You are on page 1of 12

Available online at www.sciencedirect.

com

ScienceDirect
Journal of Nutritional Biochemistry 33 (2016) 91 102

Effect of dietary docosahexaenoic acid (DHA) in phospholipids or triglycerides on


brain DHA uptake and accretion

Alex P. Kitson a , Adam H. Metherel a , Chuck T. Chen a, 1 , Anthony F. Domenichiello a , Marc-Olivier Trpanier a ,
Alvin Berger b, c , Richard P. Bazinet a,
a
Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, M5S3E2, Canada
b
Arctic Nutrition AS, NO-6155, rsta, Norway
c
Department of Food Science & Nutrition, University of Minnesota, St. Paul, MN, 55108-1038, USA

Received 6 October 2015; received in revised form 27 January 2016; accepted 11 February 2016

Abstract

Tracer studies suggest that phospholipid DHA (PL-DHA) more effectively targets the brain than triglyceride DHA (TAG-DHA), although the mechanism and
whether this translates into higher brain DHA concentrations are not clear. Rats were gavaged with [U-3H]PL-DHA and [U-3H]TAG-DHA and blood sampled over
6 h prior to collection of brain regions and other tissues. In another experiment, rats were supplemented for 4 weeks with TAG-DHA (fish oil), PL-DHA (roe PL)
or a mixture of both for comparison to a low-omega-3 diet. Brain regions and other tissues were collected, and blood was sampled weekly. DHA accretion rates
were estimated using the balance method. [U-3H]PL-DHA rats had higher radioactivity in cerebellum, hippocampus and remainder of brain, with no differences
in other tissues despite higher serum lipid radioactivity in [U-3H]TAG-DHA rats. TAG-DHA, PL-DHA or a mixture were equally effective at increasing brain DHA.
There were no differences between DHA-supplemented groups in brain region, whole-body, or tissue DHA accretion rates except heart and serum TAG where the
PL-DHA/TAG-DHA blend was higher than TAG-DHA. Apparent DHA -oxidation was not different between DHA-supplemented groups. This indicates that more
labeled DHA enters the brain when consumed as PL; however, this may not translate into higher brain DHA concentrations.
2016 The Authors. Published by Elsevier Inc. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Keywords: Docosahexaenoic acid; Brain; Triacylglycerol; Phospholipid; Serum; Uptake

1. Introduction Alzheimer's disease [711] (reviewed in [12]), major depression


[13,14], bipolar disorder [15] and schizophrenia [16], although some
Docosahexaenoic acid (DHA, 22:6n-3) is highly concentrated in the studies show no differences [1,1719]. This has led the eld to
brain where it composes approximately 10% of all fatty acids [1]. DHA speculate that strategies to increase brain DHA concentration may be
regulates many neurological processes including neuronal survival [2], effective in treatment/prevention of neurodegenerative/neuropsychi-
synaptogenesis [3], neuronal plasticity [4] and neuroinammation [5]. atric disorders, assuming that low brain DHA is causally linked to the
Experimental depletion of brain DHA in animal studies is associated disease progression [2023].
with behavioral impairments [reviewed in [6]], and some neurode- The brain can synthesize DHA from -linolenic acid (ALA, 18:3n-3)
generative and neuropsychiatric disorders have been associated with [24,25]; however, the synthesis rate (1.5 nmol/g/d) [24] is much
low brain DHA content. For example, lower postmortem brain DHA in lower than the total rate of brain DHA accretion (190 nmol/g/d) [26] in
select brain regions and lipid classes has been found in patients with rats, indicating that uptake from serum DHA is likely the main source
of DHA for the brain [24]. The plasma lipid pool from which the brain
takes up DHA is not agreed upon; however, there is evidence that

This work was funded by Arctic Nutrition AS, NO-6155 rsta, Norway. plasma unesteried DHA is the major contributor [25,27,28], although
Funding source was involved in study design, but not in analysis, data other pools may contribute, such as lysophosphatidylcholine [29,30].
handling or manuscript preparation. Alex P. Kitson received a fellowship from
Dietary interventions that target serum lipid pools important for brain
the Canadian Institutes of Health Research. Richard P. Bazinet holds a Canada
DHA uptake may be effective in targeting brain DHA.
Research Chair in Brain Lipid Metabolism.
Corresponding author at: Department of Nutritional Sciences, University Dietary DHA is typically esteried to TAG, although supplements
of Toronto, Fitzgerald Building, 150 College St. Room 306, Toronto, Ontario, with a high proportion of DHA esteried to phospholipids (PL), such as
M5S 3E2, Canada. Tel.: + 1-416-946-8276. krill oils and sh roe phospholipid extracts, are increasingly common.
E-mail address: richard.bazinet@utoronto.ca (R.P. Bazinet). Several studies have attempted to compare the bioavailability of TAG-
1
Present Address: National Institute on Alcohol Abuse and Alcoholism, or PL-enriched sources of DHA based on responses of blood
National Institutes of Health, Bethesda, MD, 20,8929410, USA. biomarkers and have concluded either higher bioavailability of PL-

http://dx.doi.org/10.1016/j.jnutbio.2016.02.009
0955-2863/ 2016 The Authors. Published by Elsevier Inc. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
92 A.P. Kitson et al. / Journal of Nutritional Biochemistry 33 (2016) 91102

DHA [3133] or equal efcacy [3437], although this area is After a 24-h recovery from surgery, a 250-l baseline blood sample was taken
followed by ushing the line with 1% heparinized saline to ensure line patency
controversial [3841].
throughout the experiment. Rats were then gavaged with 500 l of one of the two
There is also some evidence that dietary PL-DHA may target the brain tracers (n=4 for TAG-DHA, n=5 for PC-DHA) dissolved in olive oil providing 454.5 and
more effectively than TAG-DHA. Liu et al. orally administered [U-13C]DHA 192.5 mmol of the [U-3H]TAG-DHA (500 Ci) and [U-3H]DHA-PC (308 Ci), respec-
esteried in the sn-2 position of TAG or PC to neonatal piglets and found tively. The rat was then tethered to a counterbalanced sampling system (Instech
approximately twofold greater labeled DHA in gray matter and Laboratories, Plymouth Meeting, PA, USA). Blood samples (250 l) were taken 30, 60,
90, 120, 150, 180, 210, 240, 270, 300, 330 and 360 min postgavage. In the event of
synaptosomes [42] of [U-13C]DHA-PC gavaged piglets. Similarly, Graf difculty obtaining blood draw from the cannula, a tail-vein blood draw was performed.
et al. orally administered [1-14C]DHA esteried to sn-2 of PC and TAG and Blood was centrifuged at 10,000 g for isolation of serum and immediately frozen at
found higher radioactivity in brain regions of 10-week-old rats gavaged 80 C until analysis. Rats were euthanized by lethal injection of T-61 (embutramide
with [1-14C]DHA-PC, but not in 2- or 4-week-old rats [43]. 200 mg/ml, mebezodium iodide 50 mg/ml, tetrocaine hydrochloride 5 mg/ml admin-
istered at 300-l/kg body weight, Merck Animal Health, Kirkland, QUE, Canada) via the
Taken together, it appears that dietary PL-DHA may more
carotid catheter 360 min postgavage, and brain regions (cortex, cerebellum, hippo-
effectively target the brain relative to TAG-DHA; however, it is not campus, striatum, brainstem and rest of brain) as well as liver, perirenal adipose tissue,
known if this translates into higher brain DHA concentration following heart, small intestine (5 cm inferior to pyloric sphincter) and abdominal skin were
supplementation with a source of PL-DHA compared with TAG-DHA. It collected, rinsed in saline and frozen at 80 C until analysis.
is possible that ingested PL-DHA may more effectively enrich serum
2.3. Radiotracer gavage study lipid analysis
lipid pools that target brain DHA compared with TAG-DHA, as there is
some evidence that dietary PL and TAG target different serum lipid Lipids were extracted from serum and tissues by the method of Folch, Lees and
pools upon digestion and absorption [44,45]. However, a detailed Sloane Stanley [47] using 2:1:0.8 chloroform:methanol:0.88% KCl (v/v/v) as previously
time-course comparing the serum lipid class enrichment following described [48,49]. Thin-layer chromatography was used to isolate brain region PL as
well as to collect lipid classes in serum and liver (PL, monoacylglycerols, co-eluting
ingestion of DHA-containing PL and TAG tracers has not been
diacylglycerols/cholesterol, nonesteried fatty acids, TAGs, cholesteryl esters) using
performed, nor has it been assessed whether acute postprandial silica G plates (silica gel 60, EMD Millipore, MA, USA) and a mobile phase of 60:40:2
serum lipid class enrichment kinetics translate into differences in heptane:diethyl ether:glacial acetic acid (v/v/v). Lipid classes were visualized by
serum lipid class DHA concentration in response to semichronic PL- or spraying resolved plates with 0.1% (w/v) 8-anilino-1-naphthalenesulfonic acid and
TAG-DHA supplementation. identied in relation to mobility of a reference standard mixture containing lipid classes
of interest. Only total lipid extracts of heart, adipose, skin and intestine were analyzed.
Therefore, the goal of this study was to examine whether the
Radioactivity of total lipid extracts and lipid fractions was measured on a Hewlett-
higher brain tracer uptake of ingested PL-DHA compared to TAG-DHA Packard Tri-Carb 2900TR Liquid Scintillation Analyzer with detector efciency of
translated into higher brain DHA content following 4 weeks of 45.81%. Radioactive data were normalized to percentage of gavaged dose. Unidirec-
supplementation with a source of PL-DHA, TAG-DHA or a mixture of tional incorporation coefcients were also determined [50] using the average area
under the curve for radioactivity in unesteried fatty acids, esteried fatty acids and
the two. In addition, differences in serum lipid pool enrichment
total serum lipid radioactivity according to the following equation
following ingestion of PL-DHA and TAG-DHA tracer were character-
ized and compared to changes in serum lipid pool DHA concentration
C i T
following dietary PL-DHA vs. TAG-DHA supplementation. Differences k i 1
0 C si dt
T

in other tissues were also assessed.


