Life Sciences

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 7

Life Sciences 199 (2018) 16–22

Contents lists available at ScienceDirect

Life Sciences
journal homepage: www.elsevier.com/locate/lifescie

Hypolipidemic, antioxidant and antiatherogenic property of sardine T


by-products proteins in high-fat diet induced obese rats
Fouad Affane, Sabrine Louala, Nour el Imane Harrat, Fatima Bensalah, Hadjera Chekkal,

Amine Allaoui, Myriem Lamri-Senhadji
Laboratory of Clinical and Metabolic Nutrition (LNCM), Faculty of Nature and Life Sciences, University of Oran 1 Ahmed Ben Bella, BP 1524 El m'nouer, 31100 Oran,
Algeria

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

Keywords: Aims: Fish by-products valorization on account of their richness in bioactive compounds may represent a better
Fish by-products alternative to marine products with a view to economic profitability and sustainable development. In this study,
High-fat diet we compared the effect of sardine by-product proteins (SBy-P), with those of the fillets (SF-P) or casein (Cas), on
Hyperlipidemia growth parameters, serum leptin level, lipids disorders, lipid peroxidation and reverse cholesterol transport, in
Lipid peroxidation
diet-induced obese rats.
Obese rats
Main methods: Obesity was induced by feeding rats a high-fat diet (20% sheep fat), during 12 weeks. At body
Sardine proteins
weight (BW) of 400 ± 20 g, eighteen obese rats were divided into three homogenous groups and continue to
consume the high-fat diet for 4 weeks containing either, 20% SBy-P, SF-P or Cas.
Key findings: The results showed that SBy-P, compared to SF-P and Cas, efficiently reduced food intake (FI), BW
gain and serum leptin level, and improved blood lipids levels and reverse cholesterol transport by reducing total
cholesterol (TC), triacylglycerols (TG) and low-density lipoprotein cholesterol (LDL-HDL1-C) serum levels, in-
creasing the level of high-density lipoprotein cholesterol (HDL2-C and HDL3-C), and enhancing lecithin: cho-
lesterol acyltransferase (LCAT) activity. Furthermore, they attenuated lipid peroxidation by increasing ather-
oprotective activity of the paraoxonase-1 (PON-1).
Significance: Sardine by-product proteins due to their richness in certain essential amino acids, highlight weight-
loss, lipid-lowering, antioxidant and anti-atherogenic potentials, contributing to the improvement of the com-
plications associated with obesity.

1. Introduction obesity [6]. This may be the consequence of stimulating white fat de-
posits, altering feed intake and increasing pre-adipositary proliferation,
Obesity is one of the most chronic metabolic diseases in the world adipocyte differentiation and mature adipocyte size [7]. In order to
with an increasing prevalence [1]. According to World Health Organi- study the different mechanisms involved in cardiometabolic risk and
zation [2], there are approximately 600 million obese people in the obesity, many experimental studies carried out on rats have used un-
world, including 3.4 million adults who die each year from its com- balanced diets including high-fat, high-caloric or high-carbohydrate
plications. Long-term exposure to an unbalanced high-fat, high-energy diets [8–11].
and micronutrient-poor diet associated with a sedentary lifestyle, in- In humans, several anti-obesity drugs have been approved to im-
volves abnormal or excessive body fat accumulation with a defective prove weight loss; however their potential toxicity may limit their long
energy balance regulation system [3], and causes development of term use [12]. Besides that, prevention and treatment of obesity is es-
obesity and emergence of many cardiovascular risk (CVR) factors which sentially based on improving lifestyle behaviors including a regular
are associated [4]. Hyperlipidemia, which is characterized by high TC physical activity combined with good dietary habits, which are char-
and TG and decreased HDL-C levels, is an independent CVR factor acterized by consumption of marine products, source of high nutritional
which is also considered to be responsible for atherogenicity of obesity value protein, minerals, trace elements, vitamins and lipids, in parti-
[5]. In addition, oxidative stress resulting from an imbalance between cular omega-3 polyunsaturated fatty acids (PUFA n-3) thus giving fish
oxygenated reactive species and antioxidant defense is induced also by their cardioprotective effects [13]. Therefore, developing nutraceutical


Corresponding author.
E-mail address: mylamri@hotmail.fr (M. Lamri-Senhadji).

https://doi.org/10.1016/j.lfs.2018.03.001
Received 11 November 2017; Received in revised form 26 February 2018; Accepted 1 March 2018
Available online 02 March 2018
0024-3205/ © 2018 Elsevier Inc. All rights reserved.
F. Affane et al. Life Sciences 199 (2018) 16–22

