You are on page 1of 13

The FASEB Journal • Research Communication

CD36-dependent signaling mediates fatty acid-induced


gut release of secretin and cholecystokinin
Sinju Sundaresan,* Rafiq Shahid,‡ Terrence E. Riehl,† Rashmi Chandra,‡
Fatiha Nassir,* William F. Stenson,† Rodger A. Liddle,‡ and Nada A. Abumrad*
*Department of Medicine, Center for Human Nutrition, and †Division of Gastroenterology,
Washington University School of Medicine, St. Louis, Missouri, USA; and ‡Department of Medicine,
Division of Gastroenterology, Duke University, North Carolina, USA

ABSTRACT Genetic variants in the fatty acid (FA) secretin. These peptides contribute to the role of CD36 in
translocase FAT/CD36 associate with abnormal post- fat absorption and to its pleiotropic metabolic effects.—
prandial lipids and influence risk for the metabolic Sundaresan, S., Shahid, R., Riehl, T. E., Chandra, R.,
syndrome. CD36 is abundant on apical enterocyte Nassir, F., Stenson, W. F., Liddle, R. A., Abumrad, N. A.
membranes in the proximal small intestine, where it CD36-dependent signaling mediates fatty acid-induced
facilitates FA uptake and FA-initiated signaling. We gut release of secretin and cholecystokinin. FASEB J. 27,
explored whether CD36 signaling influences FA-medi- 1191–1202 (2013). www.fasebj.org
ated secretion of cholecystokinin (CCK) and secretin,
peptides released by enteroendocrine cells (EECs) in
the duodenum/jejunum, which regulate events impor- Key Words: CCK 䡠 cAMP 䡠 calcium
tant for fat digestion and homeostasis. CD36 was
immunodetected on apical membranes of secretin- and Dietary fatty acids (FAs) trigger release of a number
CCK-positive EECs and colocalized with cytosolic gran- of peptides, including cholecystokinin (CCK) and se-
ules. Intragastric lipid administration to CD36ⴚ/ⴚ mice cretin, by enteroendocrine cells (EECs) (1, 2). CCK is
released less secretin (ⴚ60%) and CCK (ⴚ50%) com- secreted by I cells (3, 4) and secretin by S cells (5–7),
pared with wild-type mice. Likewise, diminished secre- localized primarily to the duodenum and jejunum.
tin and CCK responses to FA were observed with Both peptides influence fat absorption and participate
CD36ⴚ/ⴚ intestinal segments in vitro, arguing against in nutrient sensing and regulation of energy balance
influence of alterations in fat absorption. Signaling (1, 8, 9). CCK regulates gallbladder contraction, secre-
mechanisms underlying peptide release were examined tions by the stomach and acinar pancreas, and colonic
in STC-1 cells stably expressing human CD36 or a motility (10). Secretin plays an important role in buff-
signaling-impaired mutant (CD36K/A). FA stimulation ering the acidic chyme in the intestinal lumen by
of cells expressing CD36 (vs. vector or CD36K/A) stimulating water and bicarbonate secretion and en-
released more secretin (3.5- to 4-fold) and CCK (2- to hances CCK effects on the acinar pancreas and gall-
3-fold), generated more cAMP (2- to 2.5-fold), and bladder (11). In addition, both CCK and secretin have
enhanced protein kinase A activation. Protein kinase A prosatiety effects (12, 13).
inhibition (H-89) blunted secretin (80%) but not CCK A number of cellular surface receptors for FA, includ-
release, which was reduced (50%) by blocking of cal- ing the family of G-protein-coupled receptors (GPRs)
modulin kinase II (KN-62). Coculture of STC-1 cells and the scavenger receptor CD36, have been identified.
with Caco-2 cells stably expressing CD36 did not alter Long-chain FAs are recognized by GPR120, GPR40, and
secretin or CCK release, consistent with a minimal CD36 (14 –16). CD36 facilitates cellular FA uptake (14)
effect of adjacent enterocytes. In summary, CD36 is a and, like the GPRs (17, 18), mediates FA-induced signal
major mediator of FA-induced release of CCK and transduction, influencing cellular calcium for release of
neurotransmitters (19) and arachidonic acid and pros-
taglandins (20). In taste cells, FA sensing by CD36
Abbreviations: BSA, bovine serum albumin; CaM-KII, cal-
modulin kinase II; CCK, cholecystokinin; DHA, docosa- mediates fat perception and preference (21, 22) and
hexaenoic acid; DMEM, Dulbecco’s modified Eagle’s me- induces the cephalic phase of digestion (22). In hu-
dium; EEC, enteroendocrine cell; ELISA, enzyme-linked mans, oral FA detection thresholds are higher in sub-
immunosorbent assay; FA, fatty acid; FBS, fetal bovine serum;
GFP, green fluorescent protein; GPR, G-protein-coupled re-
1
ceptor; HBSS, Hanks’ balanced saline solution; IBMX, Correspondence: Department of Medicine, Center for
3-isobutyl-1-methylxanthine; LA, linoleic acid; OA, oleic acid; Human Nutrition, 660 S. Euclid Ave., Campus Box 8031, St.
PA, palmitic acid; PKA, protein kinase A; PMSF, phenylmeth- Louis, MO 63110, USA. E-mail: nabumrad@dom.wustl.edu
anesulfonyl fluoride; RIA, radioimmunoassay; SNP, single- doi: 10.1096/fj.12-217703
nucleotide polymorphism; SSO, sulfo-N-succinimidyl oleate; This article includes supplemental data. Please visit http://
WT, wild type www.fasebj.org to obtain this information.

0892-6638/13/0027-1191 © FASEB 1191


jects carrying single-nucleotide polymorphisms (SNPs) for CCK immunohistochemistry, were developed by the Mu-
that reduce CD36 levels (23). tant Mouse Regional Resource Center (University of Mis-
CD36 and the GPRs have distinct distribution pat- souri, St. Louis, MO, USA). In these mice, GFP expression is
driven by the CCK promoter, allowing detection of CCK-
terns in the intestine. CD36 is abundant in the proxi- positive cells by immunofluorescence (33). Animals were
mal small intestine (24, 25) on villi enterocytes, where denied access to food for 16 h with ad libitum access to water
it contributes to FA and cholesterol uptake (25) and to before the small intestine was harvested.
signaling events important for initiating chylomicron
formation (26 –28). The GPRs (GPR120 and GPR40) Evaluation of gastric emptying
are more abundant in distal segments, localize primar-
ily to EECs, and were shown to mediate FA-induced Gastric emptying was measured as described previously (34).
peptide release (17, 29). The signaling pathways in- In brief, 1 ml of phenol red (100 ␮g/ml) was administered
volved in peptide release involve cAMP and protein orally to mice after overnight food withdrawal, and stomachs
kinase A (PKA) as well as PKA-independent mecha- were collected 15 min later. The residual phenol (A) was
recovered by rinsing with 10 ml of Na2HPO4 solution (0.1 M),
nisms (30 –32). Based on its ability for FA sensing and
and aliquots were diluted 1.5-fold (S1) and 5-fold (S2) before
signal transduction, CD36 could contribute to the measurement of absorbance at 570 nm (BioTek Instruments,
regulation of EEC peptide release. However, this possi- Winooski, VT, USA). The concentration was obtained from a
bility had been unexplored and was examined in the calibration curve. Dye recovered from stomachs of mice
present work. Using CD36⫺/⫺ and wild-type (WT) sacrificed immediately after dye administration served as
mice, we documented in vivo and in vitro, using intes- controls (B). Percentage of gastric emptying was calculated as
tinal segments, robust effects of CD36 on FA-induced 100 ⫺ (A/B) ⫻ 100.
release of secretin and CCK. In studies using STC-1
cells, we examined the mechanisms underlying CD36- Immunohistochemistry
mediated effects and demonstrated involvement of
cAMP and calcium. Mice were administered a mixture of 5-bromo-2=-deoxyuri-
dine (120 mg/kg) and 5-fluoro-2=-deoxyuridine (12 mg/kg)
intraperitoneally 90 min before sacrifice to label S-phase cells.
Proximal and distal small intestines were opened longitudi-
MATERIALS AND METHODS nally, fixed in 10% formalin, and paraffin embedded. Cut
sections (5 ␮m) were deparaffinized, followed by antigen
retrieval (99°C, 18 min) in a pressurized chamber (Biocare
Medical, Concord, CA, USA). Sections were incubated (1 h)
Materials in donkey serum (2%) and BSA (3%) to block nonspecific
binding and then were incubated overnight (4°C) with pri-
Antibodies used are listed in Table 1. FA-free bovine serum mary antibodies, followed by fluorescently labeled (Alexa
albumin (BSA) fraction IV, PKA inhibitor H-89 dihydrochlo- Fluor) secondary antibodies (1 h, 1:200). Images were taken
ride (N-[2-(p-bromocinnamylamino)ethyl]-5-isoquinolinesul- using a Zeiss inverted fluorescent microscope (Carl Zeiss Inc.,
fonamide), cAMP inhibitor MDL-12,330A hydrochloride [cis- Thornwood, NY, USA). For colocalization, doubly positive
N-(2-phenylcyclopentyl)-azacyclotridec-1-en-2-amine], and cells at ⫻400 were counted for 5 mice/antigen. For CCK,
calmodulin kinase inhibitor II (CaM-KII) KN-62 were from Sigma- immunohistochemical analysis was performed as described
Aldrich (St. Louis, MO, USA). previously (33).