Where C i(T) is the radioactivity of brain region i at the end of the experiment, and
2. Material and methods T0C sdt is the area under the curve of the serum radioactivity of serum lipid i. k* was
calculated for unesteried fatty acids, esteried lipids and total lipid radioactivity.
2.1. Radiotracer gavage study tracers
2.4. PL vs. TAG-DHA supplementation study animals and diets
Sn-1,3-dpalmitoyl-2-docosahexaenoylglycerol (TAG-DHA) and 1-palmitoyl-2-doc-
osahexaenoyl-sn-glycero-3-phosphocholine (DHA-PC) were purchased from Avanti Ten-day-old male LongEvans rat pups with lactating dams arrived at the
Polar Lipids (Alabaster, AL, USA) and were uniformly labeled with 3H by Moravek University of Toronto animal care facility, and the dams were immediately placed on
Biochemicals (Brea, CA, USA) to form [U-3H]TAG-DHA [U-3H]DHA-PC, respectively. a low n-3 PUFA diet (same as used in radiotracer study), and pups were weaned onto the
Radiochemical purity of the tracers was N99.8% as determined by Moravek Biochemicals low n-3 PUFA diet at 21 days of age. At 11 weeks of age, one group of rats was sacriced
and the specic activity of tracers was 1.1 mCi/mmol for [U-3H]TAG-DHA and 1.6 mCi/ by high-energy head-focused microwave xation (13.5 kW for 1.6 s; Cober Electronics
mmol for [U-3H]DHA-PC. Tracers were shipped and dissolved in toluene and, prior to Inc., Stratford, CT, USA [48,49,51]) following collection of tail vein blood, and brain
use, were dissolved in olive oil (Sigma-Aldrich, St. Louis, MO, USA) following regions (as in radiotracer study), liver, heart, adipose, small intestine, skin and perirenal
evaporation of toluene under a stream of N2. The tracer was dissolved to a nal adipose were collected to estimate baseline DHA concentrations (n=8). The rest of the
concentration of 500 Ci/ml for [U-3H]TAG-DHA and 300 Ci of [U-3H]DHA-PC. rats were randomized to one of four experimental diets: (a) a low n-3 PUFA diet
Different tracer concentrations were due to availability of material. supplemented with olive oil (OO diet; n=8); (b) a diet supplemented with sh oil (FO
diet; n=8); (c) a diet supplemented with herring caviar phospholipid concentrate (PLC
2.2. Radiotracer gavage study animals and diets diet; n=8) with 81% of DHA esteried to phospholipids; or (d) a diet supplemented
with a blend of FO and PLC (FO+PLC diet; n=7) with 30% of the DHA esteried to
All procedures performed on animals were approved by the University of Toronto phospholipids (lipid class composition analysis performed by Mylneeld, Dundee,
Animal Care Committee in accordance with guidelines set by the Canadian Council of Scotland). Oil sources and fatty acid concentration of diets are shown in Table 1. DHA-
Animal Care. Animals were housed with ad libitum access to food and water with a containing oils were prepared by Arctic Nutrition (fatty acid composition in
12:12-h light:dark cycle and controlled temperature of 221 C. Supplemental Table 1). All DHA-supplemented diets (FO, PLC, FO+PLC) were
Male LongEvans rats arrived at the University of Toronto animal care facility at formulated such that DHA would comprise 2% of total fatty acid mass, as this has
10 days of age with lactating dams. Dams were immediately placed on a low omega-3 been suggested to be the dietary DHA content at which brain DHA is maximal [52]. Rats
diet with 0.140.01% of fatty acids as n-3 PUFA (Dyet's Inc., Bethlehem Pennsylvania) were maintained on experimental diets for 4 weeks (from postnatal week 11 to week
modied from the AIN-93G formulation [46] as shown in Table 1. ALA content of the diet 15) with body weight, 24-h food intake and serum DHA (sampled via tail vein)
was consistent with previous work [46]. Pups were weaned onto the same diet at measured weekly. At the conclusion of supplementation, blood was sampled from the
21 days of age. Pups were maintained on this diet until 11 weeks of age when they tail vein, and rats were killed by high-energy head-focused microwave xation, and
underwent carotid artery cannulation. Briey, rats were anesthetized through samples were collected identically to baseline rats. Carcasses and 24-h fecal samples
isourane inhalation (5% induction, 12.5% maintenance) followed by subcutaneous were also collected and stored at 80 C.
injection of ketoprofen (5 mg/kg; Merial Canada Inc., Baie d'Urf, QC, Canada), and
incision sites (scapular, superior thoracic) were shaved and disinfected. Carotid artery 2.5. PL-DHA vs. TAG-DHA supplementation study lipid analysis
was isolated by blunt dissection and cannulated with sterile polyethylene catheter (PE
50, Intramedic; Becton Dickinson, Franklin Lakes, NJ, USA) tipped with 1.5 cm of Lipids were extracted from serum, tissues and feces as described for the radiotracer
silicone tubing (silicone tubing 0.020 in. inner diameter and 0.037 in. outer diameter, study. Carcasses were homogenized in a Waring industrial blender (model CB15) with a
VWR; Mississauga, ON, Canada). The cannula was externalized in the scapular region 4-L stainless steel pitcher (Hendrix Restaurant Equipment and Supplies, Brockville, ON,
and a skin button (Braintree Scientic, Braintree, MA, USA) was sutured to facilitate USA) with water to facilitate homogenization, and lipids were extracted in duplicate.
assembly of the sampling system. For all samples, an internal standard was added prior to lipid extraction for
A.P. Kitson et al. / Journal of Nutritional Biochemistry 33 (2016) 91102 93

Table 1
Fat source and fatty acid composition of rodent diets
Name Low n-3 PUFA Olive oil Fish oil Phospholipid concentrate Fish oil + phospholipid concentrate

Fat source g/kg diet


Hydrogenated coconut oil 66.2 50.0 50.0 50.0 50.0
Safower oil 33.8 33.8 33.8 33.8 33.8
Olive oil 0 16.2 8.8 3.4 7.85
Fish oil 0 0 7.4 0 0
Phospholipid concentrate 0 0 0 12.8 0
Phospholipid concentrate + sh oil 0 0 0 0 8.35
Total 100 100 100 100 100

Fatty acid % composition 1


C 10:0 4.440.54 2.360.45 2.320.25 2.380.08 2.350.11
C 12:0 37.450.98 25.940.51 26.50.45 27.570.11 27.140.2
C 14:0 14.210.47 10.270.12 10.980.12 11.470.08 11.120.12
C 16:0 8.900.23 9.960.19 9.620.06 10.580.14 9.850.04
C 18:0 7.380.18 6.590.17 6.770.12 7.080.1 6.820.06
C 20:0 0.170.01 0.220.005 0.230.01 0.20.01 0.220.001
SFA 75.110.56 55.781.07 56.640.69 59.530.23 57.690.45
C 16:1 0.060.01 0.350.01 0.470.004 0.490.001 0.40.003
C 18:1n-7 0.190.01 0.680.03 0.580.02 0.610.01 0.570.01
C 18:1n-9 5.250.15 16.550.39 12.250.21 9.640.07 11.720.15
C 20:1n-9 0.060.0001 0.10.01 0.340.01 0.190.004 0.150.01
C 22:1n-9 0.100.01 0.090.001 0.10.005 0.090.01 0.090.004
C 24:1n-9 TR 0.030.005 0.130.005 0.090.01 0.110.003
MUFA 5.70.17 17.790.41 13.870.25 11.140.07 13.060.14
C 18:2n-6 18.90.36 26.090.67 25.870.4 25.880.2 25.760.32
C 20:4n-6 TR TR TR TR TR
C 22:4n-6 0.10.01 0.050.013 0.080.01 0.040.01 0.060.01
N-6 19.050.38 26.180.66 26.080.39 26.020.2 25.940.33
C 18:3n-3 0.080.003 0.190.01 0.180.003 0.190.002 0.170.004
C 20:5n-3 TR TR 0.750.014 0.910.02 0.710.012
C 22:5n-3 TR TR 0.230.004 0.070.001 0.170.003
C 22:6n-3 0.050.002 0.050.002 2.240.04 2.120.04 2.250.04
N-3 0.140.01 0.250.01 3.410.05 3.310.04 3.310.06
PUFA 19.190.39 26.430.66 29.490.44 29.330.2 29.250.32
N-6/N-3 135.13.35 104.310.61 7.650.05 7.860.12 7.830.19