foods from natural sources, such as marine products, may be more at- Table 1
tractive to counter the adverse effects of this disease. Composition of the experimental dietsa (g·kg−1).
Currently the mass recovery methods of marine products are or-
Ingredients SBy-P SF-P Cas
iented primarily to production of oils, proteins and their hydrolysates
from fish muscle with hypoglycemic, hypolipidemic, antioxidant and Caseinb – – 200
cardioprotective properties [11,14]. However, fish by-products may Sardine fillets proteinc – 200 –
Sardine by-products proteinc 200 – –
represent a better alternative to deal with global demand for marine
Corn starchd 450 450 450
products that cease to grow and meet the population needs [15]. They Sucrosee 40 40 40
are considered as non-consumable parts (head, skin, viscera, tail, fins, Mutton fatf 200 200 200
crests and falls) but recoverable and usable after treatment and con- Celluloseg 50 50 50
stitute > 60% of the whole product, while only 40% of marine products Mineral mixh 40 40 40
Vitamin mixi 20 20 20
are intended for human consumption [16]. If these materials are al-
ready valued in part for animal feed or aquaculture, they can find after a
Diets are hyperlipidic (19.08 MJ/Kg of diet) and given in powdered form.
transformation more rewarding opportunities [15]. Indeed, these sub- b
Prolabo, Fontenay sous bois, France.
stances also contain numerous valuable molecules, including vitamins c
By-products and fillets proteins purified from sardine in our laboratory.
d
and minerals (hydroxyapatite) which are incorporated into fertilisers ONAB (Sidi Bel Abbes, Algeria).
e
[17], flours with anti-atherogenic properties [18], oils rich in PUFA n-3 f
Cevital (SPA, Bejaia, Algeria).
Sheep fat was obtained from a local market.
which can be used for the prevention of cardiovascular disease (CVD) g
Prolabo Paris, France.
[8], as well as proteins of high nutritional value, source of essential h
UAR 205 B (Villemoisson, 91360, Epinay/S/Orge, France), mineral mix (mg/kg of
amino acids (AA) and bioactive peptides [19–22], intended for devel- diet) CaHPO4, 17200; KCl, 4000; NaCl, 4000; MgO2, 420; MgSO4, 2000; Fe2O3, 120;
opment of new substances that can be used in dietetics, nutraceuticals, FeSO4, 7H2O, 200; MnSO4, H2SO4, H2O, 98; CuSO4, 5H2O, 20; ZnSO4, 7H2O 80; CuSO4
pharmaceuticals and therapeutics such anti-diabetic [23], antioxidants 7H2O; KI, 0,32.
i
UAR 200 (Villemoisson 91360, Epinay/S/Orge, France), vitaminic mixture (mg/kg
and anti-hypertensive products [10,20,22]. In contrast, very few studies
diet): vit A, 39600 UI; vit D3, 5000UI, vit B1, 40; vit B2, 30; vit B3, 140; vit B6, 20; vit B7,
are related in the literature on hypolepidemic, anti-obesity, anti- 300; vit B12, 0,1; vit C, 1600; vit E, 340; vit K, 3,80; vit PP, 200; choline, 2720; folic acid,
atherogenic and cardioprotective effects of proteins and peptides de- 10; para-aminobenzoїc acid, 180; biotine, 0,6; cellulose, qst, 20 g.
rived from different tissues of fish by-products [24]. In this context, the
aim of this study was to determine the effect of protein isolated from and sardine proteins was determined by the Nessler method after mi-
sardine by-product compared to those obtained from the edible portion neralization [27], and their purity was estimated by multiplying total
(fillet), and with casein, on growth parameters, serum leptin level, li- nitrogen content by the factor 6.25. The crude fat content was mea-
pids disorders, reverse cholesterol transport and lipid peroxidation by sured by Folch et al. [28] method. The moisture content was estimated
measuring the activity of certain enzymes involved in their mechanisms as the loss in weight drying at 105 °C for 24 h [29]. The ash amount was
namely, LCAT and PON-1 in high-fat diet induced obese rats. analyzed by incineration at 550 °C for 6 h [29]. The AA composition
was determined by high pressure liquid chromatography (HPLC) [30].
2. Materials and methods

2.3. Animals and diets


2.1. Sardine proteins purification process

Male Wistar rats (Pasteur Institute, Algiers, Algeria) (n = 18),


Fresh sardine (Sardina pilchardus) was purchased from a local public
weighing at the beginning of the experiment, 120 ± 10 g were sub-
fish market (Oran, Algeria), and proteins were isolated from by-pro-
jected for three months to a high-fat diet (20% of sheep fat). At
ducts and fillets according to Guillaume et al. [25] method. By-products
400 ± 20 g, obese rats were divided into three homogeneous groups
(viscera, heads, skins and edges) were separated from the fillet and then
and continue to consume the high-fat diet for 28 days containing either,
washed, kneaded and placed in an oven (Tan Steril, Italy) at 80–85 °C
20% SBy-P, SF-P or Cas. The SF-P and Cas are taken as controls. The
during 20 min. Has this step, a first separation takes place between a
diets composition is shown in Table 1. Food and water were given ad
solid phase (coagulated proteins) and a liquid phase (water and oil) and
libitum. The animals were placed in metabolic cages under standard
which is completed by a pressing. The press water was decanted and
environmental conditions (temperature of 24 °C, a humidity of 60% and
centrifuged (Eppendorf, Centrifuge 5702, Germany), the proteins in the
a 12–hours light/dark cycle). Advice for the protection and use of la-
pellet were re-incorporated into the solid phase then dried in an oven
boratory animals were followed [31]. The experimental protocol was
(40–45 °C) for 24 h, then ground into flour. To remove the lipids and
approved by the Animal Care and Use Committee of Oran 1 University
other organic solvent-soluble elements, sardine by-products and fillets
and the Algerian Ministry of Higher Education and Scientific Research.
flours were purified with n-hexane (Biochem chemopharma, UK) in oil
Rats were weighed weekly, while the food intake was recorded daily.
extractor (Soxhlet Labo-Tech LT.6, Germany) during 5 h at 50 °C. The
The food efficiency ratio (FER), used to express the animal's ability to
delipidated flour was purified by isoelectric precipitation according to
transform calories ingested into body mass [32], was calculated ac-
Undeland et al. [26] method as follow. Delipidated sardine flour was
cording to formula: FER = [body weight gain (g) / food intake
mixed with cold distilled water (1/10 w/v) containing Na2SO3 to pre-
(g)] × 100. Organs relative weight (RW) was determined referring to
serve sulfur proteins, pH was adjusted to 10.8 with NaOH (2 N). After
the equation given below: Organs RW = [organ weight (g) / BW
an overnight incubation at 4 °C, the mixture was centrifuged at
(g)] × 100.
3000 rpm for 20 min at 4 °C. Supernatant containing proteins was pre-
cipitated with H2SO4 at 98% at pH 4.5 then centrifuged at 10,000 rpm
for 30 min at 4 °C. The proteins were washed, freeze dried (Christ, 2.4. Sample collection
Alpha 1-2; LD) and reduced to powder.
At the last week of the experience, feces of each rats group were
2.2. Chemical and AA composition of casein and sardine proteins collected, dried, weighed then stored at 4 °C. After 28 days of the ex-
periment and overnight fasting, six rats of each group were anesthe-
All measurements were performed in triplicate at the Quality tized by intraperitoneal injection of a 10% chloral solution (3 mL/kg
Control and Compliance Laboratory in Oran (El Feth Etablissement). BW). Blood was collected from the abdominal aorta into tubes and
The crude protein level in casein (Prolabo, Fontenay sous bois, France) serum was obtained by low-speed centrifugation (3000 rpm for 20 min,

