Astaxanthin-antioxidant impact on excessive Reactive ... · ture of bile acids, phospholipids,...

14
Contents lists available at ScienceDirect Chemico-Biological Interactions journal homepage: www.elsevier.com/locate/chembioint Astaxanthin-antioxidant impact on excessive Reactive Oxygen Species generation induced by ischemia and reperfusion injury M. Zuluaga, V. Gueguen, D. Letourneur, G. Pavon-Djavid INSERM U1148, Laboratory for Vascular Translational Science, Cardiovascular Bioengineering, Paris 13 University, Sorbonne Paris Cite 99, Av. Jean-Baptiste Clément, 93430 Villetaneuse, France ARTICLE INFO Keywords: Astaxanthin Drug delivery Oxidative stress Ischemia and reperfusion ROS ABSTRACT Oxidative stress induced by Reactive Oxygen Species (ROS) was shown to be involved in the pathogenesis of chronic diseases such as cardiovascular pathologies. Particularly, oxidative stress has proved to mediate ab- normal platelet function and dysfunctional endothelium-dependent vasodilatation representing a key factor in the progression of ischemic injuries. Antioxidants like carotenoids have been suggested to contribute in their prevention and treatment. Astaxanthin, a xanthophyll carotenoid produced naturally and synthetically, shows interesting antioxidant and anti-inammatory properties. In vivo studies applying dierent models of induced ischemia and reperfusion (I/R) injury conrm astaxanthin's protective action after oral or intravenous admin- istration. However, some studies have shown some limitations after oral administration such as low stability, bioavailability and bioecacy, revealing a need for the implementation of new biomaterials to act as astax- anthin vehicles in vivo. Here, a brief overview of the chemical characteristics of astaxanthin, the carrier systems developed for overcoming its delivery drawbacks and the animal studies showing its potential eect to treat I/R injury are presented. 1. Introduction Reactive oxygen species (ROS) refers to a variety of highly reactive molecules and free radicals derived from molecular oxygen. ROS are formed as a normal byproducts of aerobic respiration and current cel- lular metabolism [1]. Moderate amounts of ROS have benecial eects on several physiological processes like the reduction of malignant pa- thogens, wound healing, and tissue repair processes by acting as sig- naling molecules [24]. In contrast, ROS overproduction disrupts the body homeostasis inducing oxidative tissue damage [5]. Indeed, high ROS levels leads to decreased bioavailability of nitric oxide, impairing endothelium-dependent vasodilatation thus promoting vasoconstriction [6]. These alterations occur early in the development of vascular dis- ease [7]. Moreover, overproduction of superoxide anion radical and hydroxyl radical have been considered causative agents of severe dis- eases, such as arteriosclerosis and I/R injury [810], pathologies cur- rently linked to increased rates of lipids peroxidation [8,11]. In the cell, these reactions are counteracted by the action of enzy- matic and non-enzymatic antioxidant defenses. Tissue damage takes place when these antioxidant defenses are not sucient to control the radicals generation [12]. Recent studies suggest the use of exogenous antioxidant supplementation with carotenoids would enhance antioxidant defenses thanks to their potential scavenging capabilities [1317]. Astaxanthin carotenoid is known to be a potent quencher of singlet oxygen and an ecient scavenger of superoxide anion [18], and hydroxyl radical [19,20] by acting as an antioxidant. Moreover, within the cell, it can eectively scavenge lipid radicals and eectively de- stroys peroxide chain reactions to protect fatty acids and sensitive membranes [21,22] reducing the risk of atherosclerotic plaque forma- tion [23,24]. Furthermore, the astaxanthin eect in the prevention and treatment of I/R pathologies in vivo revels its potent action as anti- oxidant molecule. However, astaxanthin as a highly unsaturated mo- lecule decomposes easily when being exposed to heat, light and oxygen. Additionally, its poor water solubility, stability and bioavailability limits its appropriate oral administration and delivery in vivo. The im- plementation of new biomaterials to act as astaxanthin vectors has been attempted through various strategies. Here, a review of in vivo studies reporting the eect of astaxanthin supplementation to counteract ischemia/reperfusion injury will be presented, including a brief review of astaxanthin carrier's system successfully developed for overcoming delivery challenges. https://doi.org/10.1016/j.cbi.2017.11.012 Received 5 July 2017; Received in revised form 3 November 2017; Accepted 21 November 2017 Corresponding author. E-mail address: [email protected] (G. Pavon-Djavid). Chemico-Biological Interactions 279 (2018) 145–158 Available online 24 November 2017 0009-2797/ © 2017 Elsevier B.V. All rights reserved. T

Transcript of Astaxanthin-antioxidant impact on excessive Reactive ... · ture of bile acids, phospholipids,...

Contents lists available at ScienceDirect

Chemico-Biological Interactions

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

Astaxanthin-antioxidant impact on excessive Reactive Oxygen Speciesgeneration induced by ischemia and reperfusion injury

M. Zuluaga, V. Gueguen, D. Letourneur, G. Pavon-Djavid∗

INSERM U1148, Laboratory for Vascular Translational Science, Cardiovascular Bioengineering, Paris 13 University, Sorbonne Paris Cite 99, Av. Jean-Baptiste Clément,93430 Villetaneuse, France

A R T I C L E I N F O

Keywords:AstaxanthinDrug deliveryOxidative stressIschemia and reperfusionROS

A B S T R A C T

Oxidative stress induced by Reactive Oxygen Species (ROS) was shown to be involved in the pathogenesis ofchronic diseases such as cardiovascular pathologies. Particularly, oxidative stress has proved to mediate ab-normal platelet function and dysfunctional endothelium-dependent vasodilatation representing a key factor inthe progression of ischemic injuries. Antioxidants like carotenoids have been suggested to contribute in theirprevention and treatment. Astaxanthin, a xanthophyll carotenoid produced naturally and synthetically, showsinteresting antioxidant and anti-inflammatory properties. In vivo studies applying different models of inducedischemia and reperfusion (I/R) injury confirm astaxanthin's protective action after oral or intravenous admin-istration. However, some studies have shown some limitations after oral administration such as low stability,bioavailability and bioefficacy, revealing a need for the implementation of new biomaterials to act as astax-anthin vehicles in vivo. Here, a brief overview of the chemical characteristics of astaxanthin, the carrier systemsdeveloped for overcoming its delivery drawbacks and the animal studies showing its potential effect to treat I/Rinjury are presented.