TR: trace (b0.05% of fatty acids), SFA: saturated fatty acids, MUFA: monounsaturated fatty acids, PUFA: polyunsaturated fatty acids.
1
Data are mean SD from triplicate analysis in our laboratory. Levels of 14:1, 18:3n-6, 20:2n-6, 20:4n-6, 22:4n-6, 22:5n-6 were measured and found to be negligible (b0.05% of
fatty acids).

phospholipids (1,2-diheptadecanoyl-sn-3-phosphatidylcholine; Avanti polar lipids), Whole-body apparent DHA -oxidation rates were determined using the following calculation:
TAGs (triheptadecanoate, Nuchek Prep) unesteried fatty acids (heptadecanoic acid,
Nuchek Prep) or cholesteryl esters (cholesteryl heptadecanoate, Nuchek Prep). Neutral Apparent daily  oxidation rate
lipid classes were isolated from serum lipid extracts by TLC as described for the Dietary DHA consumed  whole body DHA accretion  fecal DHA excretion
radiotracer study. Lysophosphatidylcholine was isolated by TLC on silica H plates 3
number of days of supplementation
(Analtech, Newark, DE, USA) with a mobile phase of chloroform:methanol:acetic
acid:acetone: water (40:25:7:4:2 by vol) [53]. Fatty acid methyl esters (FAMEs) were
prepared by heating at 100 C for 1 h in 14% boron triouride in methanol [54]. 2.8. Statistics

2.6. PL-DHA vs. TAG-DHA supplementation study gas chromatography Statistical analysis was performed using SPSS Statistics Version 22 and Prism Version
4. Signicance was inferred at Pb.05. Comparison of tissue radioactivity between
FAMEs were resolved on a Varian 430 gas chromatograph- FID (Varian, Lake Forest, [U- H]TAG-DHA and [U-3H]PL-DHA was done by independent samples t test. The effects of
3

CA, USA) with a DB-23 capillary column (30 m0.25 mm interior diameter 0.25-m time and tracer on serum lipid class radioactivity were assessed by two-way ANOVA with
lm thickness; Agilent Technologies, Mississauga, ON, USA) with 1-ul injection volume unweighted means analysis with comparisons between tracers at each time point done by
running in splitless mode. Gas chromatography conditions were as previously described independent samples t test. Mean area under the curve was also assessed for each tracer in
[48,55]. Briey, injector and detector ports were 250 C, temperature program was each serum lipid class. Comparison of tissue fatty acid concentrations between groups in
50 C for 2 min, increasing at 20 C/min, and held at 170 C for 1 min, then increased at PL-DHA versus TAG-DHA supplementation study was done by one-way ANOVA with
3 C/min and held at 212 C for 5 min to complete the run at 32 min with helium as Tukey's post-hoc test. Longitudinal changes in DHA concentration in serum lipid classes
carrier gas with ow rate of 0.7 ml/min. Peaks were identied in relation to a reference were expressed relative to OO-fed rat within each week. Effects of all diets within each
standard mixture (GLC 455 mixed with GLC-48 and supplemented with 8:0, 10:0, and week of supplementation were assessed by one-way ANOVA with Tukey's post-hoc test.
12:0 FAMEs, Nuchek Prep). Concentrations of fatty acids were determined in relation to The effects of the different FO, PLC and FO+PLC and time were assessed in longitudinal
peak area of internal standard. serum DHA concentration data by two-way ANOVA.

2.7. PL-DHA vs. TAG-DHA supplementation study calculations 3. Results


The balance method [48,5658] was used to determine brain region, tissue and
whole-body net DHA accretion rates using the following calculation: 3.1. Radiotracer study

Total radioactivity in the brain 6 h after oral gavage of was less than
DHAat beginning of study  DHAat end of study 2 105% of the gavaged dose for both tracers. Rats gavaged with
DHA daily accretion rate 2
number of days of supplementation [U-3H]PC-DHA had a signicantly greater proportion of the radiotracer
94 A.P. Kitson et al. / Journal of Nutritional Biochemistry 33 (2016) 91102

appear in cerebellum, hippocampus and remainder of brain 6-h tions of 9.9, 10.1, 10.6, 11.1, 13.4 and 13.7 mol/g in cortex, remainder
postgavage as compared with rats gavaged with [U-3H]TAG-DHA of brain, cerebellum, brainstem, hippocampus and striatum, respec-
(78%, 140% and 69% higher, respectively) with no signicant tively. Supplementation with 2% of dietary fatty acids as DHA in FO,
differences in cortex, striatum and brainstem (Fig. 1). Unidirectional PLC or FO+PLC resulted in signicantly higher brain DHA concentra-
incorporation coefcients for tracer uptake from serum were higher in tions compared with OO-fed controls (3033% higher in cortex, 35
cerebellum, hippocampus and remainder of brain of [U-3H]PC-DHA 43% higher in cerebellum, 2731% higher in hippocampus, 2832%
compared with [U-3H]TAG-DHA rats with no differences in other higher in brainstem and 2635% higher in remainder of brain) with no
regions regardless of serum lipid pool examined (data not shown). No signicant differences in brain DHA concentration between the three
signicant differences in radioactivity were observed in heart, liver, DHA-supplemented groups (Fig. 3). Striatum DHA concentrations
skin or adipose between [U-3H]TAG-DHA and [U-3H]PC-DHA, al- were not signicantly different between groups (P =.053).
though there were differences in the distribution of radioactivity The changes in fasting serum lipid class DHA concentrations in
among liver lipid classes (Supplementary Fig. 1). response to DHA supplementation are shown in Fig. 4 (full serum lipid
Ingestion of both [U- 3H]TAG-DHA and [U-3H]PC-DHA-labeled class fatty acid compositions at conclusion of study are shown in
serum lipid pools in the rank order TAG N diacylglycerol/cholesterol Supplemental Tables 811). All sources of DHA signicantly increased
N nonesteried fatty acids (NEFA) N PL N monoacylglycerols = all serum lipid DHA concentrations over the course of the study (time
cholesteryl esters (CE). Radioactivity in all fractions appeared to P b .0001 for all lipid classes). The largest increase in DHA concentra-
have plateaued or decreased by the end of the study (Fig. 2). The tion was found in serum triglycerides, with DHA supplementation
highest labeling was observed for serum TAG in the [U-3H]TAG-DHA raising DHA concentration by 2050-fold, with other fractions
gavaged animals at 5-h post gavage with 0.126% of the gavaged dose/ increasing 315-fold. DHA-supplemented rats generally had higher
ml of serum. The pattern of labeling of serum lipid pools was different serum DHA compared to OO controls based on within-week ANOVA
between [U-3H]TAG-DHA and [U-3H]PC-DHA, as two-way ANOVA analysis. In two-way ANOVA examining different DHA-containing
revealed a signicant effect of tracer in the labeling of all serum lipids diets relative to OO-control, a signicant main effect of DHA
pools examined except for total PL (Fig. 2). [U-3H]TAG-DHA tended to supplementation diet was found only for serum unesteried fatty
have higher maximum labeling, reach the maximum labeling faster acids (P =.0051), in which FO-fed rats had higher DHA at 3 weeks of
and maintain a higher value of radioactivity longer compared with supplementation compared with PLC (FO+PLC was intermediate),
[U-3H]PC-DHA in diacylglycerol/cholesterol, NEFA, monoacylglycerol while all groups were signicantly higher than control. The only other
and in particular TAG. In contrast, [U-3H]PC-DHA had higher labeling instance of signicant differences between DHA-supplemented rats
of CE. was in serum TAG after 4 weeks of supplementation, in which
FO+PLC was signicantly higher than FO.
3.2. PL-DHA versus TAG-DHA supplementation study Effects of diets on liver, heart, adipose, intestine and skin DHA
concentration is shown in Fig. 5 (full fatty acid compositions reported
No differences in body weight or food intake between the dietary in Supplemental Tables 1216). Supplementation with DHA increased
groups were observed over the course of the study (Supplemental Fig. total lipid DHA concentration by between 7 and 10 fold in liver, heart,
2). Body weight was 566 53 g at the beginning of supplementation small intestine and skin, while an increase of only three- to fourfold
and 625 64 g at the end of the 4-week supplementation period with was found in adipose tissue (P b.05 for all relative to OO). All DHA-
a signicant main effect of time but not of diet. Food intake was 31 supplemented diets were equally effective at increasing tissue total
5 g/d at the beginning of supplementation and 29 4 g/d at the end of lipid DHA concentrations in all tissues except for the heart, in which
the study with no changes over time and no differences between diets. the FO+PLC-fed rats had 27% higher heart DHA compared with FO-fed
Effects of experimental diets on brain region DHA is shown in Fig. 3 rats, with PLC being intermediate.
(full brain region fatty acid composition can be found in Supplemental The results of the DHA balance analysis are summarized in Table 2.
Tables 27). After 4 weeks of supplementation, rats in the DHA-free Brain region accretion rates were higher in FO, PLC and FO+PLC rats
OO supplemented group had brain region total lipid DHA concentra- compared to OO controls and were not different between DHA-
supplemented groups. Whole body DHA was approximately 10-fold
higher in DHA-supplemented groups compared with OO controls with
no differences between DHA sources, resulting in no differences in the
whole body accretion rates. The apparent daily DHA oxidation rate
was highest in FO+PLC-fed rats compared with PLC-fed rats, with FO-
fed being intermediate (all DHA-supplemented groups were higher
than the OO control group), although all DHA-fed groups had similar
proportions of dietary DHA that was apparently -oxidized, ranging
from 84 to 88% of ingested DHA. Among DHA-fed rats, the fecal DHA
excretion was highest in PLC-fed rats compared with FO and FO+PLC
rats, while all DHA-fed rats had higher fecal DHA excretion compared
with OO-fed controls. The percentage of dietary DHA excreted in feces
was higher in PLC-fed rats compared with FO and FO+PLC and was
less than 0.5% of dietary DHA in all groups.