Animals Secretin and CCK release in vivo

CD36-null (CD36⫺/⫺) and WT mice on the C57BL/6 After overnight food withdrawal, mice were administered an
background were housed in a facility with a 12-h light-dark intragastric load of olive oil or saline (16 ␮l/g body weight),
cycle and fed chow ad libitum (Purina, St. Louis, MO, USA). and peptide release into the blood was measured 30 min
Female mice, 3– 4 mo old, were denied access to food for later. CCK and secretin were extracted on C18 columns
16 h before euthanasia. Mouse care and use followed (Waters, Milford, MA, USA) and quantified. Plasma CCK was
guidelines of the animal ethics committee of Washington measured by bioassay using amylase secretion from isolated
University School of Medicine (St. Louis, MO, USA). rat pancreatic acini (35). Plasma from 3 mice was pooled to
Transgenic CCK-green fluorescent protein (GFP) mice, used obtain adequate sensitivity (36). Secretin was measured by

TABLE 1. Antibodies

Antibodies Host Source Dilution

CD36 Goat R&D Systems (Minneapolis, MN, USA) 1:100


␤-Actin Mouse Santa Cruz Biotechnology (Santa Cruz, CA, USA) 1:10,000
Chromogranin A Rabbit Abcam (Cambridge, MA, USA 1:200
GFP Chicken Abcam 1:1000
Phospho-PKA substrates (RRXS*/T*) Rabbit Cell Signaling Technology (Danvers, MA, USA) 1:1000
PKA C-␣ Rabbit Cell Signaling Technology 1:1000
Ran Goat Santa Cruz Biotechnology 1:10,000
Secretin Rabbit Phoenix Pharmaceuticals (Belmont, CA, USA) 1:1000

1192 Vol. 27 March 2013 The FASEB Journal 䡠 www.fasebj.org SUNDARESAN ET AL.
enzyme-linked immunosorbent assay (ELISA; Phoenix Phar- 37°C) in HBSS containing 1 mM 3-isobutyl-1-methylxanthine
maceuticals, Belmont, CA, USA). (IBMX). Cellular protein was determined in lysates from
duplicate wells (DC Protein Assay; Bio-Rad Laboratories,
Secretion by proximal intestinal loops Hercules, CA, USA). PKA activation was determined from
phosphorylation of PKA substrates (with the RRXS/T motif)
and nuclear content of the catalytic PKA subunit PKA-C␣.
The proximal small intestine isolated from WT and CD36⫺/⫺ Intracellular calcium was measured as described previously
mice after 16 h of food withdrawal was cut into two 10-cm (20), using Fura-2/AM dye (2.5 ␮M).
segments, starting at 2 cm after the pylorus. The pieces were
filled with Hanks’ balanced saline solution (HBSS) with or
without 100 or 300 ␮M linoleic acid (LA; plus protease and Western blot analyses
phosphatase inhibitors), tied at both ends, and incubated for
1 h at 37°C. The media were collected, lyophilized, rehy- Protein signals were detected using the Odyssey Infrared
drated with 500 ␮l, and assayed. Secretin was measured by System (Li-Cor Biosciences, Lincoln, NE, USA) as described
ELISA, and CCK was quantified by radioimmunoassay (RIA) previously (20). Cell proteins separated and transferred to
using 125I-labeled CCK-8 and antisera 92128, which recognize polyvinylidene fluoride membranes were blocked and incu-
biologically active CCK (37). bated overnight with primary antibodies (4°C) and then for 1
h with infrared dye-labeled secondary antibodies (room tem-
perature).
STC-1 and Caco-2 cells
Statistical analyses
The mouse EEC line (STC-1) was cultured in Dulbecco’s
modified Eagle’s medium (DMEM; Invitrogen, Carlsbad, CA,
USA) with 20% fetal bovine serum (FBS). STC-1 cells stably Statistical analyses were performed with GraphPad Prism 4
expressing human WT or mutated CD36 (CD36K/A) with software (GraphPad Software Inc., San Diego, CA, USA).
C-terminal lysines 469 and 472 substituted by alanine (38) Differences obtained by 1-way ANOVA were considered sig-
were generated by electroporation (Nucleofector Kit V; nificant at values of P ⱕ 0.05. The Bonferroni test was
Lonza, Cologne, Germany) followed by selection in gentami- performed to identify groups that were different.
cin (400 ␮g/ml). Caco-2 cells expressing WT CD36 or the
mutated form CD36K/A were generated as described for
STC-1 cells and maintained in DMEM with 20% FBS. RESULTS
Cocultures of enterocytes (Caco-2 cells) with EECs (STC-1
cells) were established by serial seeding. First, 3.0 ⫻ 106
CD36⫺/⫺ mice show reduced levels of intestinal pep-
differentiated Caco-2 cells expressing CD36 or empty vector
were seeded in DMEM containing 10% FBS. Twelve hours tides in response to a lipid load: CD36 is abundant in
later, 1 ml of culture medium containing 0.3 ⫻ 106 STC-1 the proximal intestine, where it has been shown to
cells, with or without CD36 expression, was added to the dish. facilitate uptake of FA and cholesterol (25) and to
A 1:10 ratio of STC-1 cells to Caco-2 cells was chosen to promote chylomicron formation (26). We examined
simulate the relative physiological distribution. One day later, the influence of CD36 deletion on fat-induced secre-
the mixed culture was washed, serum starved for 8 h, and tion of CCK and secretin, peptides with important roles
incubated (60 min, 37°C) in HBSS (with Ca2⫹ and Mg2⫹)
in fat absorption that are released by EECs localized
with 50 and 200 ␮M LA (plus 5 ␮M BSA) or with BSA alone
(controls). Secretin and CCK release was determined by primarily in proximal segments (3–7). Plasma CCK and
ELISA and RIA (37), respectively. secretin levels were measured in WT and CD36⫺/⫺
mice 30 min after an intragastric load of olive oil.
Peptide release by STC-1 cells CD36⫺/⫺ mice had 50% lower CCK (Fig. 1A) and 60%
lower secretin levels (Fig. 1B) than WT mice. Peptide
Cells (1⫻106) in 6-well plates were serum starved in DMEM levels were similar in saline-administered WT and
and then were stimulated (1 h) by addition of 50 –200 ␮M FA; CD36⫺/⫺ mice.
LA, docosahexaenoic acid (DHA), oleic acid (OA), and CD36 deficiency alters gut fat absorption by impair-
palmitic acid (PA) complexed to 5 ␮M BSA were added in ing chylomicron formation and shifting more luminal
HBSS (no glucose) containing aprotinin (200 kallikrein- fat to distal parts of the small intestine (26, 28). These
inhibiting units/ml). Media were collected for secretin anal-
yses by ELISA. Cells were lysed for protein assays in buffer [20
changes might contribute to the reduction of fat-
mM Tris-HCl, pH 7.5; 150 mM NaCl; 1 mM Na2EDTA; 1 mM induced CCK and secretin release in CD36⫺/⫺ mice by
EGTA; 1% Triton X-100; 2.5 mM sodium pyrophosphate; 1 altering fat exposure of EECs in the proximal part of
mM glycerol 2-phosphate; 1 mM Na3VO4; 1 ␮g/ml leupeptin; the intestine. We first compared gastric emptying rates
and 1 mM phenylmethanesulfonyl fluoride (PMSF)] contain- in WT and CD36⫺/⫺ mice and found no significant
ing a protease inhibitor mix (Roche Diagnostics, Inc., India- differences between the two groups (68.45⫾4.34 vs.
napolis, IN, USA). CCK release was measured by RIA (37). 71.32⫾6.39, n⫽4). To directly examine the role of
CD36 in FA-induced release of secretin and CCK inde-
Measurement of cAMP, PKA activation, and pendent of fat absorption, intestinal segments were
intracellular Ca2ⴙ
isolated from WT and CD36⫺/⫺ mice and tested for
FA-induced peptide release. Secretin and CCK release
Intracellular cAMP was measured using the DetectX Direct
High Sensitivity Cyclic AMP Chemiluminescent Immunoassay
were similar in WT and CD36⫺/⫺ segments under basal
Kit (Arbor Assays, Ann Arbor, MI, USA). In brief, serum- conditions (data not shown). Addition of 100 and 300
starved STC-1 cells (5⫻105/well) were stimulated with FA ␮M LA enhanced CCK release from WT segments by
(100 ␮M) complexed with BSA (5 ␮M) or BSA alone (30 min, 2.3- and 3.6-fold above basal. No enhancement was

