19 (1999) Physiol. 301 (2011), https://doi.org/10.1152/ajpgi.00425.2010.
1734–1743, https://doi.org/10.1161/01.ATV.19.7.1734. [63] L. Zhao, I. Kang, X. Fang, W. Wang, M.A. Lee, R.R. Hollins, et al., Gamma-
[43] D.J. Koumanis, N.V. Christou, X.L. Wang, B.M. Gilfix, Pilot study examining the tocotrienol attenuates high-fat diet-induced obesity and insulin resistance by
frequency of several gene polymorphisms in a morbidly obese population, Obes. inhibiting adipose inflammation and M1 macrophage recruitment, Int. J. Obes. 39
Surg. 12 (2002) 759–764, https://doi.org/10.1381/096089202320995529. (2015) 438–446, https://doi.org/10.1038/ijo.2014.124.
[44] M.A. Cornier, A.K. Salzberg, D.C. Endly, D.H. Bessesen, D.C. Rojas, J.R. Tregellas, [64] I. Cantero, I. Abete, N. Babio, F. Arós, D. Corella, R. Estruch, et al., Dietary
The effects of overfeeding on the neuronal response to visual food cues in thin and Inflammatory Index and liver status in subjects with different adiposity levels
reduced-obese individuals, PLoS One 4 (2009), https://doi.org/10.1371/journal. within the PREDIMED trial, Clin. Nutr. 37 (2018) 1736–1743, https://doi.org/
pone.0006310. 10.1016/j.clnu.2017.06.027.
[45] M. Hoekstra, A.B. Ouweneel, J. Price, R. van der Geest, R.J. van der Sluis, J. [65] Y. Qiu, S. Liu, H.-T. Chen, C.-H. Yu, X.-D. Teng, H.-T. Yao, et al., Upregulation of
J. Geerling, et al., SR-BI deficiency disassociates obesity from hepatic steatosis and caveolin-1 and SR-B1 in mice with non-alcoholic fatty liver disease, Hepatobiliary
glucose intolerance development in high fat diet-fed mice, J. Nutr. Biochem. 89 Pancreat. Dis. Int. 12 (2013) 630–636, https://doi.org/10.1016/S1499-3872(13)
(2021) 108564, https://doi.org/10.1016/j.jnutbio.2020.108564. 60099-5.
[46] M. Van Eck, M. Hoekstra, R. Out, I.S.T. Bos, J.K. Kruijt, R.B. Hildebrand, et al., [66] T. Yamazaki, S. Shiraishi, K. Kishimoto, S. Miura, O. Ezaki, An increase in liver
Scavenger receptor BI facilitates the metabolism of VLDL lipoproteins in vivo, PPARγ2 is an initial event to induce fatty liver in response to a diet high in butter:
J. Lipid Res. 49 (2008) 136–146, https://doi.org/10.1194/jlr.M700355-JLR200. PPARγ2 knockdown improves fatty liver induced by high-saturated fat, J. Nutr.
[47] R. Out, J.K. Kruijt, P.C.N. Rensen, R.B. Hildebrand, P. De Vos, M. Van Eck, et al., Biochem. 22 (2011) 543–553, https://doi.org/10.1016/j.jnutbio.2010.04.009.
Scavenger receptor BI plays a role in facilitating chylomicron metabolism, J. Biol. [67] L. Hu, C.C. Van der Hoogt, S.M.S. Espirito Santo, R. Out, K.E. Kypreos, B.J.M. Van
Chem. 279 (2004) 18401–18406, https://doi.org/10.1074/jbc.M401170200. Vlijmen, et al., The hepatic uptake of VLDL in lrp-ldlr− /− vldlr− /− mice is
[48] J.E. Hokanson, M.A. Austin, Plasma triglyceride level is a risk factor for regulated by LPL activity and involves proteoglycans and SR-BI, J. Lipid Res. 49
cardiovascular disease independent of high-density lipoprotein cholesterol level: a (2008) 1553–1561, https://doi.org/10.1194/jlr.M800130-JLR200.
metaanalysis of population-based prospective studies, Eur. J. Cardiovasc. Prev. [68] E.A. Karavia, N.I. Papachristou, G.C. Sakellaropoulos, E. Xepapadaki,
Rehabil. 3 (1996) 213–219, https://doi.org/10.1177/174182679600300214. E. Papamichail, P.-I. Petropoulou, et al., Scavenger receptor class B type I regulates
[49] B.L. Yu, S.P. Zhao, J.R. Hu, Cholesterol imbalance in adipocytes: a possible plasma apolipoprotein E levels and dietary lipid deposition to the liver,
mechanism of adipocytes dysfunction in obesity, Obes. Rev. 11 (2010) 560–567, Biochemistry 54 (2015) 5605–5616, https://doi.org/10.1021/acs.
https://doi.org/10.1111/j.1467-789X.2009.00699.x. biochem.5b00700.
[50] R. Martins Cardoso, E. Creemers, S. Absalah, M. Hoekstra, G.S. Gooris, J. [69] M. Eslam, A.J. Sanyal, J. George, A. Sanyal, B. Neuschwander-Tetri, C. Tiribelli, et
A. Bouwstra, et al., Hyperalphalipoproteinemic scavenger receptor BI knockout al., MAFLD: a consensus-driven proposed nomenclature for metabolic associated
mice exhibit a disrupted epidermal lipid barrier, Biochim. Biophys. Acta Mol. Cell fatty liver disease, Gastroenterology 158 (2020), 1999–2014.e1, https://doi.org/
Biol. Lipids 2020 (1865) 158592, https://doi.org/10.1016/j.bbalip.2019.158592. 10.1053/j.gastro.2019.11.312.

11

You might also like