4. Discussion

Fig. 1. Percent of gavaged radioactivity recovered in total phospholipids of brain regions Numerous groups have developed approaches to target the brain
6 h after gavage of [U-3H]TAG-DHA and [U-3H]PC-DHA. Signicantly higher enrichment with DHA [42,5961]; however, whether dietary strategies can also
was found in remainder of brain, cerebellum and hippocampus in rats gavaged with increase DHA supply to the brain is not clear. Ingestion of PL-DHA has
[U-3H]PC-DHA compared with [U-3H]TAG-DHA. Less than 2105 of the dose was
recovered in total brain phospholipids. Data are mean S.E.M. of 45/group. *:
been suggested to increase blood DHA more effectively compared with
signicantly different from [U-3H]TAG-DHA gavaged rats by independent samples t test TAG-DHA [3133], and previous studies have shown higher brain
(Pb.05). uptake of labeled DHA following ingestion of PL-DHA tracers as
A.P. Kitson et al. / Journal of Nutritional Biochemistry 33 (2016) 91102 95

Fig. 2. Percent of gavaged dose recovered in serum lipid classes over 6 h following gavage of [U-3H]TAG-DHA and [U-3H]PC-DHA. Radioactivity of all lipid classes increased during the
experiment, and DHA ester was a signicant factor in all lipid classes except serum phospholipids. [U-3H]TAG-DHA had higher radioactivity in TAG, unesteried fatty acids,
monoacylglycerols, and diacylglycerol/cholesterol while [U-3H]PC-DHA had higher radioactivity in serum cholesteryl esters. Data is mean S.E.M. of three to ve/time-point/group. *:
signicantly different at indicated time point by independent samples t test (Pb.05). AUC: average area under the curve.

compared with TAG-DHA [42,43]. The present study investigated brain, as no signicant differences in radiotracer enrichment were
differences in the acute brain tracer levels and serum lipid pool found in other tissues examined in general agreement with previous
enrichment following ingestion of [U-3H]PC-DHA and [U-3H]TAG- work [43]. However, the higher brain radiotracer uptake in [U-3H]PC-
DHA and compared this to the effects of dietary supplementation with DHA gavage did not translate into higher brain DHA concentrations in
sources of TAG-DHA, PL-DHA or a mixture of both. Similar to previous PL-DHA fed rats compared with TAG-DHA in the 4-week PL-DHA
studies [42,43], we show that a higher percentage of ingested versus TAG-DHA supplementation study. This indicates that while
[U-3H]PC-DHA radioactivity was found in brain lipids compared ingestion of DHA esteried to PL may increase delivery of DHA to
with [U- 3H]TAG-DHA. This effect appears to be specic to the brain lipids postprandially, this may not necessarily translate into
96 A.P. Kitson et al. / Journal of Nutritional Biochemistry 33 (2016) 91102

Fig. 3. DHA concentration of brain regions from rats fed diets supplemented with olive oil, sh oil, sh roe phospholipid concentrate or a mixture of sh oil and sh roe phospholipid
concentrate. All groups supplemented with DHA had higher brain DHA compared with the olive oil control group and were not different from each other. The only exception was
striatum, in which there was no signicant effect of diet on brain DHA concentration. Data are mean S.E.M. for seven to eight/group. Bars with a different superscript are signicantly
different by Tukey's post-hoc test following one-way ANOVA (Pb.05). OO: olive oil, FO: sh oil, PLC: phospholipid concentrate, FO+PLC: sh oil + phospholipid concentrate.

higher brain DHA concentration over the course of a more prolonged increased accretion of DHA. For example, a previous study from our
dietary intervention. group showed that rats fed ALA- or DHA-containing diets have similar
There are several possible explanations for the apparent lack of brain DHA concentrations despite DHA-fed animals having higher
agreement between the tracer and supplementation experiments. The brain DHA uptake rates [48]. In these cases, the turnover of brain DHA
DHA supplementation diets used contained 2% of fatty acids as DHA, may be increased; however, this requires investigation using in vivo
which has previously been suggested to be the dietary DHA content at kinetic modeling [50]. A greater uptake of a tracer into brain lipids also
which brain DHA is maximal [52]. The time-course of DHA repletion may not represent a major quantitative contribution to DHA
using these diets has not been investigated; however, it is possible that homeostasis and, as a result, may not translate into higher brain
brain DHA had reached maximal concentration in all DHA-fed groups DHA concentration. However, it is possible that the higher brain tracer
by the end of the supplementation period, making it difcult to concentration in [U-3H]PC-DHA compared with [U-3H]TAG-DHA rats
observe differences in DHA concentration. It is possible that rats fed indicates more rapid DHA uptake, which may be advantageous for
PL-DHA sources (PLC and FO+PLC groups) reached the steady-state rapid delivery of DHA in certain clinical settings [62]. Obese Zucker rats
plateau more rapidly compared with FO-fed rats, consistent with fed krill oil, a PL-DHA source, have higher brain PL DHA compared to
higher uptake of [U-3H]PC-DHA compared with [U-3H]TAG-DHA, sh oil-fed rats [63], possibly suggesting a selective effect of PL-DHA
although this potential effect appears to be nullied by extending the on brain DHA in obese animals that may not occur in healthy animals.
supplementation period to at least 4 weeks. In addition, the striatum The mechanism underlying the higher brain radioactivity in
exhibited the lowest DHA accretion rate of any brain region analyzed, [U-3H]PC-DHA compared with [U-3H]TAG-DHA rats is unclear, but
but exhibited no differences in DHA concentration in response to may involve differences in kinetics of serum radiotracer enrichment.
increasing PL-DHA intake. It is also possible that more DHA is entering Slightly higher labeling of serum unesteried fatty acids, the serum
the brain in response to PL-DHA intake, but that it is not resulting in lipid pool that appears to be the main pool for brain DHA uptake
A.P. Kitson et al. / Journal of Nutritional Biochemistry 33 (2016) 91102 97

Fig. 4. DHA concentration of serum lipid fractions from rats fed diets supplemented with OO, FO, PLC or FO+PLC expressed relative to olive oil-supplemented rats. DHA supplementation
increased serum lipid class DHA concentration in all DHA-supplemented groups relative to baseline. The source of DHA was only a signicant factor in the DHA concentration of serum
unesteried fatty acids, in which PLC-supplemented rats had lower DHA than FO rats after 21 days of supplementation, but this difference disappeared at 28 days. FO+PLC had higher
serum TAG DHA at 28 days compared with FO. Data are mean S.E.M. for seven to eight/group. Bars with a different superscript within a time point are signicantly different within that
time point by Tukey's post-hoc test following one-way ANOVA (Pb.05). Indicated P-values are derived from two-way ANOVA of time and DHA ester. OO: olive oil, FO: sh oil, PLC:
phospholipid concentrate, FO+PLC: sh oil + phospholipid concentrate.

[25,27], was observed in rats gavaged with [U-3H]TAG-DHA, which is proposed to be involved in brain DHA uptake [68], and HDL-associated
not in agreement with the lower brain tracer uptake in [U-3H]TAG- radioactivity is detected in mouse brain following injection of labeled
DHA rats relative to [U-3H]PC-DHA. [U-3H]PC-DHA more effectively HDL particles [69]. Maintenance of whole-brain DHA concentration
labeled serum cholesteryl esters, consistent with preferential labeling does not require the low-density or very low density lipoprotein
of HDL that has been observed for dietary PL previously [44,45]. It has receptors [70,71]; however, the role of HDL in brain DHA homeostasis
been proposed that small HDL particles can enter the brain through and targeted delivery of DHA to the brain is not clear.
the action of scavenger receptor Class B, Type I (SR-BI) on brain The postprandial distribution of [U-3H]TAG-DHA and [U-3H]PC-
capillary endothelial cells [6466], suggesting a possible mechanism DHA radioactivity among the different serum lipid pools likely reect
for the higher brain tracer uptake in [U-3H]PC-DHA-gavaged rats the differences in their digestion and absorption. Dietary TAG are
compared with [U-3H]TAG-DHA. SR-BI mediates cholesteryl ester predominantly hydrolyzed by pancreatic lipase, which primarily
inux in the liver [67], and as such may be involved in brain cholesteryl hydrolyzes fatty acids in the sn-1 and 3 positions of TAG, resulting
ester uptake in the brain as well. Brain HDL uptake has previously been in two fatty acids and a sn-2-monoacylglycerol (MAG) [72]. These
98 A.P. Kitson et al. / Journal of Nutritional Biochemistry 33 (2016) 91102

Fig. 5. DHA concentration of tissues from rats fed diets supplemented with OO, FO, PLC and FO+PLC. All DHA-containing diets had higher tissue DHA compared with OO controls.
FO+PLC rats had higher heart DHA compared with FO-fed rats, and DHA concentrations in the rest of the tissues examined were not different among DHA-supplementation groups. Data
are mean S.E.M. for seven to eight/group. Bars with a different superscript are signicantly different by Tukey's post-hoc test following one-way ANOVA (Pb.0.05). OO: olive oil, FO:
sh oil, PLC: phospholipid concentrate, FO+PLC: sh oil + phospholipid concentrate.