CD36 MEDIATES RELEASE OF GUT PEPTIDES 1193


Figure 1. CCK and secretin release in vivo or in vitro by incubated intestinal segments from WT and CD36⫺/⫺ mice. A,
B) In vivo. After overnight food withdrawal, mice were administered an intragastric load of olive oil or saline (16 ␮l/g body
weight), and peptide release into the blood was measured 30 min later. CCK (A) and secretin (B) were extracted on C18
columns and quantified by bioassay or ELISA, respectively. All assays were performed on plasma pooled from 3 mice per
genotype per treatment. Data are means ⫾ se [n of pooled (3/pool) samples⫽3]. C, D) Intestinal segments in vitro.
Proximal intestinal segments (10-cm length, beginning at 2 cm after the pylorus) isolated from mice after 16 h of food
withdrawal, were filled with HBSS buffer with or without 100 or 300 ␮M LA, tied at both ends, and incubated at 37°C for
1 h. CCK (C) and secretin (D) release was determined by RIA or ELISA, respectively. Data are means ⫾ se (n⫽3 separate
mice per genotype per treatment). Release data are expressed as fold increase above buffer controls without LA. *P ⬍ 0.05;
**P ⬍ 0.01; ***P ⬍ 0.001.

observed in CD36⫺/⫺ segments with 100 ␮M LA, and a lial CD36 expression was down-regulated in intestines
1.6-fold enhancement occurred at 300 ␮M. Secretin from fed (Supplemental Fig. S2C, D) compared with
release from WT segments increased 3.3- and 4.3-fold unfed mice (Supplemental Fig. S2A, B). CD36 was
above basal in response to 100 and 300 ␮M LA. In expressed on nonepithelial cells throughout the small
contrast, the same LA concentrations increased re- intestine, and nonepithelial expression also declined
lease only 1.4- and 1.8-fold in CD36⫺/⫺ segments. with feeding (Supplemental Fig. S2).
These data suggested a major role of CD36 in We next examined expression of CD36 on EECs.
fat-induced EEC secretion of CCK and secretin that Chromogranin A is an essential component of secretory
was independent of the effects of CD36 on luminal vesicles typical of EECs and a marker for most cell types
fat absorption. of the enteroendocrine lineage (41). We costained for
chromogranin A and CD36 in intestinal sections and
CD36 expression in the small intestine and found that a subset of chromogranin A-positive cells
colocalization with chromogranin A express CD36 in the proximal (Fig. 2A) and distal small
CD36 is expressed apically on enterocytes of the small intestine (data not shown), as determined by merge of
intestine (39) after a proximal to distal decreasing fluorescent signals. To quantify the distribution of
gradient (24, 25), and its levels are down-regulated by CD36 across chromogranin A-positive cells, doubly
small amounts of dietary fat (40). Information about positive cells (for chromogranin A and CD36) were
CD36 expression on EECs and whether it is regulated counted and expressed as a percentage of cells positive
by dietary fat is unavailable so this was examined. for chromogranin A only. Sections from 5 mice were
Consistent with previous observations, CD36 was abun- used for quantification. This process demonstrated that
dant on epithelial cells of the duodenum and jejunum ⬃35 and 16% of chromogranin A-positive EECs in the
and less abundant in the ileum (Supplemental Fig. proximal and distal small intestine, respectively, stained
S1A⫺C). No CD36 signal was detected in intestines for both CD36 and chromogranin A (Table 2). Images
from CD36⫺/⫺ mice (Supplemental Fig. S1D). Epithe- at ⫻200 showed that CD36 is expressed on one-third of

1194 Vol. 27 March 2013 The FASEB Journal 䡠 www.fasebj.org SUNDARESAN ET AL.
Figure 2. CD36 expression on intestinal EECs.
A) CD36 (green) on chromogranin A (red)-positive
EECs. B, C) Colocalization of CD36 with secretin
(red)-positive cells. CD36 staining may be present on
apical membranes (arrows) of secretin-positive cells.
Nuclei are blue (DAPI). Under the same conditions, no CD36 signal is detected in CD36⫺/⫺ intestines (Supplemental Fig.
S1D). Immunoreactivity was visualized using Alexa Fluor 488 and 594 conjugates and a Zeiss fluorescence microscope
(⫻400). D, E) CD36 is expressed on one-third of chromogranin A-positive EECs (D) and on one-fifth of secretin-positive (E)
EECs (⫻200). Formalin-fixed, paraffin-embedded sections were deparaffinized and incubated sequentially with relevant
antibodies (Supplemental Table S1). Five unfed mice (overnight food withdrawal) were tested per peptide (n⫽5). Similar
results were obtained with fed mice (data not shown).

chromogranin A-positive cells (Fig. 2D) in the proximal pressed CD36 (Table 2 and Fig. 2E). Secretin-positive
small intestine. cells in these sections were also positive for chromo-
granin A (data not shown). EECs project their apical
CD36 is expressed on subpopulations of EECs membrane into the intestinal lumen, and all secretin-
positive EECs may express CD36 on their apical mem-
Our in vivo data showed that CD36 deletion reduced brane (Fig. 2B, C; arrows).
release of secretin and CCK. We examined whether Colocalization of CD36 and CCK was studied using trans-
CD36 expression can be detected on the subpopula- genic mice expressing the GFP driven by the CCK promoter.
tions of EECs involved in release of these peptides. The A small subset of CCK-secreting cells (⬃5%) exhibited
CD36 signal was detected in the cytoplasm of a subset of intracellular costaining for CD36 (Fig. 3A, B and Table 2).
secretin-producing cells in the proximal (Fig. 2B, C) Based on immunofluorescence staining, CCK-positive cells
and distal (data not shown) small intestines. Approxi- may have apical CD36 expression (Fig. 3C, D; arrows).
mately 21 and 9% of secretin-positive cells in the
proximal and distal small intestine, respectively, ex- CD36 enhances secretin and CCK release by STC-1 cells