17
F. Affane et al. Life Sciences 199 (2018) 16–22

at 4 °C). A portion of fresh serum was used to measure LCAT and PON1 Table 2
activities, 0.1% EDTA-Na2 (w/v) was added to the serum remainder as Chemical and amino acids (AA) composition (%) of sardine by-product proteins (SBy-P),
sardine fillets proteins (SF-P) and casein (Cas).
a curator, for carrying out the remaining dosages. Liver and adipose
tissue were removed immediately, rinsed with cold saline and weighed. SBy-P SF-P Cas
Harvested samples were aliquoted and stored at −70 °C until use.
Chemical composition
Crude protein 93.7 ± 0.15 94.8 ± 0.15 95.9 ± 0.32
2.5. Serum leptin evaluation and lipids analysis
Crude fat 1.8 ± 0.11 1.3 ± 0.02 0.2 ± 0.02
Ash 2.1 ± 0.12 1.9 ± 0.01 2.3 ± 0.02
Serum leptin level was measured by radioimmunoassay using a rat Moisture 2.3 ± 0.39 2.0 ± 0.16 1.6 ± 0.27
leptin-specific kit from Assay Designs Inc. (MI, USA). Liver and fecal AA composition
lipids were extracted following Folch et al. [28] method. After total Alanine 7.0 ± 0.05 8.3 ± 0.05 2.9 ± 0.17
lipids determination, the lipids extract was redisolved in isopropanol Arginine 9.1 ± 0.05 6.4 ± 0.08 3.4 ± 0.15
for lipids analysis. Lipids digestibility was calculated as the ratio be- Aspartic acid 10.1 ± 0.05 10.9 ± 0.05 6.2 ± 0.10
Aspergillus 8.9 ± 0.01 9.2 ± 0.06 6.6 ± 0.05
tween the absorbed and the ingested lipids using the following equa-
Cystine 1.1 ± 0.02 0.9 ± 0.05 0.2 ± 0.02
tion: Lipids digestibility (%) = [lipids intake − fecal lipids / lipids in- Glutamic acid 9.9 ± 0.04 11.9 ± 0.15 19.8 ± 0.15
take] × 100. Commercial kits were used to assay TC (Biocon, Glycine 5.7 ± 0.05 3.6 ± 0.10 1.7 ± 0.10
Diagnostics, Germany), TG (GPO-POD Spinreact, Spain), unesterified Histidine 2.2 ± 0.05 3.1 ± 0.10 5.2 ± 0.15
cholesterol (UC) (CHOD-PAP Biolabo, France) and phospholipids (PL) Isoleucine 4.6 ± 0.01 5.1 ± 0.05 4.9 ± 0.06
Leucine 8.1 ± 0.01 7.7 ± 0.15 9.2 ± 0.13
(CHO-POD Cypress, Belgium). Lysine 12.1 ± 0.05 10.5 ± 0.05 7.2 ± 0.15
Methionine 2.9 ± 0.05 2.7 ± 0.06 2.4 ± 0.15
2.6. Isolation and characterization of serum lipoproteins Phenylalanine 1.9 ± 0.03 3.2 ± 0.09 4.8 ± 0.05
Proline 2.5 ± 0.04 3.5 ± 0.06 11.5 ± 0.10
Serine 4.1 ± 0.04 4.8 ± 0.10 5.4 ± 0.11
Serum lipoproteins separation was carried out by a selective pre-
Taurine 5.3 ± 0.02 3.0 ± 0.05 1.8 ± 0.05
cipitation method. Serum very low-density lipoprotein (VLDL) and LDL- Threonine 4.4 ± 0.04 4.7 ± 0.04 4.7 ± 0.05
HDL1 were isolated by precipitation using Magnesium chloride and Tyrosine 2.4 ± 0.01 2.4 ± 0.05 5.2 ± 0.15
Phosphotungstate (Sigma Chemical Company, Lyon, France) as de- Valine 5.1 ± 0.01 5.4 ± 0.10 5.9 ± 0.11
scribed by Burstein et al. [33] method. HDL2 and HDL3 were separated Lysine/arginine 1.33 ± 0.01 1.64 ± 0.02 2.1 ± 0.05
Methionine/glycine 0.51 ± 0.01 0.74 ± 0.01 1.4 ± 0.17
by precipitation according to Burstein et al. [34] method using MgCl2
and dextran sulfate (Sigma Chemical Company). TC and TG in fractions, Chemical and AA composition of SBy-P, SF-P and Cas were performed in triplicate at the
UC and PL in HDL subfractions were determined by enzymatic colori- Quality Control and Compliance Laboratory in Oran (El Feth Etablissement).
metric method described previously. The contents of HDL3 in total
apolipoproteins (apo) were estimated by protein analysis according to 2.9. Statistical analysis
the method of Lowry et al. [35]. Esterified cholesterol (EC) levels were
obtained by difference between TC and UC. HDL2-Cholesteryl esters Values were expressed as means ± standard error of mean (S.E.M.)
(CE) amounts were estimated as 1.67 times the HDL2-EC amounts. for six rats per group. Data were analyzed by one-way analysis of
variance (ANOVA) carried out by Statistica (version 5.1, Statsoft Tulsa,
2.7. Determination of lipid peroxidation in serum and lipoproteins OK, USA). After ANOVA analysis, the classification of the means was
performed using Duncan's new multiple range tests [39]. The means
Lipid peroxidation was estimated by measuring thiobarbituric acid with different superscript were considered significantly different
reactive substances (TBARS) concentrations and expressed in terms of (p < 0.05).
malondialdehyde content according to Quintanilha et al. [36] method.
One milliliter of diluted serum or lipoproteins fractions was added to
3. Results
2 mL of thiobarbituric acid solution (final concentration in the mixture,
0.017 mmol/L) containing 0.375% thiobarbituric acid, diluated on a
3.1. Chemical and AA composition of casein and sardine proteins
solvent mixture (15% trichloroacetic acid, 0.25 N chloridric acid and
distilled water to a final volume of 100 mL) plus butylated hydro-
Chemical composition of casein and sardine proteins shown in
xytoluene (concentration, 3.36 mmol/L). The final mixture was in-
Table 2 was approximately similar. Their purity level was: 96% for
cubated for 60 min at 100 °C. After cooling and centrifugation, the
casein, 94% for sardine by-product protein and 95% for the fillet. The
absorbance of the supernatant was measured at 535 nm.
AA composition of sardine proteins, in particular by-product, revealed a
richness in alanine, arginine, aspartic acid, aspergillus, glycine, lysine
2.8. Determination of LCAT and PON-1 enzymatic activities
and taurine, while their glutamic acid, histidine, phenylalanine, proline
and tyrosine levels and their lysine/arginine and methionine/glycine
LCAT activity (enzyme responsible of UC to EC conversion) was
ratios were low compared to casein. However, the levels of branched-
determined on fresh serum by an endogenous method [37]. UC dis-
chain AA, such as valine, leucine, and isoleucine, were nearly identical
appearance was evaluated between two times (4 h) by colorimetric
(Table 2).
enzymatic method (kit CHOD-PAP Biolabo, France). LCAT activity was
expressed in nanomoles of esterified cholesterol/h/mL of serum. It is
calculated according to the following formula: 3.2. Growth parameters, liver and adipose tissue relative weight
LCAT = (UCt0h − UCt4h)/4 h incubation. PON-1 activity was estimated
by Kuo et al. [38] method. This enzyme catalyses the cleavage of phenyl At day 28, a significant reduction of BW gain (−84%, −41%), FI
acetate resulting in phenol formation. To 10 μL of sample was added (−36%, −19%) and FER (−75%, −27%) were observed in SBy-P and
1 mL of 20 mM Tris-HCl buffer solution pH 8, containing 10 mM phenyl SF-P groups, respectively, compared to Cas group (Table 3). Similar,
acetate and 1 mM CaCl2. The arylesterase activity of PON-1 was cal- difference significantly was noted between SBy-P and SF-P for FI
culated relative to the phenol molar absorption coefficient (−21%), BW gain (−73%) and FER (−66%) (Table 3). Furthermore,
(1.310 M−1 cm−1). One unit of activity of this enzyme is equal to final BW, liver and adipose tissue relative RW were reduced by 6%,
1 μmoL of phenol formed/min. 11% and 18% respectively, in SBy-P compared with Cas.