products are taken up into the enterocyte, and the 2-MAG is utilized poorer substrate for enterocytic TAG synthesis as compared with
for TAG synthesis for chylomicron secretion [73]. Therefore, at least [U-3H]TAG-DHA, and the more effective labeling of serum cholesteryl
one third of the acyl-chain radioactivity of [U-3H]TAG-DHA is utilized esters may result from the preferential incorporation of acyl moieties
for TAG synthesis, and this is reected in the radioactive enrichment of from ingested PL into HDL observed previously [44,45]. [U-3H]PC-
serum TAG and its metabolites, MAG and DAG following [U-3H]TAG- DHA-gavaged rats also had a higher proportion of liver radioactivity in
DHA ingestion. As the DHA in the [U-3H]TAG-DHA is in the sn-2 the cholesteryl ester fraction, suggesting that secretion of radioactive
position, it is likely that the majority is absorbed as 2-DHA-MAG and CE by the liver may have been higher in these rats compared to
reesteried in the enterocyte. PL, on the other hand, is hydrolyzed by [U-3H]TAG-DHA, although serum lecithin-cholesterol acyltransferase
activities of phospholipase A1 (pancreatic phospholipase A1[74], may also be involved. It has been suggested that sn-1-lyso-2-DHA-PC
secondary activity of pancreatic lipase in some species [75]) and A2 is the main serum DHA pool supplying the brain [30,61]; however, we
(pancreatic phospholipase A2[76], pancreatic lipase-related protein 2 did not observe any differences in serum phospholipid labeling
[77]) into free fatty acids and sn-1/2-lysoPL. In the rat, approximately between the two tracers, and fasting serum lysoPC DHA concentration
half of the acyl chain moiety of ingested PC is absorbed as lyso-PC and was similar between the three DHA-supplemented groups. The
the other half as unesteried fatty acids, with the dominant LPC presence of PL in PL-DHA may increase intestinal absorption of lipids
species being 1-acyl-lysoPC [78], indicating that the major intestinal as biliary PC is required for intestinal TAG absorption [79] and
phospholipase activity in rats is phospholipase A2. As the DHA in the inclusion of PC in intestinal lipid infusion increases TAG absorption
[U-3H]PC-DHA tracer was in the sn-2 position, it is likely that it was [7981]. However, similar whole-body DHA accretion rates between
absorbed in the unesteried form. [U-3H]PC-DHA appears to be a FO, PLC and FO+PLC rats indicate that inclusion of PL-DHA in DHA
A.P. Kitson et al. / Journal of Nutritional Biochemistry 33 (2016) 91102 99

Table 2
Summary of DHA balance analysis
Olive oil Fish oil Phospholipid concentrate Fish oil + phospholipid concentrate

Whole-body DHA after 4 weeks (mol) 685a 68057b 79762b 68964b


Total dietary DHA intake (mol) 14620a 54021147b 5141803b 6145454b
Whole body DHA accretion (mol) 235a 63657b 75262b 64426b
Whole body DHA accretion rate (mol/d) 0.80.2a 22.72.0b 26.82.2b 23.02.3b
Metabolic DHA consumption rate (mol/d) 31a 16816bc 1589b 1946c
% Apparent Dietary DHA -oxidized 524a 871b 841b 881b
Total fecal DHA excretion (mol) 0.60.1a 9.62.1b 23.01.6c 11.60.9b
% Dietary DHA excreted in feces 0.430.10ab 0.180.04a 0.460.04b 0.190.1a
Cortex DHA accretion rate (nmol/g/d) 1518a 1315b 1284b 1249b
Cerebellum DHA accretion rate (nmol/g/d) 319a 10124b 9511b 1126b
Hippocampus DHA accretion rate (nmol/g/d) 925a 14031b 12113b 13726b
Striatum DHA accretion rate (nmol/g/d) 31105 346 369 5417
Brainstem DHA accretion rate (nmol/g/d) 1134a 10110b 10311b 11515b
Remainder of brain DHA accretion rate (nmol/g/d) 2910a 7312b 6713b 964b

Data are mean S.E.M. n=78/group. After 11-week n-3 PUFA deprivation, 8 rats were sacriced to measure baseline DHA concentrations, and the difference after 4 weeks of
supplementation with DHA or control diet were determined to measure accretion rates. At baseline, whole-body DHA was 453 mol (n=8). Different superscript letters that indicate
means are signicantly different (Pb.05) determined by Tukey's post-hoc comparison following one-way ANOVA.

formulations does not affect the amount of DHA absorbed, and in fact piglet gray matter, respectively[42, 43]) considering differences in
the PLC-fed rats had the highest fecal DHA concentration (albeit less sampling time postgavage, species, age of animals, analytical meth-
than 0.5% of dietary DHA). However, a more direct measure of odologies and diet between the studies. This suggests that only a
intestinal DHA absorption is needed to denitively conclude this. minor fraction of ingested DHA targets the brain, regardless of the
Importantly, the differences in postprandial serum lipid radioac- form in which it is consumed. The major fate of PL-DHA and TAG-DHA
tivity following gavage of [U-3H]TAG-DHA and [U-3H]PC-DHA did is oxidation, as evidenced by the high proportion of the DHA
not translate into clear differences in fasting serum lipid DHA consumed being -oxidized (8488%), consistent with previous
concentration in response to supplementing with a PL- compared studies showing high DHA oxidation using the balance method to
with TAG-DHA source for 4 weeks. Differences in serum lipid DHA examine DHA balance when fed as microalgal oil [58] or ethyl ester
among the PL- and TAG-DHA-supplemented groups were time-point [48], and the high proportion of label recovered in breath following
specic, and PL- versus TAG-DHA source was not a signicant factor for ingestion of [U-13C]DHA following sh oil supplementation [86].
any lipid class except for unesteried fatty acids. This suggests that Interestingly, the OO-fed rats had the lowest proportion of dietary
analysis of postprandial absorption kinetics will not match fasting DHA oxidized (52%), indicating that DHA may be selectively retained
blood sampling following an intervention period. A recent randomized in these animals, consistent with previous work demonstrating
controlled trial comparing krill oil (PL-EPA + DHA source) and sh oil extended DHA half-life in brain following 15-week n-3 PUFA
(TAG-EPA+DHA source) shows that krill oil had higher postprandial deprivation [46] and lower breath recovery of [U-13C]DHA label in
serum phospholipid EPA+DHA, but that this trend was reversed individuals pre- compared with postsh oil supplementation. A higher
(albeit not statistically signicant) when examining plasma TAG rate of apparent metabolic DHA consumption (i.e., apparent -
EPA+DHA [82]. Thus, the lipid pool examined can have signicant oxidation) was observed in rats fed the FO+PLC diet compared to
effects on the conclusions of DHA bioavailability studies, indicating PLC, but this appears to have resulted from slightly higher intake of
that such studies should, in the absence of any clear rationale for DHA, as the percent of ingested DHA that was -oxidized was not
investigating a particular serum lipid class, examine bioavailability of different between groups. Krill-oil fed rats have recently been shown
multiple serum lipid pools or of serum total lipids. Some of these to accrete a higher proportion of dietary DHA in whole body compared
factors have been previously proposed to introduce signicant with sh oil-fed rats after 6 weeks of supplementation [87]. No
heterogeneity in the assessment of DHA bioavailability [39]. signicant differences in whole-body DHA accretion were observed in
The mixture of the FO and PLC DHA sources appeared to have a the present study of 4-week supplementation; however, PL-fed rats
positive effect on some measures of DHA concentration. The FO+PLC had nonsignicantly higher whole-body DHA accretion compared to
group (in which approximately 30% of the total DHA was esteried to FO and FO+PLC-fed rats. It is possible that longer duration of
phospholipids) had higher heart DHA and serum TAG DHA compared supplementation is required to elicit differences in whole-body DHA
with the FO group, consistent with higher heart DHA in krill oil- versus accretion between PL- and TAG-DHA.
sh oil-fed obese Zucker rats [83]. Interestingly, the higher heart DHA There are several considerations regarding the interpretation of
in the FO+PLC rats relative to the FO rats did not correspond to this work. The isotope used was uniformly labeled, and as such, the
differences in heart radioactivity between [U- 3H]PC-DHA and conclusions rendered from this study pertain to the entire sn-1,3-
[U-3H]TAG-DHA rats, suggesting that this effect is due to either the palmitoyl-2-docosahexaenoylglycerol or 1-palmitoyl-2-docosahex-
specic mixture of PL- and TAG-DHA or the 4 weeks of supplemen- aenoyl-sn-glycero-3-phosphocholine and not necessarily the DHA
tation. In the heart, DHA is also extensively incorporated into contained within them. In [U-3H]TAG-DHA, the acyl chain radioactiv-
cardiolipin [84] which is associated with modications to mitochon- ity is one-third DHA and two-thirds palmitate, while in [U-3H]PC-DHA
drial enzyme function [85]. The acute absorption and tissue uptake it is one-half DHA and one-half palmitate. This likely does not affect
kinetics of various mixtures of various ratios PL-DHA and TAG-DHA the serum lipid labeling of the tracers, as the appearance of DHA and
warrant investigation, as do the potential cardioprotective effects palmitate in serum lipids is very similar when provided orally as
specic to certain mixtures of PL-DHA and TAG-DHA. unesteried fatty acids (TAGNNNEFANPLNDAGNCE for DHA [88] and
The amount of tracer that appeared in the brain for both [U-3H]PC- TAGNNNEFANCENPLNDAG for palmitate [27]). In addition, little
DHA and [U-3H]TAG-DHA tracers was low (less than 2 105% of the conversion of DHA to other fatty acids is likely to have occurred in
dose) but is roughly consistent with previous studies (approximately this study, as other studies have found low enrichment of other fatty
0.2% and 0.4% of dose in after 24 h in rat whole brain and 6 days in acids with label provided as DHA 22 h after oral administration in liver
100 A.P. Kitson et al. / Journal of Nutritional Biochemistry 33 (2016) 91102