To demonstrate direct CD36 involvement in FA-in-


TABLE 2. Distribution of CD36 across EECs positive for chromogranin duced peptide secretion and to better understand the
A, secretin, and CCK along the duodenal-ileum axis in SI
signaling mechanisms involved, studies were conducted
Doubly positive cells
in the EEC line STC-1 derived from an endocrine
tumor of the mouse small intestine. The STC-1 cell line
Chromogranin A Secretin and is a mix of EEC subpopulations (42) that secrete a
Location and CD36 CD36 CCK and CD36
myriad of gut hormones including secretin and CCK
Proximal SI 28/80 (35.0%) 8/39 (20.5%) 5/100 (5%) (43) and has been used extensively to investigate the
Distal SI 4/25 (16.0%) 1/11 (9.1%) mechanisms underlying peptide release. No expression
of CD36 in STC-1 cells was detected, and cells stably
Sections from 5 individual mice were used for determination of expressing WT human CD36 and a control line express-
colocalization of CD36 with chromogranin A and secretin; sections ing empty vector were generated (Fig. 4A). A cell line
from 3 mice were used for CCK. Colocalization, determined by
doubly positive cells, is expressed as a percentage (n⫽5). SI, small stably expressing a mutated form of CD36 (CD36K/A)
intestine. that lacks CD36-mediated signaling to intracellular

CD36 MEDIATES RELEASE OF GUT PEPTIDES 1195


Figure 3. CD36 expression on CCK-positive cells
in proximal intestines of CCK-GFP transgenic
mice. A, B) Subset of CCK-positive cells (green)
express cytoplasmic CD36 (red). C, D) CD36 may
also be present on apical membranes (arrows) of
CCK-positive cells. Frozen OCT-embedded sec-
tions were fixed in 10% formalin, blocked, and
incubated with CD36 and GFP antibodies. Immu-
noreactivity was visualized as in the legend to Fig.
2 (⫻400). Three mice [fed (A, B) or unfed
overnight (C, D)] were tested for CCK (n⫽3).

calcium was also used. The C terminus of CD36 is DHA (Fig. 5A) and LA (Fig. 5B) induced almost 3-fold
important for signal transduction (44, 45). In the enhancement of CCK release, whereas PA (Fig. 5D)
CD36K/A mutant lysines K469 and K472 in the C increased it by approximately 2-fold. OA was still inef-
terminus were substituted with alanine. As we reported fective. No enhancement was observed in cells express-
previously, the CD36K/A mutant has lost the ability to ing vector or CD36K/A. The specificity of CD36 in-
regulate calcium influx, calcium-induced phospho- volvement was validated further by SSO producing
lipase activation, and translocation to membranes (20), almost complete inhibition of CCK release (Fig. 5E).
but it has normal FA uptake activity (38). We hypothe-
sized that this mutant might have impaired regulation Enterocyte coculture on secretin and CCK release by
of granule exocytosis as a result of its defect in calcium STC-1 cells
signaling. All cells were treated with 50, 100, or 200 ␮M
FA (bound to 5 ␮M BSA), and the effect on peptide Our experiments using intestinal segments in vitro
secretion was monitored. suggested that CD36 regulation of EEC release of
Secretin release was induced 2-fold above basal by 50 secretin and CCK was independent of changes in fat
␮M DHA, in CD36-expressing STC-1 cells, and no absorption (Fig. 1). To examine this result further we
enhancement was observed in cells expressing either tested whether CD36 expression on enterocytes influ-
CD36K/A or the empty vector (Fig. 4B). Similar data ences peptide secretion by neighboring EECs. Studies
were observed with LA (Fig. 4C) and linolenic acid were conducted using coculture of STC-1 cells with
(data not shown). OA and PA were ineffective (Fig. 4D, Caco-2 cells, a well-studied model of enterocytes (47–
E). As the FA concentration was increased to 100 ␮M, 49). Differentiated Caco-2 cells with stable expression
secretin release increased to 3.5 times basal with DHA of CD36, which are responsive to FA signaling (data not
(Fig. 4B) and to 3 and 2 times basal with LA (Fig. 4C) shown), were generated and used for these experi-
and OA (Fig. 4D), respectively. However, modest en- ments. Cocultures of Caco-2 and STC-1 cells, with (⫹)
hancement of release (1.5–1.8 times basal) could now or without (⫺) stable expression of CD36 were tested
be observed in control (vector and CD36K/A) cells. At for LA-induced release of secretin and CCK. At 50 ␮M,
200 ␮M FA, the stimulatory effects of DHA, LA, and OA LA enhanced secretin release in STC-1 cells stably
on secretin release were slightly more than those at 100 expressing CD36 (⫹), compared with that in empty
␮M, and PA had a small effect. To further validate the vector controls (⫺) by⬃2-fold above basal whether the
CD36 specificity of observed effects, the irreversible coculture contained Caco-2 cells expressing CD36 (⫹)
CD36 inhibitor sulfo-N-succinimidyl oleate (SSO; ref. or empty vector (⫺) (Fig. 4G). A similar trend was
46) was used. In cells preincubated with SSO (20 ␮M, observed with 200 ␮M LA. Secretin release increased
15 min) and then treated with 100 ␮M FA, secretin similarly and ⬃4-fold over basal in cocultures contain-
release was blunted (Fig. 4F), whereas no significant ing STC-1 (⫹) cells whether the cocultured Caco-2 cells
effect was observed on CD36-independent release (vec- did or did not express CD36. In contrast with 50 ␮M
tor and CD36K/A-expressing cells). LA, 200 ␮M LA enhanced secretion ⬃2-fold in STC-1
cells not expressing CD36 (Fig. 4G). For CCK release,
CCK the trends were similar to those observed with secretin
(Fig. 5F). LA at 50 and 200 ␮M stimulated CCK release
DHA at 50 ␮M stimulated CCK release by 2-fold from by 2- to 2.2- and 2.7- to 2.9-fold, respectively, in STC-1
cells expressing CD36 with no effect on empty vector or cells stably expressing CD36 (⫹) compared with that in
CD36K/A-expressing cells (Fig. 5A). Similar effects vector controls (⫺), independent of CD36 expression
were observed with LA (Fig. 5B). PA induced a modest in Caco-2 cells. These data suggested limited contribu-
30% increase (Fig. 5D), whereas OA was ineffective tion of CD36 expression on adjacent enterocytes to
(Fig. 5C). At higher concentrations of FA (200 ␮M), FA-stimulated peptide release.

1196 Vol. 27 March 2013 The FASEB Journal 䡠 www.fasebj.org SUNDARESAN ET AL.
Figure 4. CD36 enhances FA-induced secretin release from STC-1 cells. A) Western blot showing CD36 expression in STC-1 cells
stably expressing empty vector, human CD36 WT, or the CD36K/A mutant. Ran is the loading control. B⫺E) Secretin release.
STC-1 cells were incubated with 50, 100, and 200 ␮M FA (with 5 ␮M BSA) or BSA alone (control) in HBSS (no glucose) for 60
min at 37°C, and secretin release was measured after DHA (B), LA (C), OA (D), and PA (E). Release is expressed as fold increase
over BSA controls after normalization to cell protein. Data are means ⫾ se of triplicates from 4 experiments (n⫽12).
F) LA-induced secretin release in the presence of the CD36 inhibitor, SSO (20 ␮M, 15 min). Cells were preincubated with 20
␮M SSO for 15 min before stimulation with 100 ␮M LA. G) Coculture of Caco-2 and STC-1 cells, with or without stable expression
of CD36. STC-1 cells expressing CD36 (STC ⫹) or empty vector (STC ⫺) were seeded with Caco-2 cells with (Caco ⫹) or without
(Caco ⫺) CD36 expression. Cocultures were serum starved overnight and then were incubated in HBSS with 50 or 200 ␮M LA
(plus 5 ␮M BSA) or BSA alone (controls) for 60 min at 37°C. Secretin release was determined and is expressed as fold increase
over BSA alone after normalization to cell protein. Data are means ⫾ se of triplicates from 3 experiments (n⫽8 –12). Points with
different letter symbols are significantly different (P⬍0.05). **P ⬍ 0.01.