18
F. Affane et al. Life Sciences 199 (2018) 16–22

Table 3 Table 5
Growth parameters and organs relative weight (RW). Serum leptin level, lipids concentrations and distribution of cholesterol and triacylgly-
cerols in lipoproteins fractions.
SBy-P SF-P Cas
SBy-P SF-P Cas
Growth parameters
Initial BW (g) 400 ± 18 399 ± 19 400 ± 20 Serum leptin (ng·mL−1) 3.35 ± 0.36c 4.08 ± 0.24b 4.73 ± 0.30a
Final BW (g) 405 ± 18b 417 ± 15ab 431 ± 17a Serum lipids (mmol·L−1)
BW gain (g/day/rat) 0.17 ± 0.07c 0.64 ± 0.19b 1.10 ± 0.19a Total cholesterol 1.51 ± 0.23b 1.60 ± 0.21b 2.20 ± 0.33a
Food intake (g/day/rat) 16.13 ± 1.59c 20.49 ± 1.73b 25.27 ± 1.49a VLDL-C 0.39 ± 0,07b 0.46 ± 0.06b 1.03 ± 0.15a
FER (%) 1.07 ± 0.58c 3.16 ± 0.40b 4.35 ± 0.61a LDL-HDL1-C 0.25 ± 0.04c 0.33 ± 0.04b 0.59 ± 0.08a
Organs RW (%) HDL2-C 0.53 ± 0.08a 0.49 ± 0.06a 0.33 ± 0.05b
Liver 2.08 ± 0.16b 2.23 ± 0.11ab 2.35 ± 0.19a HDL3-C 0.33 ± 0.04a 0.30 ± 0.04a 0.24 ± 0.03b
Adipose tissue 2.58 ± 0.18b 2.87 ± 0.40ab 3.14 ± 0.26a Triacylglycerols 0.84 ± 0.06b 0.88 ± 0.05b 1.05 ± 0.09a
VLDL-TG 0.30 ± 0.03b 0.35 ± 0.05b 0.51 ± 0.04a
Data are shown as the mean ± S.E.M. for six values per group. The three groups fed an Liver lipids (μmol·g−1)
obesogenic diet containing 20% sardine by-products proteins (SBy-P), sardine fillets Total cholesterol 19.79 ± 2.02c 25.16 ± 2.53b 37.63 ± 2.16a
proteins (SF-P) or casein (Cas). Statistical analysis was performed using the Duncan's Triacylglycerols 23.52 ± 2.61b 26.47 ± 4.16b 37.13 ± 3.59a
multiple range tests. The means with different superscript were considered significantly
different (p < 0.05). Data are shown as the mean ± S.E.M. for six values per group. The three groups fed an
obesogenic diet containing 20% sardine by-products proteins (SBy-P), sardine fillets
proteins (SF-P) or casein (Cas). Statistical analysis was performed using the Duncan's
3.3. Lipids intake, fecal lipids and lipids digestibility
multiple range tests. The means with different superscript were considered significantly
different (p < 0.05).
At the end of the experiment, lipids intake values were, lower in
SBy-P (−36%) and SF-P (−19%) groups compared to Cas group, with a respectively, by an increase in HDL3-apo (+49%, +19%) and HDL2-CE
significant reductions in SBy-P (−21%) compared to SF-P (Table 4). (+108%, +88%) and a decrease in HDL3-PL (−48%, −43%) and
Furthermore, an increase in fecal lipids and fecal cholesterol were HDL3-UC (−35%, −26%), Furthermore, LCAT activity and HDL3-apo
noted, respectively, between SBy-P (+35%, +47%) and SF-P groups were respectively, +16% and +26% higher in rats fed SBy-P diet
(+21%, +27%) compared with Cas group. Moreover, the values noted compared to SF-P group (Table 6).
in SBy-P were +13% and +15% higher compared to SF-P group.
Whereas there was no significant difference in lipid digestibility be-
3.6. PON-1 activity, serum and lipoprotein lipid peroxidation
tween the three groups (Table 4).

The results of lipid peroxidation represented in Table 7, showed that


3.4. Serum leptin, lipoproteins and liver lipids concentrations serum, LDL-HDL1, HDL2 and HDL3 TBARS levels were, respectively,
reduced by 34%, 54%, 64% and 63% in SBy-P group, and by 20%, 33%,
Serum leptin and LDL-HDL1-C levels were, respectively, lower in 39% and 32% in SF-P group, compared to Cas group. Moreover, the
SBy-P (−29%, −56%) and SF-P (−14%, −43%) groups compared values of TBARS noted in SBy-P, were respectively, reduced on serum
with Cas group (Table 5). Moreover, the values noted in the SBy-P were (−18%), LDL-HDL1 (−31%), HDL2 (−40%) and HDL3 (−46%) com-
−18% and −23% lower than SF-P group. A significant reduction in pared with SF-P group (Table 7). Serum PON-1 activity was increased in
serum TC (−31%, −27%), VLDL-C (−62%, −55%), TG (−19%, rats fed SBy-P (2.2-fold) and SF-P (1.7-fold) diets compared to Cas.
−16%), VLDL-TG (−41%, −31%) and liver TG (−37%, −29%) Likewise, this activity was also enhanced in HDL2 (2- and 1.5-fold) and
concentrations, were observed in SBy-P and SF-P groups, respectively, HDL3 (1.9- and 1.5-fold). Nevertheless, these increases were more
compared to Cas values (Table 5). Liver TC levels were lower in SBy-P marked in SBy-P group (1.3-fold in HDL2 and 1.3-fold in HDL3) com-
group (−47%) and SF-P group (−33%) compared with Cas group pared to SF-P group (Table 7).
(Table 5). Moreover, the value noted in SBy-P was −21% lower than at
SF-P rats. However, an increase in HDL2-C (+62%, +50%) and HDL3-C
4. Discussion
contents (+35%, +25%), was noted respectively, in SBy-P and SF-P
groups compared with the Cas group.
The purpose of the present work was to investigate whether proteins
isolated from sardine by-products, might reduce growth parameters and
3.5. LCAT activity, HDL3-UC, -PL, -apo and HDL2-CE serum leptin level, improve lipids metabolism and attenuate serum and
lipoproteins oxidation in obese rats.
Reverse cholesterol transport exploration has shown that the LCAT Rats, in response to a high-fat diet develop obesity as characterized
activity was increased by 57% and 35%, respectively, in SBy-P and SF-P by weight gain, reduced satiety and increased blood lipids disorders
groups compared with Cas group. This augmentation was accompanied, [4,8]. Furthermore, this type of diet stimulates visceral lipogenesis,