1. Introduction

Reactive oxygen species (ROS) refers to a variety of highly reactivemolecules and free radicals derived from molecular oxygen. ROS areformed as a normal byproducts of aerobic respiration and current cel-lular metabolism [1]. Moderate amounts of ROS have beneficial effectson several physiological processes like the reduction of malignant pa-thogens, wound healing, and tissue repair processes by acting as sig-naling molecules [2–4]. In contrast, ROS overproduction disrupts thebody homeostasis inducing oxidative tissue damage [5]. Indeed, highROS levels leads to decreased bioavailability of nitric oxide, impairingendothelium-dependent vasodilatation thus promoting vasoconstriction[6]. These alterations occur early in the development of vascular dis-ease [7]. Moreover, overproduction of superoxide anion radical andhydroxyl radical have been considered causative agents of severe dis-eases, such as arteriosclerosis and I/R injury [8–10], pathologies cur-rently linked to increased rates of lipids peroxidation [8,11].

In the cell, these reactions are counteracted by the action of enzy-matic and non-enzymatic antioxidant defenses. Tissue damage takesplace when these antioxidant defenses are not sufficient to control theradicals generation [12]. Recent studies suggest the use of exogenousantioxidant supplementation with carotenoids would enhance

antioxidant defenses thanks to their potential scavenging capabilities[13–17]. Astaxanthin carotenoid is known to be a potent quencher ofsinglet oxygen and an efficient scavenger of superoxide anion [18], andhydroxyl radical [19,20] by acting as an antioxidant. Moreover, withinthe cell, it can effectively scavenge lipid radicals and effectively de-stroys peroxide chain reactions to protect fatty acids and sensitivemembranes [21,22] reducing the risk of atherosclerotic plaque forma-tion [23,24]. Furthermore, the astaxanthin effect in the prevention andtreatment of I/R pathologies in vivo revels its potent action as anti-oxidant molecule. However, astaxanthin as a highly unsaturated mo-lecule decomposes easily when being exposed to heat, light and oxygen.Additionally, its poor water solubility, stability and bioavailabilitylimits its appropriate oral administration and delivery in vivo. The im-plementation of new biomaterials to act as astaxanthin vectors has beenattempted through various strategies. Here, a review of in vivo studiesreporting the effect of astaxanthin supplementation to counteractischemia/reperfusion injury will be presented, including a brief reviewof astaxanthin carrier's system successfully developed for overcomingdelivery challenges.

https://doi.org/10.1016/j.cbi.2017.11.012Received 5 July 2017; Received in revised form 3 November 2017; Accepted 21 November 2017

∗ Corresponding author.E-mail address: [email protected] (G. Pavon-Djavid).

Chemico-Biological Interactions 279 (2018) 145–158

Available online 24 November 20170009-2797/ © 2017 Elsevier B.V. All rights reserved.

T

2. Astaxanthin: A powerful antioxidant molecule

2.1. Astaxanthin sources

The carotenoid astaxanthin is found in various microorganisms andmarine animals, such us yeast, microalgae, salmon, krill, shrimp,complex plants and some birds [25–29]. As in general with all car-otenoids, astaxanthin is not synthesized by humans and therefore re-quires to be ingested in the diet, seafood being the main source [30,31].

Haematococcus pluvialis (H. pluvialis), a unicellular biflagellate greenmicroalgae, is believed to have the highest capacity to accumulate as-taxanthin in nature under environmental stresses such as starvation,high salt or pH, elevated temperature, or irradiation [25,32]. Underthese unfavorable conditions, microalgae modify their cellular mor-phology, increasing their size to become red cysts charged with ∼80%of astaxanthin pigment [54] and 20% comprised a mixture of othercarotenoids [33]. Due to the high astaxanthin concentration, micro-algae represent the primary natural source of processed astaxanthin forhuman applications such as dietary supplements, cosmetics, and foodand beverages [34], while the synthetic [35,36], yeast (mutated Xan-thophyllomyces dendrorhous) [37] and bacteria sources (from Paracoccuscarotinifaciens, an aerobic bacteria) [38] are predominantly used in theaquaculture sector [35]. Moreover, dietary supplements containing H.pluvialis astaxanthin have proved to be safe and accepted by theAmerican Food and Drug Administration at daily doses of 2–12 mg perday [39,40].

2.2. Chemical characteristics

Astaxanthin (3,3′-dihydroxy-β,β′-carotene-4,4′-dione) carotenoid isa fat-soluble orange-red color pigment with the molecular formulaC40H52O4 and molar mass of 596.84 g/mol. Astaxanthin structureconsists of 40 carbon atoms which contain two oxygenated β-ionone-type ring systems linked by a chain of conjugated double bonds

(polyene chain). The oxygen presence in astaxanthin ionone rings inboth hydroxyl (OH) and keto (C]O) groups, makes it a member of thexanthophyll carotenoid family and confers to astaxanthin a more polarnature than other carotenoids [41]. Additionally, the conjugated doublebonds allow astaxanthin to act as a strong antioxidant by electron do-nation and by reacting with free radicals [42] (Fig. 1A).

In its free form, astaxanthin is considerably unstable and particu-larly susceptible to oxidation, therefore, this form is mainly producedsynthetically or from yeast [43]. In nature, it is found either conjugatedwith proteins (e.g., salmon muscle or lobster exoskeleton) or esterifiedby hydroxyl reaction with one (monoester) or two (diester) fatty acids,which stabilize the molecule. Natural astaxanthin from H. pluvialiscontains 70–90% of monoesters, about 8% of diester and 2% of freeform [41,44,45] (Fig. 1B–C). A protective role against high light andoxygen radical has been attributed to astaxanthin accumulation in H.pluvialis [33]. The stereogenic carbons in the 3 and 3′ positions on the β-ionone moieties define astaxanthin conformation as chiral [(3S, 3′S) or(3R, 3′R)] or as meso form (3R, 3′S), with the chiral conformation themost abundant in nature [27] (Fig. 1D–E). Astaxanthin from microalgaeH. pluvialis biosynthesizes the (3S, 3′S) isomer whereas yeast produces(3R, 3′R) isomer [41]. The synthetic source consists of isomers (3S, 3′S)(3R, 3′S) and (3R, 3′R) [27].