CD36 MEDIATES RELEASE OF GUT PEPTIDES 1197


Figure 5. CD36 mediates FA-induced CCK release. A⫺D) CCK release from STC-1 cells expressing empty vector, CD36 WT, or
the mutant CD36K/A in response to DHA (A), LA (B), OA (C), and PA (D). E) LA-induced CCK release after treatment with
SSO (20 ␮M, 15 min). Cells were preincubated with 20 ␮M SSO for 15 min before stimulation with 100 ␮M LA. STC-1 cells were
FA treated (as in the legend to Fig. 4), and CCK release was determined by RIA. Data are means ⫾ se of triplicates from 3
experiments (n⫽9). Data points with different letter symbols are significantly different (P⬍0.05). *P ⬍ 0.05; **P ⬍ 0.01.
F) Coculture of Caco-2 and STC-1 cells, with (⫹) or without (⫺) stable expression of CD36. Cocultures were serum starved for
8 h and then were tested for CCK release as in the legend to Fig. 4. CCK was determined by RIA and is expressed as fold over
cells with BSA alone after normalization to cell protein. Data are means ⫾ se of triplicates from 2 experiments (n⫽4 for HBSS
controls; n⫽6 for treatments). Points with different letter symbols are significantly different (P⬍0.05). **P ⬍ 0.01.

cAMP production is involved in CD36-dependent no significant effect on release of these peptides in the
peptide release CD36K/A mutant. These data implicated cAMP gener-
ation in CD36-mediated FA-induced release of secretin
cAMP regulates a variety of secretory events including and CCK.
EEC release of secretin and CCK (30, 50). Its role in
CD36-mediated effects was examined. LA treatment CD36-mediated secretin release is dependent on PKA
increased intracellular cAMP (⬃50%) compared with activation
that in BSA controls, and this effect was strongly
amplified in CD36-expressing cells in which cAMP A major effector of cAMP is PKA, which, when acti-
increased by 240% (Fig. 6A). DHA increased cAMP vated, phosphorylates an array of protein substrates
(⬃88%) only in these cells, whereas PA had no effect (51). We determined the effect of FA on PKA activity by
(Fig. 6A). Further, the cAMP inhibitor MDL12230A (10 measuring phosphorylation of its substrates (RRXS*/T*
␮M, 30 min) reduced CD36-dependent release of both motifs). Addition of LA (Fig. 7A) or DHA (data not
secretin (Fig. 6B) and CCK (Fig. 6C). MDL12230A had shown) in the presence of 0.5 mM IBMX (to block

Figure 6. CD36-mediated enhancement of peptide release involves cAMP generation. A) Intracellular cAMP levels in response
to LA, DHA, and PA in STC-1 cells with vector, CD36 WT, and CD36K/A. STC-1 cells were FA treated at the indicated
concentration for 30 min (in the presence of the phosphodiesterase inhibitor IBMX, 0.5 mM), and cAMP was measured in cell
lysates. B, C) Secretin (B) and CCK (C) release after treatment with the cAMP inhibitor MDL12230A (10 ␮M, 30 min) in the
presence of 100 ␮M LA. Data are means ⫾ se of triplicates from 3 experiments (n⫽9). **P ⬍ 0.01; ***P ⬍ 0.001.

1198 Vol. 27 March 2013 The FASEB Journal 䡠 www.fasebj.org SUNDARESAN ET AL.
Figure 7. CD36-mediated effects on peptide release involve activation of PKA and CaM-KII. A, B) Phospho-PKA substrates (with
RRXS*/T* motif) in cell lysates (A) and PKA C-␣ in nuclear fractions (B) of STC-1 cells expressing vector, CD36 WT, or
CD36K/A after LA stimulation (in the presence of 0.5 mM IBMX). Histone H-3 was used as a loading control for nuclear lysates
(n⫽4). C, D) LA-induced secretin (C) and CCK release (D) after treatment with the PKA inhibitor H-89. E) Representative
intracellular calcium transients in STC-1 cells stably expressing vector, CD36 WT, or CD36K/A. F) LA (100 ␮M) induced CCK
release in CD36-expressing STC-1 cells with or without treatment with the CaM-KII inhibitor KN-62 (2.5 ␮M, 15 min). Data are
means ⫾ se of triplicates from 3 experiments (n⫽9). *P ⬍ 0.05; **P ⬍ 0.01.

cAMP degradation) strongly enhanced phosphoryla- in Chinese hamster ovary, macrophages, and taste bud
tion of PKA substrates in STC-1 cells expressing WT cells (20, 22). We investigated effect of CD36 expres-
CD36 but not in cells expressing the signaling impaired sion on calcium transients in response to LA in the
mutant CD36K/A (Fig. 7A). Under the same condi- absence or presence of extracellular calcium. A rapid
tions, PA treatment was ineffective (Supplemental Fig. increase in intracellular calcium was observed in LA-
S3). Activation of the PKA tetramer associates with treated STC-1 cells expressing CD36 WT on addition of
translocation of the catalytic subunit to the nucleus medium calcium, whereas no effect was observed in
(51). LA treatment induced nuclear translocation of cells expressing the vector or CD36K/A mutant (Fig.
catalytic PKA C-␣ in control cells, and this effect was 7E). Calcium influx activates CaM-KII, which is involved
amplified 3-fold in cells expressing CD36 WT (Fig. 7B). in release of CCK (32). Indeed, treatment with the
LA increased nuclear protein kinase C content by CaM-KII inhibitor KN-62 (2.5 ␮M, 15 min) blunted
⬃2.4-fold in vector cells, by 7.4-fold in CD36WT cells, CCK release in CD36-expressing cells (Fig. 7F), sup-
and by 1.7-fold in CD36K/A cells. The responses of porting involvement of CaM-KII in CD36-mediated
vector and CD36K/A-expressing cells were similar and CCK release.
significantly less than that of CD36WT cells (P⬍0.01).
Pretreatment with the PKA inhibitor H-89 (10 ␮M, 30
min) reduced secretin release only in CD36 WT-ex- DISCUSSION
pressing cells (Fig. 7C), suggesting that release involved
PKA activation. In contrast, CD36-mediated, FA-in- CD36 contributes to different steps of the fat absorp-
duced CCK release was not affected by H-89 (Fig. 7D), tion process (53); it mediates fat perception in taste
implicating PKA-independent mechanisms downstream of bud cells and the initiation of the cephalic phase of
cAMP. digestion (22). It also influences intestinal fat process-
ing and chylomicron formation (25, 26, 28). This study
CD36-mediated CCK release involves CaM-KII documented a novel and potentially important meta-
bolic function of CD36: its mediation of gut peptide
An increase in intracellular calcium after cAMP gener- release in response to dietary FA. We showed that CD36
ation has been implicated in release of CCK (50, 52). is expressed on EECs positive for secretin and CCK and
CD36 was recently reported to influence calcium flux the release of these peptides in response to intragastric