Table 4 Table 6
Lipids intake, fecal lipids and lipids digestibility. Serum LCAT activity and HDL3-UC, HDL3-PL, HDL3-apo and HDL2-CE contents.

SBy-P SF-P Cas SBy-P SF-P Cas

c b a −1 −1 a b
Lipids intake (mg/day/rat) 3226 ± 319 4098 ± 347 5055 ± 298 LCAT (nmol·h ·mL ) 19.34 ± 1.26 16.63 ± 1.24 12.35 ± 1.80c
Fecal lipids HDL3-UC (mmol·L−1) 0.05 ± 0.01b 0.06 ± 0.01b 0.08 ± 0.008a
Fecal total lipids (mg/day/rat) 76.8 ± 4.0a 68.5 ± 3.5b 56.9 ± 6.4c HDL3-PL (mmol·L−1) 0.48 ± 0.11b 0.53 ± 0.09b 0.93 ± 0.18a
Fecal CT (mg/day/rat) 6.87 ± 0.33a 5.97 ± 0.45b 4.68 ± 0.61c HDL3-apo (g·L−1) 0.77 ± 0.06a 0.61 ± 0.04b 0.51 ± 0.04c
Lipids digestibility (%) 97.6 ± 0.3 98.3 ± 0.2 98.9 ± 0.1 HDL2-CE (mmol·L−1) 0.75 ± 0.12a 0.68 ± 0.10a 0.36 ± 0.08b

Data are shown as the mean ± S.E.M. for six values per group. The three groups fed an Data are shown as the mean ± S.E.M. for six values per group. The three groups fed an
obesogenic diet containing 20% sardine by-products proteins (SBy-P), sardine fillets obesogenic diet containing 20% sardine by-products proteins (SBy-P), sardine fillets
proteins (SF-P) or casein (Cas). Statistical analysis was performed using the Duncan's proteins (SF-P) or casein (Cas). Statistical analysis was performed using the Duncan's
multiple range tests. The means with different superscript were considered significantly multiple range tests. The means with different superscript were considered significantly
different (p < 0.05). different (p < 0.05).