in suckling rats [89] or 5 h after intravenous administration in plasma Appendix A. Supplementary data
in maternal baboons [90]. Similarly, appearance of orally administered
palmitate label as 18:0 and 16:1 is approximately 8% after 7 days in Supplementary data to this article can be found online at http://dx.
humans [91] and has been below the detection limit 24 h after oral doi.org/10.1016/j.jnutbio.2016.02.009.
dose in humans in one study [92], suggesting that the majority of
labeled palmitate ingested remains as labeled palmitate over the
course of this study. The higher brain radioactivity resulting from References
[U-3H]PC-DHA compared with [U-3H]TAG-DHA gavage presently is
consistent with previous work in which only the sn-2 DHA is labeled [1] Igarashi M, Ma K, Gao F, Kim HW, Greenstein D, Rapoport SI, et al. Brain lipid
concentrations in bipolar disorder. J Psychiatr Res 2010;44:17782.
[42,43], indicating that this higher brain radioactivity in [U-3H]PC- [2] Rao JS, Ertley RN, Lee HJ, DeMar Jr JC, Arnold JT, Rapoport SI, et al. n-3
DHA rats was, at least in part, due to higher brain uptake of the polyunsaturated fatty acid deprivation in rats decreases frontal cortex BDNF via a
[3H]DHA moiety. Interestingly, baboon neonate brain takes up more p38 MAPK-dependent mechanism. Mol Psychiatry 2007;12:3646.
[3] Cao D, Kevala K, Kim J, Moon HS, Jun SB, Lovinger D, et al. Docosahexaenoic acid
arachidonate when fed as sn-2-arachidonate-PC compared with sn-2- promotes hippocampal neuronal development and synaptic function. J Neuro-
arachidonate-TAG [93], suggesting that this effect is not limited to sn- chem 2009;111:51021.
2-DHA-PC. The tracers were both dissolved in olive oil (virtually [4] Su HM. Mechanisms of n-3 fatty acid-mediated development and maintenance of
learning memory performance. J Nutr Biochem 2010;21:36473.
entirely TAG) prior to gavage, and as such, the absorption matrix for [5] Orr SK, Palumbo S, Bosetti F, Mount HT, Kang JX, Greenwood CE, et al. Unesterified
both TAG and PL tracers was TAG. This may have had some effects on docosahexaenoic acid is protective in neuroinflammation. J Neurochem 2013;
the absorption kinetics of the PL tracer. The DAG and cholesterol 127:37893.
[6] Brenna JT. Animal studies of the functional consequences of suboptimal
fractions were collected together in the radiotracer gavage study, and polyunsaturated fatty acid status during pregnancy, lactation and early post-
so the distribution of radioactivity between these two fractions is natal life. Matern Child Nutr 2011;7(Suppl. 2):5979.
unclear as carbon recycling into cholesterol could account for some of [7] Igarashi M, Ma K, Gao F, Kim HW, Rapoport SI, Rao JS. Disturbed choline
plasmalogen and phospholipid fatty acid concentrations in Alzheimer's disease
the radioactivity. However, the appearance of carbon in serum saturated
prefrontal cortex. J Alzheimers Dis 2011;24:50717.
fatty acids is low for DHA in rats over this time course (b 3% of serum [8] Soderberg M, Edlund C, Kristensson K, Dallner G. Fatty acid composition of brain
label) [90], suggesting that cholesterol labeling via carbon recycling of phospholipids in aging and in Alzheimer's disease. Lipids 1991;26:4215.
labeled DHA was likely low in this study. Another factor is the baseline [9] Lukiw WJ, Cui JG, Marcheselli VL, Bodker M, Botkjaer A, Gotlinger K, et al. A role for
docosahexaenoic acid-derived neuroprotectin D1 in neural cell survival and
diet used in these studies, which is a low n-3 PUFA diet that has Alzheimer disease. J Clin Invest 2005;115:277483.
previously been shown to increase brain DHA half-life [46] and hepatic [10] Astarita G, Jung KM, Berchtold NC, Nguyen VQ, Gillen DL, Head E, et al. Deficient
synthesis rate [94]. The present studies therefore represent n-3 PUFA liver biosynthesis of docosahexaenoic acid correlates with cognitive impairment
in Alzheimer's disease. PLoS One 2010;5, e12538.
repletion on a very low n-3 PUFA background. However, the association [11] Cunnane SC, Schneider JA, Tangney C, Tremblay-Mercier J, Fortier M, Bennett DA,
between low n-3 PUFA status and neurodegenerative [711] and et al. Plasma and brain fatty acid profiles in mild cognitive impairment and
neuropsychiatric disorders [1316] and cardiovascular mortality [95] Alzheimer's disease. J Alzheimers Dis 2012;29:6917.
[12] Cunnane SC, Chouinard-Watkins R, Castellano CA, Barberger-Gateau P. Docosa-
suggests that this experimental approach is relevant for determining hexaenoic acid homeostasis, brain aging and Alzheimer's disease: can we
the relative efcacy of different dietary forms of DHA on tissue DHA reconcile the evidence? Prostaglandins Leukot Essent Fatty Acids 2013;88:6170.
uptake and concentration. Also, as the diet is essentially free of ALA [13] McNamara RK, Jandacek R, Tso P, Dwivedi Y, Ren X, Pandey GN. Lower
docosahexaenoic acid concentrations in the postmortem prefrontal cortex of
(b 0.2% of dietary fatty acids), DHA synthesis from dietary ALA likely adult depressed suicide victims compared with controls without cardiovascular
does not contribute appreciably to changes in tissue and blood DHA disease. J Psychiatr Res 2013;47:118791.
status throughout the study between DHA-supplemented groups [48]. [14] McNamara RK, Hahn CG, Jandacek R, Rider T, Tso P, Stanford KE, et al. Selective
deficits in the omega-3 fatty acid docosahexaenoic acid in the postmortem
The three DHA-supplemented groups were matched for DHA intake;
orbitofrontal cortex of patients with major depressive disorder. Biol Psychiatry
however, there were small differences in the EPA and 22:5n-3 dietary 2007;62:1724.
content. These differences amounted to a range in total dietary n-3 PUFA [15] McNamara RK, Jandacek R, Rider T, Tso P, Stanford KE, Hahn CG, et al. Deficits in
docosahexaenoic acid and associated elevations in the metabolism of arachidonic
content between the three DHA-supplemented groups of only 3.31
acid and saturated fatty acids in the postmortem orbitofrontal cortex of patients
3.41% of total fatty acids. with bipolar disorder. Psychiatry Res 2008;160:28599.
This study conrms greater brain uptake following an oral tracer [16] McNamara RK, Jandacek R, Rider T, Tso P, Hahn CG, Richtand NM, et al.
dose of DHA esteried to PL compared to TAG [42,43] but shows that Abnormalities in the fatty acid composition of the postmortem orbitofrontal
cortex of schizophrenic patients: gender differences and partial normalization
supplementing diets with a source of PL-DHA, TAG-DHA or a mixture with antipsychotic medications. Schizophr Res 2007;91:3750.
of both results in similar increases in brain DHA compared to a low n-3 [17] Hamazaki K, Hamazaki T, Inadera H. Abnormalities in the fatty acid composition of
diet. This suggests that the higher tracer uptake of PL-DHA compared the postmortem entorhinal cortex of patients with schizophrenia, bipolar
disorder, and major depressive disorder. Psychiatry Res 2013;210:34650.
to TAG-DHA does not necessarily translate into higher brain DHA [18] Hamazaki K, Hamazaki T, Inadera H. Fatty acid composition in the postmortem
concentration. In addition, signicant differences were observed in the amygdala of patients with schizophrenia, bipolar disorder, and major depressive
postprandial serum lipid radioactivity prole following gavage of disorder. J Psychiatr Res 2012;46:10248.
[19] Fraser T, Tayler H, Love S. Fatty acid composition of frontal, temporal and parietal
[U-3H]PC-DHA compared with [U-3H]TAG-DHA, while only time neocortex in the normal human brain and in Alzheimer's disease. Neurochem Res
point-specic differences were observed in fasting serum DHA 2010;35:50313.
concentrations during the 4-week PL-DHA versus TAG-DHA supple- [20] Oster T, Pillot T. Docosahexaenoic acid and synaptic protection in Alzheimer's
disease mice. Biochim Biophys Acta 2010;1801:7918.
mentation study. This indicates that the study and sampling protocol
[21] Cole GM, Frautschy SA. Docosahexaenoic acid protects from amyloid and
can have signicant effects on the conclusions obtained from studies dendritic pathology in an Alzheimer's disease mouse model. Nutr Health 2006;
evaluating the effects of PL-DHA compared with TAG-DHA on blood 18:24959.
DHA levels. Overall, there appear to be differences in the digestion, [22] Zugno AI, Chipindo HL, Volpato AM, Budni J, Steckert AV, de Oliveira MB, et al.
Omega-3 prevents behavior response and brain oxidative damage in the ketamine
absorption and brain uptake of DHA esteried to PL compared to TAG; model of schizophrenia. Neuroscience 2014;259:22331.
however, these postprandial differences may not translate into [23] Park Y, Moon HJ, Kim SH. N-3 polyunsaturated fatty acid consumption produces
increased longer term blood and tissue DHA status following neurobiological effects associated with prevention of depression in rats after the
forced swimming test. J Nutr Biochem 2012;23:9248.
supplementation with PL-DHA compared with TAG-DHA. [24] Igarashi M, DeMar Jr JC, Ma K, Chang L, Bell JM, Rapoport SI. Docosahexaenoic acid
synthesis from alpha-linolenic acid by rat brain is unaffected by dietary n-3 PUFA
Potential conflict of interest deprivation. J Lipid Res 2007;48:11508.
[25] Moore SA, Yoder E, Murphy S, Dutton GR, Spector AA. Astrocytes, not neurons,
produce docosahexaenoic acid (22:6 omega-3) and arachidonic acid (20:4
This study was funded by Arctic Nutrition AS, NO-6155 rsta, Norway. omega-6). J Neurochem 1991;56:51824.
A.P. Kitson et al. / Journal of Nutritional Biochemistry 33 (2016) 91102 101