CD36 MEDIATES RELEASE OF GUT PEPTIDES 1199


fat was halved in CD36⫺/⫺ mice. This reduction was CD36-mediated FA signaling enhances cAMP
most likely independent of changes in fat absorption generation and calcium transients
because it was reproduced in isolated intestinal seg-
ments. Using the EEC line STC-1 with stable expres- Endocrine cells release granules in response to factors
sion of CD36 or a signaling-impaired CD36 mutant, that increase intracellular Ca2⫹ or cAMP levels. The
specific CD36 mediation of FA-induced peptide re- cAMP pathway modulates granule exocytosis in a vari-
lease was demonstrated to involve increases in cellu- ety of cells (51) and has been implicated in release of
lar cAMP and calcium. gut peptides (30, 50). We showed that CD36 signaling
regulates cAMP levels and that this was involved in its
enhancement of FA-induced release of secretin and
CD36 regulates peptide release CCK. FA activation of PKA, the major cAMP effector,
and enhanced nuclear PKA content were documented
CD36 deletion was associated with 50 – 60% lower in CD36-expressing cells. However, only release of
plasma levels of secretin and CCK at 30 min after an secretin was markedly attenuated by the PKA inhibitor
intragastric triglyceride load. In vitro, the effect of FA to H-89, whereas that of CCK was not, suggesting PKA-
induce release of these peptides was specific to cells independent mechanisms. CCK release is triggered by
expressing functional WT CD36. The calcium signal- an increase in intracellular Ca2⫹ that is coupled to
ing-impaired CD36K/A mutant was ineffective despite cAMP generation (50) and CaM-KII activation (32).
the fact that it expresses normally, localizes to the Consistent with CD36 regulation of this pathway, FA
plasma membrane, and functions in FA uptake (20, induced an increase in intracellular Ca2⫹ specifically in
38). Our findings using WT and CD36⫺/⫺ mice, given STC-1 cells expressing CD36, and the CCK response
a high oil load, support the existence of both CD36- was suppressed by the CaM-KII inhibitor KN-62. The
dependent and CD36-independent peptide release be- differential effects of signaling inhibitors, implying
cause secretin and CCK release was suppressed by 60 involvement of distinct pathways downstream of cAMP
and 50%, respectively. The work in vitro with proximal in the regulation of secretin vs. CCK release, probably
intestinal segments and with STC-1 cells showed that reflect the contribution of different cell subpopulations
the CD36-independent component becomes more ap- because the STC-1 cell line derived from an endocrine
parent as the FA concentration is increased and the tumor of the intestine is heterogeneous (42, 43). Our
high-affinity CD36-mediated pathway is saturated. In data with STC-1 cells are consistent with previous work
vivo, this could imply that one function of CD36 is to supporting involvement of Ca2⫹ and cAMP in release of
enable secretin and CCK release at relatively low FA secretory granules in most cell types and with selectivity
concentrations possibly early during the digestion pro- of granule exocytosis, reflecting variable sensitivity to
cess, thus optimizing subsequent absorption. Other FA Ca2⫹ and selective downstream phosphorylation path-
receptors such as GPR120 (17) and GPR40 (2), which ways (55). Some EECs in the mouse intestine synthesize
are localized more distally in the intestine, would and secrete more than one peptide, including CCK and
contribute to FA-induced FA release at a later stage in secretin (56). There is also evidence for different
the absorptive process. GPR120 has been mainly impli- composition of granules in CCK- or secretin-releasing
EECs (57), consistent with differential regulation of
cated in mediation of FA-induced incretin release (17,
granule release. The possibility that some secretion of
18). Its role in CCK release has been briefly docu-
CCK and secretin may occur from the same STC-1
mented in STC-1 cells, in which small interfering RNA
subpopulation cannot be ruled out.
directed against GPR120 but not GPR40 inhibited a
In summary, our data document CD36 expression on
2-fold effect of linolenic acid to induce CCK secretion
secretin- and CCK-releasing EECs and its role in FA-
by 30 – 40% (54). More convincing evidence docu- induced release of these peptides in vivo. The more
mented involvement of GPR40 in long-chain FA-in- abundant localization of CD36 in the proximal than in
duced release of CCK (2) in vivo using GPR40⫺/⫺ mice. the distal intestine and the relatively higher FA affinity
A load of intragastric olive oil comparable to that used of CD36-mediated vs. CD36-independent peptide re-
in our study resulted at 30 min in a 4.2-fold increase of lease suggest that CD36 would play a critical role in the
plasma CCK vs. a 2.8-fold increase in GPR40⫺/⫺ mice, early phase of fat absorption. Other FA receptors such
meaning a reduction of 33%. These findings together as GPR40, which is more abundant in the distal intes-
with our observation of a 50% reduction of plasma tine, would contribute at later phases of absorption.
CCK, 30 min after oil gavage in CD36⫺/⫺ mice (Fig. 1) Our data, together with previous findings, suggest that
support the interpretation that CD36 and GPR40 might CD36 and GPR40 account for most FA-induced CCK
account for most FA-induced CCK release in vivo. In release and that CD36 is the predominant regulator of
the case of secretin, our study provides, to our knowl- FA-induced secretin release. CCK and secretin play
edge, the first information related to the FA receptor important regulatory roles in fat absorption because
involved in secretin release by documenting a major they influence gastric emptying and acid secretion and
role of CD36 based on the 60% reduction of plasma pancreatic and intestinal bicarbonate secretion in ad-
secretin release in CD36⫺/⫺ mice, given a high intra- dition to gallbladder contractility and intestinal motility
gastric oil load (Fig. 1). (3, 58). Both peptides influence satiety and energy