2.3. Extraction, storage, stability of astaxanthin

When the stress is induced, the microalgae H. pluvialis becomesencysted cells and accumulates high quantities of astaxanthin [46]. Thisgrowth stage is usually produced in either enclosed outdoor systems orclosed indoor photo-bioreactors, which are preferred to avoid con-tamination by other microorganisms and to guarantee optimal andcontrolled growth conditions [36]. Different methods had been carriedout to extract the greatest quantity of the carotenoid from H. pluvialisbiomass by cracking the cell [33]. Some of them are based on the use ofsolvents [47], edible oils [48], enzymatic digestion [49], but

Fig. 1. (A) Structure of free astaxanthin with a numbering scheme in the stereoisomer form 3S, 3′S. Astaxanthin (B) monoester and (C) diester form. (D–E) Astaxanthin stereoisomers3R,3′S and 3R,3′R. Natural H. pluvialis produced astaxanthin 3S,3′S containing 2% free, 90% monoester and 8% of diester, while synthetic astaxanthin exists as free form constituted by3S,3′S, 3R,3′S and 3R, 3′R in a ratio of 1:2:1, respectively [25,43,44].

M. Zuluaga et al. Chemico-Biological Interactions 279 (2018) 145–158

146

supercritical fluid extraction still represents the method most widelyused in the ago-alimentary industry [50–52]. An innovative method forastaxanthin extraction from H. pluvialis using supramolecular solvents(SUPRAS) is currently under development [53]. Once astaxanthin isextracted from the biomass, its stability and storage must be assured toavoid degradation by environmental factors such as temperature, pHand light [26].

2.4. Pharmacokinetics and toxicity of astaxanthin

The low bioavailability of astaxanthin and all xanthophyll car-otenoids after oral administration has been attributed to their poorwater solubility/dispersibility. Particularly, the limited solubility indigestive fluids compromise the uptake of astaxanthin by intestinalepithelial cells and their final secretion to lymph as chylomicrons[30,54]. After ingestion, xanthophyll carotenoids are solubilized in themixed micelles in the small intestine. These micelles represent a mix-ture of bile acids, phospholipids, cholesterol, fatty acids and mono-acylglycerols surrounded by the bile acids [55]. Then carotenoidstransfer from the micelles to the epithelial cells by simple and fa-cilitated diffusion across the phospholipid bilayers of the cytoplasmicmembrane [30]. Once degraded, carotenoids are stored in the liver andre-secreted as very low-density lipoproteins (VLDL), low density lipo-proteins (LDL), and high-density lipoproteins (HDL) reaching a higherlevel of bioavailability, and eventually to be transported to the tissuesvia the circulation [56].

Due to the presence of polar ends in its structure, astaxanthin can beabsorbed better than other non-polar carotenoids, such as lycopene andβ-carotene [57–59]. In the case of esterified astaxanthin, before LDLtransport, esters need to be hydrolyzed by cholesterol esterase [21,22].Coral-Hinostroza et al. [58] showed that after oral administration, as-taxanthin esters are hydrolyzed selectively during absorption, sug-gesting that unesterified astaxanthin may be preferentially absorbed orselectively transported through circulation in human. Additionally,astaxanthin blood levels have been reported as up to 0.19 μmol/L after

1–12 mg human intake for 1 year [60].Conversely, animal studies showed a higher uptake of astaxanthin

diesters than esters after oral administration [61]. H. D Choi et al. [62]evaluated the pharmacokinetics of astaxanthin in rats, reporting theelimination of an important portion of intravenous administered as-taxanthin at doses up to 20 mg/kg via a non-renal route. Moreover, alonger astaxanthin half-life and a hepatic and gastrointestinal first-passextraction ration of 0.490 and 0.901, respectively, were obtained when200 mg/kg of astaxanthin was administered using an oral (1460 min)than intravenous (569 min) pathways. These results could indicate thatbioavailability and a half-life of astaxanthin is influenced by its ester-ification status [34] and suggests that the lipophilic properties of themolecule require the use of additives and surfactants to incorporate itinto carrier systems for use in foods, beverages and pharmaceuticalproducts [63].

The safety of astaxanthin has been assessed in Sprague rats afterreceiving daily oral administration of astaxanthin-rich H. pluvialis bio-mass at concentrations up 500 mg astaxanthin/kg/day for 90 days [64],or synthetic astaxanthin in a range between 880 and 1240 mg/kg bw/day, for 13 weeks, [65]. No adverse effects were reported on the ana-lyzed health-related parameters. The toxicity of synthetic astaxanthinwas also tested in pregnant New Zealand white rabbits at concentra-tions up to 400 mg/kg bw/day without showing harmful effects onreproduction or fetal development [66]. Additionally, Katsumata et al.[67] performed a sub-chronic-toxicity evaluation of a natural astax-anthin-rich carotenoid extract produced from the natural bacteriaParacoccus carotinifaciens suspended in olive oil and administered dailyto rats by oral gavage at doses of up to 1000 mg/kg/day for 13 weeks.The only result highlighted was the excretion of dark-red color feceswithout reporting any considerable adverse effect.

A. Satoh et al. [68] evaluated the human clinical toxicity and effi-cacy of long-term administration of soft capsules containing an oilbased natural astaxanthin-rich product by measuring biochemical andhematological blood parameters and by analyzing brain function. Theparticipants received an astaxanthin concentration up to 20 mg daily

Fig. 2. I/R injury enhance ROS levels and oxidative stress conditions. Adapted from Refs. [15,71,82].

M. Zuluaga et al. Chemico-Biological Interactions 279 (2018) 145–158

147

Table1

Summaryof

seve

ralin

vivo

stud

iesev

alua

ting

theeff

ectof

astaxa

nthintreatm

enton

indu

cedI/Rinjury.

Ref.

Source

Mod

elPa

thway

Doses/

Duration

Effects

ofAstax

anthin

treatm

ent

Gross

and

Look

woo

-d20

04[98]

Disod

ium

Disuc

cina

teAstax

anthin

(from

Dr.

Samue

lF.

Lockwoo

d,Haw

aii

Biotech,

Inc.)

Sterile

DI

Water

Rat

myo

cardial

I/R

I:30

min

R:2

h

Intrav

enou

sinjection

One

of3

doses(25,

50,a

nd75

mg/

kg)for4

days

prior

toI/R

Carda

xat

50an

d75

mg/

kgfor4da

yssign

ificantly

redu

cesinfarctsize

atarea

atrisk

to35

±3%

(41%

salvag

e)an

d26

±2%

(56%

salvag

e),respective

ly.