19
F. Affane et al. Life Sciences 199 (2018) 16–22

Table 7 reductase activity and/or an increase of its excretion via its hepatic
Thiobarbituric acid reactive substances (TBARS) and paraoxonase (PON-1) activity in catabolism by cholesterol 7α-hydroxylase [14,46]. Furthermore, the
serum and lipoproteins.
high contents in taurine and glycine in SBy-P and SF-P groups, could be
SBy-P SF-P Cas play an important role in bile acids secretion. Indeed, according to
Liaset et al. [43], the fish promotes the excretion of bile acids by their
Serum and lipoproteins conjugation with taurine and glycine, which increases their affinity for
TBARS
membrane hepatic bile acids transporters. Likewise, the inhibition of
Serum-TBARS 18.61 ± 2.10c 22.65 ± 1.29b 28.33 ± 2.24a
(μmol·mL−1) lipids and cholesterol absorption in gut is also possible. Indeed, Hosomi
VLDL-TBARS (μmol·mL−1) 5.39 ± 0.86 4.53 ± 0.85 6.53 ± 0.94 et al. [14], had demonstrated that fish protein have high binding affi-
LDL-HDL1-TBARS 3.56 ± 0.80c 5.19 ± 1.07b 7.80 ± 1.05a nity to bile acids which inhibits the cholesterol emulsifying power. As
(μmol·mL−1) result, the non-reabsorption of bile acids and cholesterol, leading to an
HDL2-TBARS (μmol·mL−1) 2.77 ± 0.64c 4.65 ± 1.11b 7.64 ± 1.32a
increase in the expression of cholesterol 7α-hydroxylase [46]. Conse-
HDL3-TBARS (μmol·mL−1) 3.12 ± 1.00c 5.76 ± 1.09b 8.48 ± 1.23a
Serum and lipoproteins quently, sardine proteins (by-products and fillet) were characterized by
PON-1 a low lysine/arginine (1.33 and 1.64, respectively) and methionine/
Serum-PON-1 (U·mL−1) 102.3 ± 18.0a 81.3 ± 15.3a 46.8 ± 12.4b glycine (0.51 and 0.74, respectively) ratios compared to casein (2,1 and
HDL2-PON-1 (U·mL−1) 9.4 ± 1.5a 7.3 ± 1.4b 4.7 ± 1.0c
1.4, respectively), that seem to favor a cholesterol-lowering effect
HDL3-PON-1 (U·mL−1) 16.3 ± 1.4a 12.9 ± 1.7b 8.6 ± 1.0c
[14,47], probably by increasing hepatic LDL receptor expression [47].
Data are shown as the mean ± S.E.M. for six values per group. The three groups fed an In hypercholesterolemic rats, Mir et al. [48] found that diet con-
obesogenic diet containing 20% sardine by-products proteins (SBy-P), sardine fillets taining sardine proteins increased HDL-C levels, in particular its con-
proteins (SF-P) or casein (Cas). Statistical analysis was performed using the Duncan's tents in cholesteryl esters (CE) and apo-HDL3, indicating an important
multiple range tests. The means with different superscript were considered significantly LCAT activity; an enzyme responsible for reverse cholesterol transport
different (p < 0.05).
and which plays an important role in HDL maturation [49]. In this
study, decreased PL-HDL3 (LCAT substrate) and UC-HDL3 (acyl group
thereby causing lipid peroxidation [6,9]. Similar observations were acceptor) and increased apo-HDL3 (cofactor-activator) and HDL2-CE
found in our study after three months of a high-fat diet consumption at (LCAT product) in response to both SBy-P and SF-P diets could be in-
20% of sheep fats [4,8], compared to those observed in normal rats timately linked to a higher LCAT activity, which could possibly related
consuming a normocaloric diet [9]. to an increased expression of its apo AI cofactor activator and/or its
Interestingly, one of the first results observed in our study discloses hepatic genes synthesis in the liver [47]. In an early study, we had
that SBy-P and SF-P exhibited a significant decrease in FI, FER and BW shown that sardine by-products flour (with approximately 60% of
gain compared to Cas diet. The observed effects are more prominent protein contents) effectively reduces the pro-atherogenic risk in rats fed
with by-products proteins. Similar results were reported in studies an obesogenic diet by improving lipid profile and reverse cholesterol
targeting decreased food intake and weight loss with fish by-products transport [18].
(collagen peptides) and fillet proteins [10,11]; Their low content of It is well established that visceral fat accumulation is correlated
certain AA such as phenylalanine, tyrosine, histidine and proline or with abnormalities of lipoprotein metabolism by an increased release of
their high content of alanine, arginine, glycine and taurine compared to free fatty acids circulating which are primarily directed to liver [5].
casein could be implicated in these effects [19,23]. Bougatef et al. [40] This mechanism could increase TG liver synthesis and VLDL produc-
reported that rats fed smooth hound muscle proteins hydrolysates, tion, leading to long-term dyslipidemia [5]. In our study, reduction in
obtained by digestive proteases, showed a significant decrease in BW, as hepatic TG and hypotriglyceridemia associated with decreased VLDL-
well as a decrease of FI and explained that by a satietogenic effect of TG in obese rats treated with both sources of sardine proteins were
fish peptides. This results suggested that sardine muscle and especially similar to those observed in rats subjected to a high-fat diet treated with
by-products proteins release more efficiently peptides in digestive tract fish proteins and their hydrolysates [11]. This result could be probably
with satietogenic properties, by stimulating Glucagon-like peptide-1 attributed to the increase in lipoprotein lipase activity by decreasing
(GLP-1) activity [10,41]. The rise in endogenous GLP-1 signaling was VLDL hepatic synthesis enriched on TG and/or accelerated VLDL and
seen as a strategy leading to weight loss [42]. The regulation of body apo B-100 catabolism [47].
weight may also depend on the tissue mass (liver and adipose tissue) Recent data suggest that oxidative stress may be the mechanistic
that is reduced in the SF-P group, and particularly in SBy-P group. This linked between obesity and its complications [6]. Besides that, many
reduction is concomitant with the leptin concentration reduction. Sar- studies reported that LCAT may have a dual role in lipoproteins oxi-
dine proteins could by their hypolipidemic properties, increase the li- dation, allowing dose-reactive action as a potent pro-oxidant during
ver's ability to excrete excess lipids [43] or reduce their accumulation in VLDL oxidation, but as an antioxidant during LDL oxidation which
adipose tissue [10,41,44]. Furthermore, it's known that leptin is a sa- would be in favor to reduce an atherogenic acceleration process related
tiety-inducing hormone and involved in the adipose tissue regulation, to obesity [50]. In this study, we have found that sardine proteins de-
its blood concentration tends to increase during weight gain and de- creased serum and lipoproteins TBARS levels and improved LCAT and
crease after weight loss [45]. As well, SBy-P could mitigate the pro- PON-1 activities, probably by their high taurine content and their
gression of obesity by stimulating fatty acid oxidation through the up- richness in AA with an hypocholesterolemic and antioxidant properties
regulation of AMP-activated protein kinase (AMPK) genes, as well as by [19,22], by inhibiting the production of oxidized LDL [20]. The in-
inhibiting the synthesis of adipogenic and lipogenic enzymes [46]. creased PON-1 activity could also explain the low TBARS levels in
In experimental studies, it has been shown that proteins from dif- serum and lipoproteins observed in our study. This antioxidant enzyme
ferent fish species have cholesterol-lowering properties compared to is closely associated with HDL synthesis secondary to the high LCAT
casein [10,11,14]. In this context, our results showed that sardine activity and prevents LDL oxidation [51].
proteins (by-products and fillet) improve cholesterolemia by decreasing Furthermore, high quality fish proteins hydrolysates prepared from
serum VLDL-C and LDL-HDL1-C and increasing HDL-C levels, with viscera, skin and gelatin by-products, also demonstrated high in vitro
higher fecal fat and cholesterol content in rats fed SBy-P. Though, the antioxidant properties and inhibitory activity against lipid peroxidation
exact mechanisms involved in the cholesterol-lowering effect of fish [20–22], and protect DNA damage caused by hydroxyl radicals [52],
proteins have not been fully identified, many hypotheses could be probably due to their richness in certain bioactive peptides. The anti-
proposed to explain them. An inhibition of endogenous cholesterol oxidant activity of proteins (or their peptides) fish is not limited to a
biosynthesis by decreasing 3-hydroxy-3-methyl-glutaryl-coenzyme A single mechanism. They contain various AA with different antioxidant