1200 Vol. 27 March 2013 The FASEB Journal 䡠 www.fasebj.org SUNDARESAN ET AL.
homeostasis (12, 13). Regulation of FA-induced CCK mice through the activation of the melanocortin system. Neuro-
and secretin release contributes to the role of CD36 in psychopharmacology 36, 459 –471
13. Dockray, G. J. (2012) Cholecystokinin. Curr. Opin. Endocrinol.
intestinal handling of dietary fat. In humans, common Diabetes Obes. 19, 8 –12
SNPs in the CD36 gene influence plasma lipids (59), 14. Hajri, T., and Abumrad, N. A. (2002) Fatty acid transport across
and CD36 deficiency is characterized by altered chylo- membranes: relevance to nutrition and metabolic pathology.
Annu. Rev. Nutr. 22, 383–415
micron production (60). In addition, SNPs that reduce 15. Oh, D. Y., and Lagakos, W. S. (2011) The role of G-protein-
the CD36 level associate with diminished oral FA sen- coupled receptors in mediating the effect of fatty acids on
sitivity (23). Whether intestinal FA sensing is also inflammation and insulin sensitivity. Curr. Opin. Clin. Nutr.
altered in these subjects remains to be determined. FA Metab. Care 14, 322–327
16. Vinolo, M. A., Hirabara, S. M., and Curi, R. (2012) G-protein-
sensing in the small intestine with consequent effects coupled receptors as fat sensors. Curr. Opin. Clin. Nutr. Metab.
on gut peptide release and on satiety were reported to Care 15, 112–116
be attenuated in subjects with reduced oral FA sensitiv- 17. Hirasawa, A., Tsumaya, K., Awaji, T., Katsuma, S., Adachi, T.,
Yamada, M., Sugimoto, Y., Miyazaki, S., and Tsujimoto, G.
ity or after high-fat feeding (8). (2005) Free fatty acids regulate gut incretin glucagon-like
peptide-1 secretion through GPR120. Nat. Med. 11, 90 –94
The authors acknowledge valuable assistance by Terri 18. Oh, D. Y., Talukdar, S., Bae, E. J., Imamura, T., Morinaga, H.,
Pietka (Adipocyte Biology and Molecular Nutrition Core, Fan, W., Li, P., Lu, W. J., Watkins, S. M., and Olefsky, J. M.
Nutrition and Obesity Research Center, Washington Univer- (2010) GPR120 is an omega-3 fatty acid receptor mediating
sity School of Medicine; P30-DK056341) and are grateful to potent anti-inflammatory and insulin-sensitizing effects. Cell
Dr. Ondrej Kuda for help with the calcium flux studies and to 142, 687–698
Dr. Doug Hanahan for the permission to obtain STC-1 cells 19. El-Yassimi, A., Hichami, A., Besnard, P., and Khan, N. A. (2008)
Linoleic acid induces calcium signaling, Src kinase phosphory-
from American Type Culture Collection (Manassas, VA, lation, and neurotransmitter release in mouse CD36-positive
USA). This work was supported by funding from U.S. Na- gustatory cells. J. Biol. Chem. 283, 12949 –12959
tional Institutes of Health grants DK033301, DK60022 (to 20. Kuda, O., Jenkins, C. M., Skinner, J. R., Moon, S. H., Su, X.,
N.A.A.), DK33165, DK55753 (to W.F.S.), and DK091946 (to Gross, R. W., and Abumrad, N. A. (2011) CD36 protein is
R.A.L.). R.S., T.E.R., R.C., and F.N. assisted in data acquisition involved in store-operated calcium flux, phospholipase A2 acti-
and reviewed the manuscript; W.F.S. and R.A.L. critically vation, and production of prostaglandin E2. J. Biol. Chem. 286,
reviewed the manuscript; S.S. obtained and analyzed data; 17785–17795
and S.S. and N.A.A. were involved in development of the 21. Khan, N. A., and Besnard, P. (2009) Oro-sensory perception of
study concept and design, data analysis and interpretation, dietary lipids: new insights into the fat taste transduction.
Biochim. Biophys. Acta 1791, 149 –155
and manuscript preparation. 22. Laugerette, F., Passilly-Degrace, P., Patris, B., Niot, I., Febbraio,
M., Montmayeur, J. P., and Besnard, P. (2005) CD36 involve-
ment in orosensory detection of dietary lipids, spontaneous fat
preference, and digestive secretions. J. Clin. Invest. 115, 3177–
REFERENCES 3184
23. Pepino, M. Y., Love-Gregory, L., Klein, S., and Abumrad, N. A.
1. Beglinger, C., and Degen, L. (2004) Fat in the intestine as a (2012) The fatty acid translocase gene CD36 and lingual lipase
regulator of appetite—role of CCK. Physiol. Behav. 83, 617–621 influence oral sensitivity to fat in obese subjects. J. Lipid Res. 53,
2. Liou, A. P., Lu, X., Sei, Y., Zhao, X., Pechhold, S., Carrero, R. J., 561–566
Raybould, H. E., and Wank, S. (2011) The G-protein-coupled 24. Lobo, M. V., Huerta, L., Ruiz-Velasco, N., Teixeiro, E., de la
receptor GPR40 directly mediates long-chain fatty acid-induced Cueva, P., Celdran, A., Martin-Hidalgo, A., Vega, M. A., and
secretion of cholecystokinin. Gastroenterology 140, 903–912 Bragado, R. (2001) Localization of the lipid receptors CD36 and
3. Liddle, R. A. (2000) Regulation of cholecystokinin secretion in CLA-1/SR-BI in the human gastrointestinal tract: towards the
humans. J. Gastroenterol. 35, 181–187 identification of receptors mediating the intestinal absorption
4. Buchan, A. M., Polak, J. M., Solcia, E., Capella, C., Hudson, D., of dietary lipids. J. Histochem. Cytochem. 49, 1253–1260
and Pearse, A. G. (1978) Electron immunohistochemical evi- 25. Nassir, F., Wilson, B., Han, X., Gross, R. W., and Abumrad, N. A.
dence for the human intestinal I cell as the source of CCK. Gut (2007) CD36 is important for fatty acid and cholesterol uptake
19, 403–407 by the proximal but not distal intestine. J. Biol. Chem. 282,
5. Lam, I. P., Lee, L. T., Choi, H. S., and Chow, B. K. (2006) 19493–19501
Localization of small heterodimer partner (SHP) and secretin 26. Drover, V. A., Ajmal, M., Nassir, F., Davidson, N. O., Nauli,
in mouse duodenal cells. Ann. N. Y. Acad. Sci. 1070, 371–375 A. M., Sahoo, D., Tso, P., and Abumrad, N. A. (2005) CD36
6. Polak, J. M., Bloom, S., Coulling, I., and Pearse, A. G. (1971) deficiency impairs intestinal lipid secretion and clearance of
Immunofluorescent localization of secretin in the canine duo- chylomicrons from the blood. J. Clin. Invest. 115, 1290 –1297
denum. Gut 12, 605–610 27. Hsieh, J., Longuet, C., Maida, A., Bahrami, J., Xu, E., Baker,
7. Polak, J. M., Coulling, I., Bloom, S., and Pearse, A. G. (1971) C. L., Brubaker, P. L., Drucker, D. J., and Adeli, K. (2009)
Immunofluorescent localization of secretin and enteroglucagon Glucagon-like peptide-2 increases intestinal lipid absorption
in human intestinal mucosa. Scand. J. Gastroenterol. 6, 739 –744 and chylomicron production via CD36. Gastroenterology 137,
8. Little, T. J., and Feinle-Bisset, C. (2011) Effects of dietary fat on 997–1005, 1005.e1⫺1005.e4
appetite and energy intake in health and obesity— oral and 28. Nauli, A. M., Nassir, F., Zheng, S., Yang, Q., Lo, C. M.,
gastrointestinal sensory contributions. Physiol. Behav. 104, 613– Vonlehmden, S. B., Lee, D., Jandacek, R. J., Abumrad, N. A.,
620 and Tso, P. (2006) CD36 is important for chylomicron forma-
9. Sclafani, A., and Ackroff, K. (2012) Role of gut nutrient sensing tion and secretion and may mediate cholesterol uptake in the
in stimulating appetite and conditioning food preferences. Am. proximal intestine. Gastroenterology 131, 1197–1207
J. Physiol. Regul. Integr. Comp. Physiol. 302, R1119 –R1133 29. Edfalk, S., Steneberg, P., and Edlund, H. (2008) Gpr40 is
10. Berna, M. J., and Jensen, R. T. (2007) Role of CCK/gastrin expressed in enteroendocrine cells and mediates free fatty acid
receptors in gastrointestinal/metabolic diseases and results of stimulation of incretin secretion. Diabetes 57, 2280 –2287
human studies using gastrin/CCK receptor agonists/antago- 30. Chang, C. H., Chey, W. Y., Erway, B., Coy, D. H., and Chang,
nists in these diseases. Curr. Top. Med. Chem. 7, 1211–1231 T. M. (1998) Modulation of secretin release by neuropeptides in
11. Chey, W. Y., and Chang, T. M. (2003) Secretin, 100 years later. secretin-producing cells. Am. J. Physiol. 275, G192–G202
J. Gastroenterol. 38, 1025–1035 31. Parker, H. E., Reimann, F., and Gribble, F. M. (2010) Molecular
12. Cheng, C. Y., Chu, J. Y., and Chow, B. K. (2011) Central and mechanisms underlying nutrient-stimulated incretin secretion.
peripheral administration of secretin inhibits food intake in Expert Rev. Mol. Med. 12, e1