Gross

and

Look

woo

-d20

05[99]

Dog

myo

cardial

I/R

I:60

min

R:3

h

Intrav

enou

sinjection

50mg/

kg2hor

4da

ysprior

toI/R

Red

uction

ininfarctsizeat

area

atrisk

to11

.0±

1.7%

(47.3%

salvag

e)in

dogs

treatedon

lyon

ceIV

at2hpriorto

occlusion,

and6.6

±2.8%

(68.4%

salvag

e)in

dogs

treatedfor4da

ys.

Lauv

eret

al.,

2005

[100

]

Rab

bit

myo

cardial

I/R

I:30

min

R:3

h

Intrav

enou

sinjection

(1mL/

min)

50mg/

kg/d

ay4 co

nsecu-

tive

sprior

toI/R

Infarctsize

redu

ctionexpressedas

ape

rcen

tage

ofthearea

atrisk

(25.8

±4.7%

)in

theDDA-treated

.Myo

cardialsalvag

eof

51%.

Red

uced

erythroc

ytehe

molysis

indicatedby

high

lyfavo

rablemeanmyo

cardium/serum

ratios

(10.1

±1.6μM

)Red

ucede

position

ofCRPan

dMACan

tibo

dies

intheinfarctregion

Gross

and

Look

woo

-d 20

06[101

]

Rat

myo

cardial

I/R

I:30

min

R:2

h

Oral

administra-

tion

asfeed

supp

lemen

t

0.1an

d0.4%

;∼12

5an

d50

0mg/

kg/d

ay,

respec-

tive

lyfor

seve

nda

ys

Carda

xTM

at0.1an

d0.4%

infeed

for7da

ysresulted

inasign

ificant

meanredu

ctionin

infarctsize

atarea

atrisk

to45

±2.0%

(26%

salvag

e)an

d39

±1.5%

(36%

salvag

e),r

espe

ctively.

Myo

cardialleve

lsof

Carda

xachiev

edafter7-da

ysupp

lemen

tation

ateach

ofthetw

oco

ncen

trations

400

±65

nMan

d16

34±

90nM

,respe

ctively.

Red

uction

ofarachido

nicacid

andlin

oleicacid

(lipid

peroxida

tion

prod

ucts)in

plasmaleve

ls

Adluriet

al.,

2013

[102

]

VitaePro(2%

astaxa

nthin,

8.1%

lutein

and1.23

%zeax

anthin;

VitaeLa

bAS,

Eneb

akkv

eie

Oslo,

Norway

)

Rat

myo

cardial

I/R

I:30

min

R:2

h

Oralga

vage

Dissolved

insafflow

eroil

70mg/

kgbo

dyweigh

tfor

21da

ys

Increasedleft

ventricu

larfunc

tion

alreco

very

afterI/R

Decreaseinfarctsize

(27.68

±1.7)

Decreaseap

optoticcardiomyo

cytes(61.7

±10

.6)measuredby

TUNEL

assay

Decreasethioba

rbituric

acid

reactive

substanc

esleve

ls(80

±3)

Wan

get

al.,

2017

[96]

Astax

anthin

(from

Jian

heBiotechCo.

Ltd.

Jian

he,

Heb

ei,

China

)

Mice

tran

svers

aortic

constriction

Oralga

vage

200mg/

kg/d

ayfor12

days

prior

toTA

C

Mitigationof

TACindu

cedcardiacdy

sfun

ction,

myo

cardialfibrosis

andmyo

cardialdisorder

show

ingthat

SIRT1

participates

intheseprotective

func

tion

sby

attenu

atingR-SMAD

acetylation.

Red

uction

intheexpression

ofproteinan

dtran

script

leve

lsof

TGF-β,

α-SM

Aan

dCOLI.

Luet

al.,20

10[103

]Astax

anthin

(from

Sigm

a-Aldrich

,St.

Louis,

MO,

USA

)

Rat

cerebral

I/R

I:2h

R:2

4h

Oralga

vage

20,5

0,80

mg/

kgintrag

as-

trically

twiceat

5han

d1hprior

to isch

emia

Red

uction

ofinfarctsize

volume14

.6±

5.4%

and11

.4±

4.9%

afterdo

sesof

50an

d80

mg/

kgrespective

lyIm

prov

edne

urolog

ical

deficitto

1.5

±0.7an

d0.9

±0.8afterdo

sesof

50an

d80

mg/

kgrespective

lyNeu

rons

cells

protection

upto

99.5

±12

.6.

Leeet

al.,

2010

[104

]

Rat

cerebral

I/RI:1

0min

Intrap

eriton

-ealinjection

30mg/

kgof

at0an

d90

min

of

Protective

effectof

59.5%

atdo

sesof

30mg/

kgon

CA1hipp

ocam

palne

uron

s.Inhibition

ofpo

ly(A

DP-ribo

se)po

lymerase(PARP-1,

apop

toticmarke

r)cleava

geat

ado

seof

20mg/

kg

(con

tinuedon

next

page)

M. Zuluaga et al. Chemico-Biological Interactions 279 (2018) 145–158

148

Table1(con

tinued)

Ref.

Source

Mod

elPa

thway

Doses/

Duration

Effects

ofAstax

anthin

treatm

ent

R:0

and

90min

Dissolved

inpu

reetha

nol

cerebral

repe

rfu-

sion

.Li

etal.,20

15[105

]Micerena

lI/R

I:60

min

R:2

h,8h

and24

h

Oralga

vage

Dissolved

inoliveoil

30mg/

kgor

60mg/

kg)for14

days

Decreaseof

ALT

andAST

enzymes

inado

se-dep

ende

ntman

ner:

Low

astaxa

nthindo

sesredu

cerena

lne

crotic

area,w

hile

high

dosesincrease

protection

Red

uction

ofinflam

matorycytokine

sleve

ls(TNF-αan

dIL-6)in

serum

andtissue

.Prom

oted

anti-apo

ptotic

Bcl-2

andinhibitedpro-ap

optoticBa

xleve

lsA

downw

ardtren

din

proteinkina

ses(p-P38

MAPK

,p-ER

Kan

dp-JN

K)expression

inliv

ertissue

Curek

etal.,

2010

[106

]

ASX

(from

Sigm

a–Aldrich

Che

mie,

Steinh

eim,

German

y)