20
F. Affane et al. Life Sciences 199 (2018) 16–22

properties. Some are more effective as radical scavengers or lipid per- 3111–3120.
oxidation inhibitors, while others are metal chelators or reducers [18] F. Affane, N. Boukhari Benahmed, S. Louala Daidj, A.N. Munezero, M.Y. Lamri-
Senhadji, Effects of an obesogenic diet enriched in sardine by-products on pro-
[19,20]. atherogenic markers in Wistar rats, Ann. Cardiol. Angeiol. 65 (2016) 214–218.
[19] M. Chalamaiah, B. Dinesh kumar, R. Hemalatha, T. Jyothirmayi, Fish protein hy-
5. Conclusion drolysates: proximate composition, amino acid composition, antioxidant activities
and applications: a review, Food Chem. 135 (2012) 3020–3038.
[20] S.M. Halldorsdottir, H. Sveinsdottir, A. Gudmundsdottir, G. Thorkelsson,
Sardine proteins and in particular those of by-products because of H.G. Kristinsson, High quality fish protein hydrolysates prepared from by-product
their richness in certain essential amino acids, favored the weight body material with Fucus vesiculosus extract, J. Funct. Foods 9 (2014) 10–17.
[21] M. Nikoo, S. Benjakul, Potential application of seafood-derived peptides as bi-
loss by their satiating capacity, and ameliorated lipid profile by de- functional ingredients, antioxidant cryoprotectant: a review, J. Funct. Foods 19
creasing the feedback inhibition of cholesterol biosynthesis and/or in- (2015) 753–764.
creasing bile acid excretion. Sardine proteins improved reverse cho- [22] O. Villamil, H. Váquiro, J.F. Solanilla, Fish viscera protein hydrolysates: production,
potential applications and functional and bioactive properties: a review, Food
lesterol transport by increasing LCAT activity, leading to serum
Chem. 224 (2017) 160–171.
cholesterol regulation. This anti-atherogenic action is particularly fa- [23] P.A. Harnedy, V. Parthsarathy, C.M. McLaughlin, M.B. O'Keeffe, P.J. Allsopp
vored by increasing PON-1 activity which prevents lipoproteins from E.M. McSorley, F.P.M. O'Harte, R.J. FitzGerald, Atlantic salmon (Salmo salar) co-
oxidation, thus reducing the complications related to obesity. product-derived protein hydrolysates: a source of antidiabetic peptides, Food Res.
Int. 19 (2018) 598–606.
[24] N.R.A. Halim, H.M. Yusof, N.M. Sarbon, Functional and bioactive properties of fish
Conflict of interest statement protein hydolysates and peptides: a comprehensive review, Trends Food Sci.
Technol. 51 (2016) 24–33.
[25] J. Guillaume, S. Kaushik, P. Bergot, R. Méttailler, Fabrication des farines de poisson
The authors declare that there are no conflicts of interest. et contrôle de qualité: Principales matières premières d'origine animale, in: INRA
(Ed.), Nutrition et alimentation des poissons et crustacés, 1999, pp. 346–351.
Acknowledgements [26] I. Undeland, S.D. Kelleher, H.O. Hultin, Recovery of functional proteins from her-
ring (Clupea harengus) light muscle by an acid or alkaline solubilization process, J.
Agric. Food Chem. 50 (2002) 7371–7379.
This work was funded by the Ministry of Higher Education and [27] M.C. Michel, Application de la méthode de Nessler au microdosage de l'azote alpha-
Scientific Research- Algeria (CNEPRU number D01N01UN310120150023). aminé en milieu Biologique, Ann. Biol. Anim. Biochim. Biophys. 1 (1961) 449–456.
[28] J. Folch, M. Lees, G.H. Sloane-Stanley, A simple method for isolation and pur-
ification of total lipids from animal tissues, J. Biol. Chem. 226 (1957) 497–499.
References [29] C.L. Audigie, J. Figarelle, Z.F. Zons, Manipulation d'analyses biochimiques,
Éditions. Doin. Paris, (1980), pp. 88–97.
[30] E. Soufleros, A. Bertrand, Evaluation of an HPLC method adapted for the de-
[1] C.J. Lavie, A. De Schutter, P. Parto, E. Jahangir, P. Kokkinos, F.B. Ortega, R. Arena,
termination of amino acids in wine, Vitis 37 (1998) 43–53.
R.V. Milani, Obesity and prevalence of cardiovascular diseases and prognosis-the
[31] Council of European Communities, Council instructions about the protection of
obesity paradox updated, Prog. Cardiovasc. Dis. 58 (2016) 537–547.
living animals used in scientific investigations, Off. J. Eur. Communities 358 (1986)
[2] WHO: World Health Organization, BMI Classification, Available from, 2015. http://
1–28.
www.who.int/mediacentre/factsheets/fs311/en/, .
[32] M.T. De Sibio, R.A. Luvizotto, R.M. Olimpio, C.R. Corrêa, J. Marino, M. de Oliveira,
[3] J.O. Hill, H.R. Wyatt, J.C. Peters, Energy balance and obesity, Circulation 126
S.J. Conde, A.L. Ferreira, C.R. Padovani, C.R. Nogueira, A comparative genotoxicity
(2012) 126–132.
study of a supraphysiological dose of triiodothyronine (T3) in obese rats subjected
[4] L. Huang, J. Chen, P. Cao, H. Pan, C. Ding, T. Xiao, P. Zhang, J. Guo, Z. Su, Anti-
to either calorie-restricted diet or hyperthyroidism, 8 (2013) 1–9.
obese effect of glucosamine and chitosan oligosaccharide in high-fat diet-induced
[33] M. Burstein, H.R. Scholnick, R. Morfin, Rapid method for the isolation of lipopro-
obese rats, Mar. Drugs 13 (2015) 2732–2756.
teins from human serum by precipitation with polyanions, J. Lipid Res. 11 (1970)
[5] B. Klop, J.W.F. Elte, M.C. Cabezas, Dyslipidemia in obesity: mechanisms and po-
583–595.
tential targets, Nutrients 5 (2013) 1218–1240.
[34] M. Burstein, A. Fine, V. Atger, E. Wirbel, A. Girard-Globa, Rapid method for iso-
[6] F. Santilli, M.T. Guagnano, N. Vazzana, S. La Barb, G. Davi, Oxidative stress drivers
lation of two purified subfractions of high density lipoproteins by differential
and modulators in obesity and cardiovascular disease: from biomarkers to ther-
dextran sulfate–magnesium chloride precipitation, Biochemistry 71 (1989)
apeutic approach, Curr. Med. Chem. 22 (2015) 582–595.
741–746.
[7] M. Higuchi, G.J. Dusting, H. Peshavariya, F. Jiang, S.T. Hsiao, E.C. Chan, G.S. Liu,
[35] O.H. Lowry, N.J. Rosebrough, A.L. Farr, R.J. Randall, Protein measurement with
Differentiation of human adipose-derived stem cells into fat involves reactive
folin phenol reagent, J. Biol. Chem. 193 (1951) 265–275.
oxygen species and Forkhead box O1 mediated upregulation of antioxidant en-
[36] A.T. Quintanilha, L. Packer, J.M. Szyszlo, T.L. Racanelly, K.J. Davies, Membrane
zymes, Stem Cells Dev. 22 (2013) 878–888.
effects of vitamine E deficiency bioenergetic and surface charge density studies of
[8] N. Boukhari, D. Taleb-Senouci, F.Z. Chabane, M. Besbes, M.Y. Lamri-Senhadji, Fish
skeletal muscle and liver mitochondria, Ann. N. Y. Acad. Sci. 393 (1982) 32–37.
by-products oil corrects dyslipidemia, improves reverse cholesterol transport and
[37] J.J. Albers, C.H. Chen, A.G. Lacko, Isolation, characterization, and assay of le-
stimulates paraoxonase-1 activity in obese rat, Ann. Cardiol. Angeiol. 62 (2013)
cithin–cholesterol acyltransferase, Methods Enzymol. 129 (1986) 763–783.
149–154.
[38] C.L. Kuo, B.N. La Du, Comparison of purified and rabbit serum paraoxonases, Drug
[9] S. Louala, A. Benyahia-Mostefaoui, M.Y. Lamri-Senhadji, La restriction énergétique
Metab. Dispos. 23 (1995) 935–944.
réduit le stress oxydant de l'aorte et du cœur et corrige le risque athérogène chez le
[39] D.B. Duncan, Multiple range and multiple range F-tests, Biometrics 11 (1955)
rat rendu obèse, Ann. Cardiol. Angeiol. 62 (2013) 155–160.
41–42.
[10] E.J. Lee, J. Hur, S.A. Ham, Y. Jo, S. Lee, M.J. Choi, H.G. Seo, Fish collagen peptide
[40] A. Bougatef, R. Ravallec, N. Nedjar-Arroume, A. Barkia, D. Guillochon, M. Nasri,
inhibits the adipogenic differentiation of preadipocytes and ameliorates obesity in
Evidence of in vivo satietogen effect and control of food intake of smooth hound
high fat diet-fed mice, Int. J. Biol. Macromol. 104 (2017) 281–286.
(Mustelus mustelus) muscle protein hydrolysate in rats, J. Funct. Foods 2 (2010)
[11] I. Jemil, O. Abdelhedi, R. Nasri, L. Mora, R. Marrekchi, K. Jamoussi, A. El Feki,
10–16.
M. Hajji, F. Toldra, M. Nasri, Hypolipidemic, antiobesity and cardioprotective ef-
[41] Z. Madani, K. Louchami, A. Sener, W.J. Malaisse, D. Ait Yahia, Dietary sardine
fects of fermented protein hydrolysates from sardinelle (Sardinella aurita) in high-fat
protein lowers insulin resistance, leptin and TNF-α and beneficially affects adipose
and fructose diet fed Wistar rats, Life Sci. 176 (2017) 54–66.
tissue oxidative stress in rats with fructose-induced metabolic syndrome, Int. J. Mol.
[12] A.J. Krentz, A.K. Fujioka, M. Hompesch, Evolution of pharmacological obesity
Med. 29 (2012) 311–318.
treatments: focus on adverse side-effect profiles, Diabetes Obes. Metab. 18 (2016)
[42] C.R. Hutch, D. Sandoval, The role of GLP-1 in the metabolic success of bariatric
558–570.
surgery, Endocrinology 158 (2017) 4139–4151.
[13] R. Larsen, K.E. Eilertsen, E.O. Elvevoll, Health benefits of marine foods and in-
[43] B. Liaset, L. Madsen, Q. Hao, G. Crials, G. Mellgren, H.U. Marschall, P. Hallenborg,
gredients, Biotechnol. Adv. 29 (2011) 508–518.
M. Espe, L. Frøyland, K. Kristiansen, Fish protein hydrolysate elevates plasma bile
[14] R. Hosomi, K. Fukunaga, H. Arai, T. Nisniyama, M. Yoshida, Effect of combination
acids and reduces visceral adipose tissue mass in rats, Biochim. Biophys. Acta Mol.
of dietary fish protein and fish oil on lipid metabolism in rats, J. Food Sci. Technol.
Cell Biol. Lipids 1791 (2009) 254–262.
50 (2013) 266–274.
[44] G. Pilon, J. Ruzzin, L.E. Rioux, C. Lavigne, P.J. White, L. Frøyland, H. Jacques,
[15] FAO, The State of World Fisheries and Aquaculture. FAO Fisheries and Aquaculture
P. Bryl, L. Beaulieu, A. Marette, Differential effects of various fish proteins in al-
Department. Food and Agriculture Organization of the United Nations. Contributing
tering body weight, adiposity, inflammatory status, and insulin sensitivity in high-
to Food Security and Nutrition for All. Rome, 200pp. http://www.fao.org, .
fat–fed rats, Metabolism 60 (2011) 1122–1130.
[16] E. Dekkers, S. Raghavan, H.G. Kristinsson, M.R. Marshall, Oxidative stability of
[45] K.W. Williams, M.M. Scott, J.K. Elmquist, From observation to experimentation:
mahimahi red muscle dipped in tilapia protein hydrolysates, Food Chem. 124
leptin action in the mediobasal hypothalamus, Am. J. Clin. Nutr. 89 (2009)
(2011) 640–645.
985S–990S.
[17] V. Ferraro, A.P. Carvalho, C. Piccirillo, M.M. Santos, P.M.L. Castro, M.E. Pintado,
[46] R. Hosomi, K. Fukunaga, H. Arai, T. Nisniyama, M. Yoshida, Effects of dietary fish
Extraction of high added value biological compounds from sardine, sardine-type
protein on serum and liver lipid concentrations in rats and the expression of hepatic
fish and mackerel canning residues: a review, Mater. Sci. Eng. 33 (2013)
genes involved in lipid metabolism, J. Agric. Food Chem. 57 (2009) 9256–9262.