[26] Contreras MA, Greiner RS, Chang MC, Myers CS, Salem Jr N, Rapoport SI. [53] Wang WQ, Gustafson A. One-dimensional thin-layer chromatographic separation
Nutritional deprivation of alpha-linolenic acid decreases but does not abolish of phospholipids and lysophospholipids from tissue lipid extracts. J Chromatogr
turnover and availability of unacylated docosahexaenoic acid and docosahex- 1992;581:13942.
aenoyl-CoA in rat brain. J Neurochem 2000;75:2392400. [54] Morrison WR, Smith LM. Preparation of fatty acid methyl esters and dimethy-
[27] Purdon D, Arai T, Rapoport S. No evidence for direct incorporation of esterified lacetals from lipids with boron fluoride-methanol. J Lipid Res 1964;4:6008.
palmitic acid from plasma into brain lipids of awake adult rat. J Lipid Res 1997;38: [55] Chen CT, Liu Z, Bazinet RP. Rapid de-esterification and loss of eicosapentaenoic
52630. acid from rat brain phospholipids: an intracerebroventricular study. J Neurochem
[28] Chen CT, Kitson AP, Hopperton KE, Domenichiello AF, Lin L, Trepanier MO, et al. 2011;116:36373.
Plasma non-esterified docosahexaenoic acid is the major pool supplying the brain. [56] Yang J, Chen ZY, Cunnane SC. Application of the balance method to determining
Sci Rep 2015 [in press]. accumulation, metabolism, and apparent oxidation of linoleic and alpha-linolenic
[29] Nguyen LN, Ma D, Shui G, Wong P, Cazenave-Gassiot A, Zhang X, et al. Mfsd2a is a acids in the pregnant rat. Metabolism 1994;43:9404.
transporter for the essential omega-3 fatty acid docosahexaenoic acid. Nature [57] Cunnane SC, Anderson MJ. The majority of dietary linoleate in growing rats is
2014;509. beta-oxidized or stored in visceral fat. J Nutr 1997;127:14652.
[30] Thies F, Pillon C, Moliere P, Lagarde M, Lecerf J. Preferential incorporation of sn-2 [58] Poumes-Ballihaut C, Langelier B, Houlier F, Alessandri JM, Durand G, Latge C, et al.
lysoPC DHA over unesterified DHA in the young rat brain. Am J Physiol 1994;267: Comparative bioavailability of dietary alpha-linolenic and docosahexaenoic acids
R12739. in the growing rat. Lipids 2001;36:793800.
[31] Ramprasath VR, Eyal I, Zchut S, Jones PJ. Enhanced increase of omega-3 index in [59] Belayev L, Khoutorova L, Atkins KD, Eady TN, Hong S, Lu Y, et al. Docosahexaenoic
healthy individuals with response to 4-week n-3 fatty acid supplementation from acid therapy of experimental ischemic stroke. Transl Stroke Res 2011;2:3341.
krill oil versus fish oil. Lipids Health Dis 2013;12:178. [60] Hachem M, Geloen A, Van AL, Foumaux B, Fenart L, Gosselet F, et al. Efficient
[32] Ulven SM, Kirkhus B, Lamglait A, Basu S, Elind E, Haider T, et al. Metabolic effects of Docosahexaenoic Acid Uptake by the Brain from a Structured Phospholipid. Mol
krill oil are essentially similar to those of fish oil but at lower dose of EPA and DHA, Neurobiol 2015. http://dx.doi.org/10.1007/s12035-015-9228-9.
in healthy volunteers. Lipids 2011;46:3746. [61] Lagarde M, Bernoud N, Brossard N, Lemaitre-Delaunay D, Thies F, Croset M, et al.
[33] Maki KC, Reeves MS, Farmer M, Griinari M, Berge K, Vik H, et al. Krill oil Lysophosphatidylcholine as a preferred carrier form of docosahexaenoic acid to
supplementation increases plasma concentrations of eicosapentaenoic and the brain. J Mol Neurosci 2001;16:2014 [discussion 15-21].
docosahexaenoic acids in overweight and obese men and women. Nutr Res [62] Barrett EC, McBurney MI, Ciappio ED. Omega-3 fatty acid supplementation as a
2009;29:60915. potential therapeutic aid for the recovery from mild traumatic brain injury/
[34] Schuchardt JP, Schneider I, Meyer H, Neubronner J, von Schacky C, Hahn A. concussion. Adv Nutr 2014;5:26877.
Incorporation of EPA and DHA into plasma phospholipids in response to different [63] Di Marzo V, Grinari M, Carta G, Murru E, Ligresti A, Cordeddu L, et al. Dietary krill
omega-3 fatty acid formulationsa comparative bioavailability study of fish oil vs. oil increases docosahexaenoic acid and reduces 2-arachidonoylglycerol but not N-
krill oil. Lipids Health Dis 2011;10:145. acylethanolamine levels in the brain of obese Zucker rats. Int Dairy J 2010;20:
[35] Vaisman N, Kaysar N, Zaruk-Adasha Y, Pelled D, Brichon G, Zwingelstein G, et al. 2315.
Correlation between changes in blood fatty acid composition and visual sustained [64] Goti D, Hrzenjak A, Levak-Frank S, Frank S, van der Westhuyzen DR, Malle E, et al.
attention performance in children with inattention: effect of dietary n-3 fatty Scavenger receptor class B, type I is expressed in porcine brain capillary
acids containing phospholipids. Am J Clin Nutr 2008;87:117080. endothelial cells and contributes to selective uptake of HDL-associated vitamin
[36] Carnielli VP, Verlato G, Pederzini F, Luijendijk I, Boerlage A, Pedrotti D, et al. E. J Neurochem 2001;76:498508.
Intestinal absorption of long-chain polyunsaturated fatty acids in preterm infants [65] Vitali C, Wellington CL, Calabresi L. HDL and cholesterol handling in the brain.
fed breast milk or formula. Am J Clin Nutr 1998;67:97103. Cardiovasc Res 2014;103:40513.
[37] Yurko-Mauro K, Kralovec J, Bailey-Hall E, Smeberg V, Stark JG, Salem Jr N. Similar [66] Panzenboeck U, Balazs Z, Sovic A, Hrzenjak A, Levak-Frank S, Wintersperger A,
eicosapentaenoic acid and docosahexaenoic acid plasma levels achieved with fish et al. ABCA1 and scavenger receptor class B, type I, are modulators of reverse
oil or krill oil in a randomized double-blind four-week bioavailability study. Lipids sterol transport at an in vitro bloodbrain barrier constituted of porcine brain
Health Dis 2015;14:99. capillary endothelial cells. J Biol Chem 2002;277:427819.
[38] Salem Jr N, Kuratko CN. A reexamination of krill oil bioavailability studies. Lipids [67] Acton S, Rigotti A, Landschulz KT, Xu S, Hobbs HH, Krieger M. Identification of
Health Dis 2014;13:137. scavenger receptor SR-BI as a high density lipoprotein receptor. Science 1996;
[39] Ghasemifard S, Turchini GM, Sinclair AJ. Omega-3 long chain fatty acid 271:51820.
"bioavailability": a review of evidence and methodological considerations. Prog [68] Polozova A, Salem Jr N. Role of liver and plasma lipoproteins in selective transport
Lipid Res 2014;56:92108. of n-3 fatty acids to tissues: a comparative study of 14C-DHA and 3H-oleic acid
[40] Nichols PD, Kitessa SM, Abeywardena M. Commentary on a trial comparing krill tracers. J Mol Neurosci 2007;33:5666.
oil versus fish oil. Lipids Health Dis 2014;13:2. [69] Polozova A, Gionfriddo E, Salem Jr N. Effect of docosahexaenoic acid on tissue
[41] Ramprasath VR, Eyal I, Zchut S, Jones PJ. Response to commentary on a trial targeting and metabolism of plasma lipoproteins. Prostaglandins Leukot Essent
comparing krill oil versus fish oil. Lipids Health Dis 2014;13:17. Fatty Acids 2006;75:18390.
[42] Liu L, Bartke N, Van Daele H, Lawrence P, Qin X, Park HG, et al. Higher efficacy of [70] Rahman T, Taha AY, Song BJ, Orr SK, Liu Z, Chen CT, et al. The very low
dietary DHA provided as a phospholipid than as a triglyceride for brain DHA density lipoprotein receptor is not necessary for maintaining brain polyunsatu-
accretion in neonatal piglets. J Lipid Res 2014;55:5319. rated fatty acid concentrations. Prostaglandins Leukot Essent Fatty Acids 2010;82:
[43] Graf BA, Duchateau GS, Patterson AB, Mitchell ES, van Bruggen P, Koek JH, et al. 1415.
Age dependent incorporation of 14C-DHA into rat brain and body tissues after [71] Chen CT, Ma DW, Kim JH, Mount HT, Bazinet RP. The low density lipoprotein
dosing various 14C-DHA-esters. Prostaglandins Leukot Essent Fatty Acids 2010; receptor is not necessary for maintaining mouse brain polyunsaturated fatty acid
83:8996. concentrations. J Lipid Res 2008;49:14752.
[44] Zierenberg O, Grundy SM. Intestinal absorption of polyenephosphatidylcholine in [72] Tso P, Hayashi H. The physiology and regulation of the intestinal absorption and
man. J Lipid Res 1982;23:113642. transport of omega-3 and omega-6 fatty acids. Adv Prostaglandin Thromboxane
[45] Amate L, Gil A, Ramirez M. Feeding infant piglets formula with long-chain Leukot Res 1989;19:6236.
polyunsaturated fatty acids as triacylglycerols or phospholipids influences the [73] Schuchardt JP, Hahn A. Bioavailability of long-chain omega-3 fatty acids.
distribution of these fatty acids in plasma lipoprotein fractions. J Nutr 2001;131: Prostaglandins Leukot Essent Fatty Acids 2013;89:18.
12505. [74] Arima N, Inoue A, Makide K, Nonaka T, Aoki J. Surface loops of extracellular
[46] DeMar Jr JC, Ma K, Bell JM, Rapoport SI. Half-lives of docosahexaenoic acid in rat phospholipase A(1) determine both substrate specificity and preference for
brain phospholipids are prolonged by 15 weeks of nutritional deprivation of n-3 lysophospholipids. J Lipid Res 2012;53:51321.
polyunsaturated fatty acids. J Neurochem 2004;91:112537. [75] Aloulou A, Frikha F, Noiriel A, Bou Ali M, Abousalham A. Kinetic and structural
[47] Folch J, Lees M, Sloane Stanley GHS. A simple method for the isolation and characterization of triacylglycerol lipases possessing phospholipase A1 activity.
purification of total lipides from animal tissues. J Biol Chem 1957;226:497509. Biochim Biophys Acta 1841;2014:5817.
[48] Domenichiello AF, Chen CT, Trepanier MO, Stavro PM, Bazinet RP. Whole body [76] Ono T, Tojo H, Inoue K, Kagamiyama H, Yamano T, Okamoto M. Rat pancreatic
synthesis rates of docosahexaenoic acid (DHA) from alpha-linolenic acid are phospholipase A2: purification, characterization, and N-terminal amino acid
greater than brain DHA accretion and uptake rates in adult rats. J Lipid Res 2013; sequence. J Biochem (Tokyo) 1984;96:78592.
55:6274. [77] Roussel A, Yang Y, Ferrato F, Verger R, Cambillau C, Lowe M. Structure and activity
[49] Chen CT, Domenichiello AF, Trepanier MO, Liu Z, Masoodi M, Bazinet RP. The low of rat pancreatic lipase-related. protein 2. J Biol Chem 1998;273:321218.
levels of eicosapentaenoic acid in rat brain phospholipids are maintained via [78] Le Kim D, Betzing H. Intestinal absorption of polyunsaturated phosphatidylcho-
multiple redundant mechanisms. J Lipid Res 2013;54:241022. line in the rat. Hoppe Seylers Z Physiol Chem 1976;357:132131.
[50] Robinson PJ, Noronha J, DeGeorge JJ, Freed LM, Nariai T, Rapoport SI. A [79] Tso P, Kendrick H, Balint JA, Simmonds WJ. Role of biliary phosphatidylcholine in
quantitative method for measuring regional in vivo fatty-acid incorporation the absorption and transport of dietary triolein in the rat. Gastroenterology 1981;
into and turnover within brain phospholipids: review and critical analysis. Brain 80:605.
Res Brain Res Rev 1992;17:187214. [80] Mansbach II CM, Arnold A, Cox MA. Factors influencing triacylglycerol delivery
[51] Lin LE, Chen CT, Hildebrand KD, Liu Z, Hopperton KE, Bazinet RP. Chronic dietary into mesenteric lymph. Am J Physiol 1985;249:G6428.
n-6 PUFA deprivation leads to conservation of arachidonic acid and more rapid [81] Nishimukai M, Wakisaka T, Hara H. Ingestion of plasmalogen markedly increased
loss of DHA in rat brain phospholipids. J Lipid Res 2015;56:390402. plasmalogen levels of blood plasma in rats. Lipids 2003;38:122735.
[52] Orr SK, Tong JY, Kang JX, Ma DW, Bazinet RP. The fat-1 mouse has brain [82] Kohler A, Sarkkinen E, Tapola N, Niskanen T, Bruheim I. Bioavailability of fatty
docosahexaenoic acid levels achievable through fish oil feeding. Neurochem Res acids from krill oil, krill meal and fish oil in healthy subjectsa randomized, single-
2010;35:8119. dose, cross-over trial. Lipids Health Dis 2015;14:19.
102 A.P. Kitson et al. / Journal of Nutritional Biochemistry 33 (2016) 91102