Rat

liver

I/R

I:60

min

R:6

0min

Oralga

vage

Dissolved

inoliveoil

5mg/

kg/

day

14da

yspriorto

I/R

Decreased

oxidativestress

byhe

paticco

nversion

ofXDH

toxa

nthine

oxidasean

dtissue

proteincarbon

ylleve

lsPa

rtialredu

ctionof

cellda

mag

e,sw

ellin

gof

mitoc

hond

riaan

ddisarran

gemen

tof

roug

hen

doplasmatic

reticu

lum

Red

uced

ofproteincarbon

ylform

ationbu

tno

sign

ificant

effecton

GSH

andnitrite/nitrateleve

ls

Qiu

etal.,

2015

[107

]

Free

astaxa

nthin

(≥97

%,

Sigm

a-Aldrich

,St.L

ouis,M

O,

USA

)

Micerena

lI/R

I:45

min

R:1

2han

d24

h

Oralga

vage

Dissolved

inoliveoil

5mg/

kg/

day

during

14da

yspriorto

I/R

Preserva

tion

ofrena

lfunc

tion

after12

and24

hpo

stI/Rreflectedby

redu

cedbloo

dserum

urea

nitrog

enan

durinecreatinine

leve

ls.

Decreaseof

apop

toticcells

andα-sm

ooth

muscleactinexpression

assessed

byTU

NEL

assay

Decreaseexpression

sof

TNF-α,

IL-1β,

andIL-6

inflam

matoryproteins

Red

uceox

idativestress

reflectedby

sign

ificantly

increasedof

supe

roxide

dism

utaseleve

lan

dde

creasedleve

lof

malon

dialde

hyde

Hussein

etal.,

2005

[24]

ASX

-O,

compo

sedof

5.5%

astaxa

nthin

(FujiChe

mical

Indu

stry

Co.,

Ltd.,T

oyam

a,Japa

n)

Mice

cerebral

isch

emia

I:20

min

Oralga

vage

Dissolved

inan

edible

oil

base

ASX

-Oat

5,50

and

500mg/

kg Sing

leor

daily

dose

for2

weeks

ASX

-Oshow

edarterial

bloo

dpressure

loweringeff

ectfrom

thefirstweekat

thedo

seof

50mg/

kg.

Nosign

ificant

chan

gein

thehe

artrate

afterASX

-Otreatm

ent

ASX

-O(50mg/

kg)sign

ificantly

delaye

dtheincide

nceof

stroke

M. Zuluaga et al. Chemico-Biological Interactions 279 (2018) 145–158

149

Table2

Somepo

lymeric

system

sprop

osed

toprotectan

den

hanc

eastaxa

nthinprop

erties.

Ref.

Astax

anthin

source

System

Results

Antioxida

ntev

alua

tion

Polymeric

system

sHigue

ra-Ciapa

raet

al.,20

04[121

]Sy

nthe

ticastaxa

nthin(from

Sigm

aChe

mical

Co.

St.L

ouis,M

O,U

SA)

Microen

capsulationin

chitosan

matrix

cross-lin

kedwithglutaralde

hyde

bymultipleem

ulsion

/solve

ntev

aporation

Non

-hom

ogen

eous

size

anddiam

eter

(5–5

0μm

)Goo

dstorag

eat

T°<

45°C

for8weeks

Yield

of92

%an

dmoistureco

nten

tof

11.25%

77%

ofastaxa

nthinextraction

inmetha

nol/dich

lorometha

ne

Not

performed

Dai

etal.,20

13[125

]Astax

anthin

(from

Sigm

aAldrich

Co·St.

Louis,

MO,U

SA).

Extractedin

dich

lorometha

ne

Microen

capsulationusinga

Mon

odispe

rseDroplet

SprayDryer

tech

niqu

e

Micropa

rticleswithrelative

lyna

rrow

size

distribu

tion

(Meansize:

122μm

)Moistureco

nten

tof

4.55

wt%

Astax

anthin

released

immed

iately

from

themicropa

rticlesin

amixture

ofPB

Sbu

ffer

andaceton

e(16%

released

inthefirst6h).

Limited

loss

ofan

tiox

idan

tcapa

bilityat

140°C

calculated

byDPP

Hmetho

d(EC50

:1.872

×10

−4g/

mL)

Zhan

get

al.,20

17[126

]Astax

anthin

(nutraceutical

grad

e,ob

tained

from

Sigm

aChe

mical

Co.,U

SA)

Microen

capsulated

incalcium

algina

teHighen

capsulationeffi

cien

cy(>

85%)an

dHighstorag

estab

ility

atT°

<50

°CUniform

size

distribu

tion

(1.61mm)

Sustaine

dastaxa

nthinrelease

Not

cytotoxiceff

ecton

adipose-de

rive

dstem

cells

Determinationof

lipid

peroxida

tion

inhibitory

activity

bythe

thioba

rbituric

acid

(TBA

)metho

d

Kittika

iwan

etal.,

2007

[127

]H.p

luvialis

Astax

anthin

biom

ass(from

Wak

oChe

micals,

Japa

n)Algabe

adsco

atingwithmultiplelaye

rof

chitosan

fil

Uniform

size

andshap

e(0.431

±0.02

8cm

)Con

servationof

astaxa

nthinbiom

ass

Improv

edthermal

stab

ility

at80

°COptim

alstorag

eat

18°C

unde

rN2atmosph

erein

theda

rk

Loss

of3%

ofan

tiox

idan

tactivity

afteren

capsulationcalculated

byABT

Sassay

Leeet

al.,20

11[128

]Astax

anthin

rich

Xan

thop

hyllo

myces

dend

rorhou

s(A

stax

anthin,nu

traceu

tical

grad

eIG

ENEBiotechn

olog

yInc.

Colum

bia,

MD,U

SA

Astax

anthin-calcium

algina

tege

l(C

AG)

bead

sprep

ared

byionicge

lation

Bead

swithov

oidshap

ean

dsm

ooth

surface(ave

rage

size

2.41

mm)

23-31%tof

entrap

men

teffi

cien

cyCon

stan

tastaxa

nthinreleaserate

over

12h

Lipidpe

roxida

tion

inhibition

using

FTCan

dTB

Ametho

dsshow

edto

inhibitox

idationof

linoicacid

Bustos-G

arza

etal.,20

13[129

]H.p

luvialis

cyst

cells

(fFu

ture

Food

sSA

deCV,M

éxico)

oleo

resin

Extractedin

HClan

dethy

lacetate

Microen

capsulationusinglecithin

asem

ulsifier

andwhe

yproteinan

dgu

mArabicas

wallmaterials.