21
F. Affane et al. Life Sciences 199 (2018) 16–22

[47] A. Shukla, A. Bettzieche, F. Hirche, C. Brandsch, G.I. Stangl, K. Eder, Dietary fish (2015) 1–9.
protein alters blood lipid concentrations and hepatic genes involved in cholesterol [50] P.A. McPherson, I.S. Young, J. McEneny, A dual role for lecithin: cholesterol
homeostasis in the rat model, Br. J. Nutr. 96 (2006) 674–682. acyltransferase (EC 2.3.1.43) in lipoprotein oxidation, Free Radic. Biol. Med. 43
[48] H.Mir.D. Krouf, D. Taleb-Senouci, N. Taleb-Dida, Combined treatment with sardine (2007) 1484–1493.
proteins and Citrus latifolia extract corrects dyslipidemia, prevents lipid peroxida- [51] H. Soran, J.D. Schofild, P.N. Durrington, Antioxidant properties of HDL, Front.
tion and improves lecithin: cholesterol acyltransferase and paraoxonase 1 activities Pharmacol. 6 (2015) 1–6.
in hypercholesterolemic rats, Br. J. Appl. Sci. Technol. 7 (2015) 156–167. [52] R.A. Nazeer, N.S.S. Kumar, Purification and identification of antioxidant peptide
[49] M.C. Sprandel, W.A. Hueb, A. Segre, J.A. Ramires, R. Kalil-Filho, R.C. Maranhão, from black pomfret, Parastromateus niger (Bloch, 1975) viscera proteinhydrolysate,
Alterations in lipid transfers to HDL associated with the presence of coronary artery Food Sci. Biotechnol. 20 (2011) 1087–1094.
disease in patients with type 2 diabetes mellitus, Cardiovasc. Diabetol. 14

22

You might also like