CD36 MEDIATES RELEASE OF GUT PEPTIDES 1201


32. Prpic, V., Basavappa, S., Liddle, R. A., and Mangel, A. W. (1994) dyl derivatives of long-chain fatty acids: inhibition of fatty acid
Regulation of cholecystokinin secretion by calcium-dependent transport. J. Membr. Biol. 121, 261–268
calmodulin kinase II: differential effects of phenylalanine and 47. Hussain, M. M. (2000) A proposed model for the assembly of
cAMP. Biochem. Biophys. Res. Commun. 201, 1483–1489 chylomicrons. Atherosclerosis 148, 1–15
33. Chandra, R., Samsa, L. A., Vigna, S. R., and Liddle, R. A. (2010) 48. Levy, E., and Bendayan, M. (2000) Use of immunoelectron
Pseudopod-like basal cell processes in intestinal cholecystokinin microscopy and intestinal models to explore the elaboration of
cells. Cell Tissue Res. 341, 289 –297 apolipoproteins required for intraenterocyte lipid transport.
34. Suzuki, S., Suzuki, H., Horiguchi, K., Tsugawa, H., Matsuzaki, J., Microsc. Res. Tech. 49, 374 –382
Takagi, T., Shimojima, N., and Hibi, T. (2010) Delayed gastric 49. Mathias, A., Duc, M., Favre, L., Benyacoub, J., Blum, S., and
emptying and disruption of the interstitial cells of Cajal network Corthesy, B. (2010) Potentiation of polarized intestinal Caco-2
after gastric ischaemia and reperfusion. Neurogastroenterol. Motil. cell responsiveness to probiotics complexed with secretory IgA.
22, 585–593, e126 J. Biol. Chem. 285, 33906 –33913
35. Liddle, R. A., Goldfine, I. D., and Williams, J. A. (1984) Bioassay 50. Scott, L., Prpic, V., Capel, W. D., Basavappa, S., Mangel, A. W.,
of plasma cholecystokinin in rats: effects of food, trypsin inhib- Gettys, T. W., and Liddle, R. A. (1996) ␤-Adrenergic regulation
itor, and alcohol. Gastroenterology 87, 542–549 of cholecystokinin secretion in STC-1 cells. Am. J. Physiol. 270,
36. Tashiro, M., Samuelson, L. C., Liddle, R. A., and Williams, J. A. G291–G297
(2004) Calcineurin mediates pancreatic growth in protease 51. Seino, S., and Shibasaki, T. (2005) PKA-dependent and PKA-
inhibitor-treated mice. Am. J. Physiol. Gastrointest. Liver Physiol. independent pathways for cAMP-regulated exocytosis. Physiol.
286, G784 –G790 Rev. 85, 1303–1342
37. Rehfeld, J. F. (1998) Accurate measurement of cholecystokinin 52. Sidhu, S. S., Thompson, D. G., Warhurst, G., Case, R. M., and
in plasma. Clin. Chem. 44, 991–1001 Benson, R. S. (2000) Fatty acid-induced cholecystokinin secre-
38. Smith, J., Su, X., El-Maghrabi, R., Stahl, P. D., and Abumrad, tion and changes in intracellular Ca2⫹ in two enteroendocrine
N. A. (2008) Opposite regulation of CD36 ubiquitination by cell lines, STC-1 and GLUTag. J. Physiol. 528 (Pt. 1), 165–176
fatty acids and insulin: effects on fatty acid uptake. J. Biol. Chem. 53. Abumrad, N. A., and Davidson, N. O. (2012) Role of the gut in
283, 13578 –13585
lipid homeostasis. Physiol. Rev. 92, 1061–1085
39. Poirier, H., Degrace, P., Niot, I., Bernard, A., and Besnard, P.
54. Tanaka, T., Katsuma, S., Adachi, T., Koshimizu, T. A., Hirasawa,
(1996) Localization and regulation of the putative membrane
A., and Tsujimoto, G. (2008) Free fatty acids induce cholecysto-
fatty-acid transporter (FAT) in the small intestine. Comparison
kinin secretion through GPR120. Naunyn-Schmiedebergs Arch.
with fatty acid-binding proteins (FABP). Eur. J. Biochem. 238,
Pharmacol. 377, 523–527
368 –373
55. Burgoyne, R. D., and Morgan, A. (2003) Secretory granule
40. Tran, T. T., Poirier, H., Clement, L., Nassir, F., Pelsers, M. M.,
Petit, V., Degrace, P., Monnot, M. C., Glatz, J. F., Abumrad, exocytosis. Physiol. Rev. 83, 581–632
N. A., Besnard, P., and Niot, I. (2011) Luminal lipid regulates 56. Habib, A. M., Richards, P., Cairns, L. S., Rogers, G. J., Bannon,
CD36 levels and downstream signaling to stimulate chylomicron C. A., Parker, H. E., Morley, T. C., Yeo, G. S., Reimann, F., and
synthesis. J. Biol. Chem. 286, 25201–25210 Gribble, F. M. (2012) Overlap of endocrine hormone expres-
41. Varndell, I. M., Lloyd, R. V., Wilson, B. S., and Polak, J. M. sion in the mouse intestine revealed by transcriptional profiling
(1985) Ultrastructural localization of chromogranin: a potential and flow cytometry. Endocrinology 153, 3054 –3065
marker for the electron microscopical recognition of endocrine 57. Portela-Gomes, G. M., Stridsberg, M., Johansson, H., and Grime-
cell secretory granules. Histochem. J. 17, 981–992 lius, L. (1997) Complex co-localization of chromogranins and
42. Cheung, A. T., Dayanandan, B., Lewis, J. T., Korbutt, G. S., neurohormones in the human gastrointestinal tract. J. His-
Rajotte, R. V., Bryer-Ash, M., Boylan, M. O., Wolfe, M. M., and tochem. Cytochem. 45, 815–822
Kieffer, T. J. (2000) Glucose-dependent insulin release from 58. Lam, I. P., Siu, F. K., Chu, J. Y., and Chow, B. K. (2008) Multiple
genetically engineered K cells. Science 290, 1959 –1962 actions of secretin in the human body. Int. Rev. Cytol. 265,
43. Rindi, G., Grant, S. G., Yiangou, Y., Ghatei, M. A., Bloom, S. R., 159 –190
Bautch, V. L., Solcia, E., and Polak, J. M. (1990) Development of 59. Love-Gregory, L., Sherva, R., Schappe, T., Qi, J. S., McCrea, J.,
neuroendocrine tumors in the gastrointestinal tract of trans- Klein, S., Connelly, M. A., and Abumrad, N. A. (2011) Common
genic mice. Heterogeneity of hormone expression. Am. J. Pathol. CD36 SNPs reduce protein expression and may contribute to a
136, 1349 –1363 protective atherogenic profile. Hum. Mol. Genet. 20, 193–201
44. Lipsky, R. H., Eckert, D. M., Tang, Y., and Ockenhouse, C. F. 60. Masuda, D., Hirano, K., Oku, H., Sandoval, J. C., Kawase, R.,
(1997) The carboxyl-terminal cytoplasmic domain of CD36 is Yuasa-Kawase, M., Yamashita, Y., Takada, M., Tsubakio-
required for oxidized low-density lipoprotein modulation of Yamamoto, K., Tochino, Y., Koseki, M., Matsuura, F., Nishida,
NF-␬B activity by tumor necrosis factor-␣. Recept. Signal Trans- M., Kawamoto, T., Ishigami, M., Hori, M., Shimomura, I., and
duct. 7, 1–11 Yamashita, S. (2009) Chylomicron remnants are increased in
45. Silverstein, R. L. (2009) Type 2 scavenger receptor CD36 in the postprandial state in CD36 deficiency. J. Lipid Res. 50,
platelet activation: the role of hyperlipemia and oxidative stress. 999 –1011
Clin. Lipidol. 4, 767
46. Harmon, C. M., Luce, P., Beth, A. H., and Abumrad, N. A. Received for publication July 30, 2012.
(1991) Labeling of adipocyte membranes by sulfo-N-succinimi- Accepted for publication November 26, 2012.

1202 Vol. 27 March 2013 The FASEB Journal 䡠 www.fasebj.org SUNDARESAN ET AL.
Copyright of FASEB Journal is the property of Federation of American Society for
Experimental Biology and its content may not be copied or emailed to multiple sites or posted
to a listserv without the copyright holder's express written permission. However, users may
print, download, or email articles for individual use.

You might also like