Prod

uctyieldof

theastaxa

nthinoleo

resin61

.2–7

0%8.31

–11.17

%of

moistureco

nten

tin

theastaxa

nthinoleo

resin

microen

capsulates

particle

sizesfrom

1to

10μm

Not

performed

Macha

doet

al.

2014

[130

]H.p

luvialis

(Eliz

abethAda

irMicroalga

eCollection,

Brazil)

Extractedin

dich

lorometha

ne

Co-po

lymer

poly

(hyd

roxy

butirate-co-

hydrox

yvalerate)

(PHBV

)

Minor

rigidity

andcrystalline

Precipitationpressure

influe

ncepa

rticle

size

(0.128

μm)

Increaseden

capsulationeffi

cien

cyby

increasing

astaxa

nthinbiom

ass

Not

performed

Suga

nyaet

ashe

eba,

2015

[131

]

Astax

anthin

isolated

from

crab

s(G

andh

imarke

t,Trichy

,Tam

ilnad

u,India).

Solven

t:he

xane

:isop

ropa

nol

Microen

capsulationprep

ared

by2%

sodium

algina

tean

d3%

calcium

chloride

usingiono

trop

icge

lation

metho

d.

Heterog

eneo

usmicrosphe

resin

size

(5.65μm

–8.98μm

)Rou

ndshap

edwithsm

ooth

surface.

Drugco

nten

t35

.25–

42.21mg

Not

performed

Shen

-FuLinet

al.,

2016

[123

]

Astax

anthin

(from

Orgch

emTe

chno

logies,

Hsinc

hu,T

aiwan

)Microen

capsulationin

calcium

algina

tebe

ads

Solution

calcium

chloride

,sod

ium

algina

te,a

ndTw

een20

assurfactant

Themicroen

capsulationyieldrang

edfrom

97.94to

25.83%

,and

load

ingeffi

cien

cyrang

edfrom

100%

to82

%.

Size

=68

5.9–

1044

.4μm

Goo

dstab

ility

after21

days

ofstorag

eat

25°C

Not

performed

M. Zuluaga et al. Chemico-Biological Interactions 279 (2018) 145–158

150

Table 3Some lipid based carriers evaluated to protect and enhance astaxanthin properties.

Ref. Astaxanthin source System Results Antioxidant evaluation

Lipid basedcarriers

Barroset al., 2001[138]

Astaxanthin fromSigma-Aldrich SwedenABn-hexane as solvent

Incorporation into egg-yolkphosphatidylcholine liposomes(PCL)

Astaxanthin was successfully incorporated intoPCL

Reduction of lipid damage causedby addition of:Ferrous ions + H2O2 = 45%t-ButOOH: 45%Ascorbate = 33%Reduction of H2O2-inducedlipoperoxidation in Iron PCL by26%.

Peng et al.,2010 [139]

Astaxanthin fromSigma Chemical Co. St.Louis, MO, USA)Solvent: Chloroform/methanol (2:1 v/v)

Liposomes Astaxanthin content 89 mg/gImprove stability after 25 h exposure in cell-freemediumHomogeneous dispersion in water (size251 ± 23 nm)Disintegration after 8 ± 1.8 minBetter access to cell surfaces after 2 hToxicity on Hep3B and HepG2 cell lines atconcentration between 5 and 40 μg/mL and nottoxicity on BNL CL2 line

Not performed

Acevedoet al., 2014[140]

Astaxanthin (fromSigma Chemical Co·St.Louis, MO, USA).

Microencapsulation in oil bodiescarriers based on oleosomesisolated from plant seeds in aratio (0.01–0.3).

Morphological stability (size average:3.444 ± 0.479 μm)Reduction of astaxanthin degradation after air andlight exposure by twice.Decrease of endothelial cell viability (28% at25–100 mg/mL of astaxanthin-microcapsules)

Intra-and extracellular ROSreduction up to 74.49% and47.7% respectively at 1000 mg/mL

Hamaet al., 2012[19,20]Kamezakiet al., 2016[141]

Astaxanthin (fromSigma Aldrich Co·St.Louis, MO, USA).

LiposomesEgg phosphatidylcholine (EPC)as based lipid

Liposomes size between 151.4 and 33.6 nmProtection of mouse skin fibroblast NIH3T3 cellsfrom hydroxyl radical induced cytotoxicity.

High hydroxyl radical scavengingat concentration< 20 μMReduction of singlet oxygenproduction up to 88%Inhibition of skin damageinduced by UV irradiation aftertopical application (40 μM)Co-encapsulation of astaxanthinand tocotrienol induce asynergistic scavenging activity

Bustamant-e et al.,2016 [142]

Astaxanthin (98%purity, fromSigma–Aldrich,Steinheim, Germany)Solvents: (1:1 v/v)diethyl ether:hexane

Microencapsulation withdifferent fatty acid compositionobtained by supercritical fluidextraction (SFE)Fatty acids: sunflower oil (SO) orhigh oleic sunflower oil (HOSO)

Lower degradation rateDroplet sizes of the SO + SFE and HOSO + SFEemulsions ranged from 0.31 to 0.56 μm and0.29–0.76 μm, respectively

Not performed

Odeberget al., 2003[143]

Commercialformulation of algalmeal and dextrin inhard gelatin capsules(Napro Pharma,Brattvaag, Norway).

Incorporation in three lipidbased formulations containingpolysorbate 80 and:1.Long-chain triglyceride (palmoil)2. glycerol mono- and dioleate3. glycerol mono- and dioleate,and sorbitan monooleate

Enhanced bioavailability, ranging from 1.7 to 3.7times Highest bioavailability using formulation Bafter a one dose human trial. Astaxanthin doses upto 40 mg were well tolerated.