[83] Batetta B, Griinari M, Carta G, Murru E, Ligresti A, Cordeddu L, et al. [90] Sheaff Greiner RC, Zhang Q, Goodman KJ, Giussani DA, Nathanielsz PW, Brenna JT.
Endocannabinoids may mediate the ability of (n-3) fatty acids to reduce ectopic Linoleate, alpha-linolenate, and docosahexaenoate recycling into saturated and
fat and inflammatory mediators in obese zucker rats. J Nutr 2009;139:1495501. monounsaturated fatty acids is a major pathway in pregnant or lactating adults
[84] Berger A, Gershwin ME, German JB. Effects of various dietary fats on cardiolipin and fetal or infant rhesus monkeys. J Lipid Res 1996;37:267586.
acyl composition during ontogeny of mice. Lipids 1992;27:60512. [91] Rhee SK, Kayani AJ, Ciszek A, Brenna JT. Desaturation and interconversion of
[85] Malis CD, Weber PC, Leaf A, Bonventre JV. Incorporation of marine lipids into dietary stearic and palmitic acids in human plasma and lipoproteins. Am J Clin
mitochondrial membranes increases susceptibility to damage by calcium and Nutr 1997;65:4518.
reactive oxygen species: evidence for enhanced activation of phospholipase A2 in [92] Emken EA, Rohwedder WK, Adlof RO, Rakoff H, Gulley RM. Metabolism in humans
mitochondria enriched with n-3 fatty acids. Proc Natl Acad Sci U S A 1990;87:88459. of cis-12,trans-15-octadecadienoic acid relative to palmitic, stearic, oleic and
[86] Plourde M, Chouinard-Watkins R, Rioux-Perreault C, Fortier M, Dang MT, Allard linoleic acids. Lipids 1987;22:495504.
MJ, et al. Kinetics of 13C-DHA before and during fish-oil supplementation in [93] Wijendran V, Huang MC, Diau GY, Boehm G, Nathanielsz PW, Brenna JT. Efficacy
healthy older individuals. Am J Clin Nutr 2014;100:10512. of dietary arachidonic acid provided as triglyceride or phospholipid as substrates
[87] Ghasemifard S, Hermon K, Turchini GM, Sinclair AJ. Metabolic fate (absorption, for brain arachidonic acid accretion in baboon neonates. Pediatr Res 2002;51:
beta-oxidation and deposition) of long-chain n-3 fatty acids is affected by sex and 26572.
by the oil source (krill oil or fish oil) in the rat. Br J Nutr 2015:19. [94] Igarashi M, DeMar Jr JC, Ma K, Chang L, Bell JM, Rapoport SI. Upregulated liver
[88] Chen CT, Kitson AP, Hopperton KE, Domenichiello AF, Trepanier MO, Lin LE, et al. conversion of alpha-linolenic acid to docosahexaenoic acid in rats on a 15 week n-
Plasma non-esterified docosahexaenoic acid is the major pool supplying the brain. 3 PUFA-deficient diet. J Lipid Res 2007;48:15264.
Sci Rep 2015;5:15791. [95] Chowdhury R, Warnakula S, Kunutsor S, Crowe F, Ward HA, Johnson L, et al.
[89] Sinclair AJ. Incorporation of radioactive polyunsaturated fatty acids into liver and Association of dietary, circulating, and supplement fatty acids with coronary risk:
brain of developing rat. Lipids 1975;10:17584. a systematic review and meta-analysis. Ann Intern Med 2014;160:398406.

You might also like