Not performed

Ribeiroet al., 2005[144]

Crystalline astaxanthin(80% purity, BASF,Ludwigshafen,Germany)

O/W Emulsions prepared byrepeated premix membraneemulsificationDispersed in medium chaintriglyceride oil (palm oil)

30% of Astaxanthin degradation during storageDroplets stability up to 3 weeks

Not performed

Tachapruti-n et al.,2009 [145]

Astaxanthin (97% (w/w) purity, AcrosOrganics, Geel,Belgium)Solubilized in DMF//water

Polymeric nanocarries using:poly (ethylene oxide)-4-methoxycinnamoylphthaloylchi-tosan (PCPLC),

Stable aqueous suspensionNanospheres diameter: 68.3 ± 0.35 to312 ± 5.83 nm40% (w/w) astaxanthin loadingImproved thermal stability at 70 °C

Not performed

Anarjan etTan 2013[63,146,1-47]

Astaxanthin (> 90%,from Kailu EverBrillianceBiotechnology Co., Ltd.Beijing, China)

Nanodispersions using S1:Polysorbate 20 (PS20, 29% w/w), sodium caseinate (SC, 65%w/w) and gum Arabic (GA, 6%w/w); S2: Tween 20 with 62%w/w acetone and 38% w/wdichloromethane

Spherical-shaped with particle size of 88.9 nm forS2 and 114.6 nm using S1Astaxanthin loss of 32.4% (w/w) S2 and 20% (w/w) at 25 °C after 8 weeks of storage using S1Higher HT-29 uptake of astaxanthinnanodispersions

Not performed

Tamjidiet al., 2014[148]

H.Pluvialis oleoresin(astaxanthin content40%; from WuhanEreli Import & ExportCo. Ldt Wuhan, China)Extracted indichloromethane

Nanostructured lipid carriers(NCL) using tween80 andlecithin as emulsifiers and oleicacid and glycerol behenate aslipids

Good storage stability for 25 daysHigh drug loadingParticle size 85.1–138 nmDecrease in astaxanthin content due to hothomogenization method but increase in long-termstability

Not performed

Not performed(continued on next page)

M. Zuluaga et al. Chemico-Biological Interactions 279 (2018) 145–158

151

for 4 weeks. No safety concerns were reported and a positive effect onmetabolic syndrome and cognitive function was suggested.

Astaxanthin has been approved by the United States Food and DrugAdministration (US FDA) as well as the European Food Safety Authority(EFSA) for used as a food ingredient and feed additive. However, onlyastaxanthin from H. pluvialis and P. carotinifaciens have been approvedas a human dietary supplement at dosages from 12 to 24 mg/day and6 mg/day respectively, for no more than 30 days [28,43,69]. Althoughseveral toxicology studies confirmed astaxanthin safety and no toxicityrisks, it is important to consider that animal studies are of doubtfulhuman relevance and that there is a need for further evaluations ofastaxanthin toxicity in humans.

3. Influence of oxidative stress in I/R: Astaxanthin antioxidanttreatment

3.1. ROS production during I/R injury

Reperfusion injury is induced after blood flow is restored followingan ischemic period. Recent studies shown that an oxidative stress couldbe a critical factor involved in the pathogenesis of this injury [70]. Theinduction of an ischemic state leads to an imbalance in the oxygenproduction and consumption. This lack of oxygen further restricts bloodsupply, inducing a hypoxic state which leads to microvascular dys-function [71]. Restoration of blood flow and reoxygenation is fre-quently associated with an increased amount of tissue injury and anelevated inflammatory response [72], also called ‘reperfusion injury’[73]. Reperfusion injury leads to the depression of the inner body de-fense mechanism, inducing an imbalance between a burst of ROS pro-duction and the inability of reoxygenated cells to handle this radicalload [8]. Under these conditions, cell death programs including apop-tosis, autophagy-associated cell death and necrosis [74] are activated,leading to a multi-organ failure, even if only one organ underwent I/R[75]. Here, extracellular ATP depletion from apoptotic cells acts as a‘find-me’ signal that attracts phagocytes [76,77]. Additionally, limitedoxygen availability is associated with the activation of inflammatorysignals which control the stability of the transcription nuclear factorNF-kB [73] through a mechanism involving hypoxia-dependent in-hibition of oxygen sensors [78], and adaptive immune responses thatinvolves the infiltration of various types of inflammatory cells (neu-trophils, T lymphocytes, monocyte/macrophages) [79,80].

In normal physiological conditions the undamaged endotheliumprevents adhesion and activation of platelets and leukocytes by severalmechanisms [81].

During reperfusion injury, adhesion of platelets and leukocytes toendothelial cells are enhanced leading a procoagulant state as well asplatelet and leukocyte activation [83]. This activation results in theinduction of proinflammatory cytokines and chemokines (TNF and IL-

1B) [84] that are further released by activated leukocytes in the re-perfused blood [85]. Under these circumstances, the endothelial cellbarrier gets weak, increasing vascular permeability and leakage [86].Endothelial damage is enhanced by the hydroxyl radical, superoxideand peroxynitrite overproduction that are formed following the reac-tion of NO with oxygen in the reperfused blood [82]. L-arginine is themain precursor of endothelial nitric oxide synthase, eNOS, which is incharge of NO synthesis which further enhances inducible NOS and iNOS[87]. Moreover, activation of NADPH oxidase in activated neutrophils,induces peroxynitrite which, then, in turn, generates more ROS by theincreased availability of free iron during ischemia [88]. This excessiveradical generation leads to lipid peroxidation of tissues within thesubendothelial space. Accumulation of oxidized low-density lipopro-teins (ox-LDL) within monocytes-derived macrophages generates foamcells that further amplify the inflammatory cascade [89], ultimatelyleading to the formation of thrombus and occlusion of the vessel [90](Fig. 2).

3.2. Astaxanthin: An antioxidant ROS blocking agent

Natural carotenoids have shown particular abilities to entrap ROSand enhance the cellular capacity to block oxidative stress [91]. Theeffects of carotenoids vary depending on how they interact with cellmembranes [92,93]. Astaxanthin carotenoid has been shown to reducelipid peroxidation damages by the preservation of membranes struc-tures using a polyunsaturated fatty acid enriched membrane model[94]. This action was attributed to its polar end groups which extendedtoward the polar regions of the membrane bilayer [95].

Astaxanthin has been currently studied in the cardiovascular fieldthanks to its antioxidants and anti-inflammatory properties [95]. As-taxanthin showed reduction of blood coagulation, platelet aggregationand promoted fibrinolytic activity in a high-fat diet-induced hyperli-pidemic rats. These positive effects were correlated with decrease ofserum lipid and lipoproteins levels, antioxidants production and pro-tection of endothelial cells [96]. Moreover, the protective effect of as-taxanthin has been studied on different in vivo models of I/R such asmyocardial, cerebral, liver and renal (Table 1). In which animals re-ceived an oral or intravenous injection of astaxanthin dose rangingbetween 5 and 500 mg/kg/day. In these cases, astaxanthin was ad-ministered in both hydrophilic (solubilized on DI water and other or-ganic solvents) and lipophilic (solubilized on oils) formulations beforeinducing the ischemic damage, thus acting as a preventive agent. Awater soluble synthetic astaxanthin derivative or disodium disuccinateastaxanthin (Cardax Hawaii Biotech, Inc., USA) was studied on ex-perimental myocardial I/R models in rat, rabbit and dogs (Table 1).After parental administration this derivative showed a potential effi-cacy to reduce infarct size and plasma lipid peroxidation levels attrib-uted the direct scavenging of superoxide anion [97]. Similar results

Table 3 (continued)

Ref. Astaxanthin source System Results Antioxidant evaluation

Meor MohdAffandiet al., 2011[149]

Astareal 10FC grade(an oil extractcontaining 10% w/wof standardizedastaxanthin, from FujiChemical Industry,Nakaniikawa, Toyama,Japan).

Nanoemulsion using Tween 80and lecithin as emulsifiers (2.5%w/w)

Particles size at 5 cycles of homogenizing pressure122.9 ± 1.55 nm.Conservation of stability and storage at25 ± 2 °C/60% ± 5% relative humidity for 3months.

Chun-HungChiu et al.,2016 [150]Secondpart of thestudy[139].

Astaxanthin (> 99%,from the Fuji ChemicalIndustry Co., Ltd.Toyama Prefecture,Japan).

Liposomes Particles size distribution of 240 ± 58 nmAstaxanthin instantaneous pharmacokineticsrelease from the nanoliposome particles.Efficient and stable transport allowing a higherintrahepatic uptake

Attenuation of nuclear levels ofiNOS and NF-kBHepatoprotective effects andcompletely alleviated the acuteinflammatory status at a 10 mg/kg-day dosage.

M. Zuluaga et al. Chemico-Biological Interactions 279 (2018) 145–158

152

were obtained with natural astaxanthin and carotenoids mixtures (Vi-taePro, astaxanthin, lutein and zeaxanthin) in a rat model of I/R.

Oil-based astaxanthin formulations, administered mostly by oralgavage, showed a potential reduction of inflammatory cytokines ex-pression, decreased organ infarct size area and reduction of arterialblood pressure lowering the risk of strokes. These studies are brieflysummarized on Table 1.

Animal studies supports the potential preventive effect of astax-anthin supplementation to reduce the impact of cardiovascular dis-eases. The potential astaxanthin effects in the prevention and treatmentof cardiovascular disease have been extensively studied [23,25,28].Moreover, first preclinical studies support its antioxidant abilities toprevent oxidative processes. For instance, patients who received as-taxanthin supplementation showed an increased resistance to LDLoxidation when administered at doses of 1.8–21.6 mg/day during 14days [108] and a slight glucose-lowering effect at doses of 4–20 mg/dayin other studies [109]. Additionally, the amelioration of triglycerideand HDL-cholesterol in correlation with increased serum adiponectinlevels after administration at doses of 12–18 mg/days during 12 weekswas also reported in another patient group [110]. In a clinical study,Park et al. [47] examined the action of dietary astaxanthin (2 and 8 mg/day for 8 days) in regulating immune response, oxidative damage andinflammation in humans. Results showed an enhancement of immunemarkers and reduction in DNA oxidative damage biomarker and in-flammation.

Additionally, there is evidence of the effects of astaxanthin againstoxidative damage in other disorders such as diabetes, obesity andneurodegenerative diseases.

Animal studies showed the astaxanthin potential to reduce the al-tered oxidative stress environment in diabetic and obese rats. Yeh et al.[111] studied the astaxanthin capacity to protect against oxidativedamage in the ocular tissues of streptozotocin-induced diabetic Wistarrats receiving 3 mg/kg daily of astaxanthin for 8 weeks. Their resultssuggested a protective effect in the preservation and reduction of dia-betic retinopathy mediated by downregulation of NF-κB activity, anincrease of antioxidant enzymes, and reduction of downstream in-flammatory mediators' expression. Mimoun-Benar et al. [112] studiedthe capacity of free-astaxanthin to reduce plasmatic triglycerides in pre-obesity diet-induced dyslipidaemia mice, showing a positive effect inreducing triglycerides concentrations up to 45% but not cholesterollevels after astaxanthin supplementation for 8 weeks. Also, Al-bulishet al. [113] showed astaxanthin capacity to prevent the increasingoxidative stress biomarkers in rats presenting streptozotocin-inducedhyperglycemia and pancreatic cell injury, after the animals receivedoral astaxanthin administration (20 mg/kg of body weight) for 12weeks.

Human studies have also been conducted to identify the potential ofusing this antioxidant to reduce overweight and obesity problemslinked to oxidative stress induction. Satoh et al. [68] showed the re-duction of systolic blood pressure, triglyceride, and fasting glucosevalues after astaxanthin intake by patients with borderline diabetesmellitus or persons at risk for metabolic syndrome. Choi et al. [114]confirmed the capacity of astaxanthin to improve oxidative stress bio-markers by suppressing lipid peroxidation and stimulating the activityof the antioxidant defense system in overweight and obese adults inKorea, these patients daily received an oral administration of astax-anthin at concentrations up to 20 mg for 3 weeks.

Oxidative stress plays an important role in the induction of neuro-logical diseases by damaging macromolecules and leading to neuronaldysfunction [115]. Astaxanthin has been evaluated as a potential neu-roprotective agent due to its capacity to cross the brain blood barrierprotecting the brain for acute injury and chronic neurodegeneration[116]. Grimmig et al. [117] reviewed the potential of astaxanthin topromote or maintain neural plasticity, suggesting that astaxanthincould increase the cognitive function by promoting neurogenesis andbehavioral performance on hippocampal-dependent tasks. Additionally,Ta

ble4

Cyclode

xtrininclusionco

mplexes

evalua

tedto

protectan

den

hanc

eastaxa

nthinprop

erties.

Referen

ceAstax

anthin

source

System

Results

Antioxida

ntev

alua

tion

Cyclode

xtrininclusion

complex

Don

get

al.,20

14[155

]Astax

anthin

(purity>

98%,from

Dr.

Ehrenstorfer

Co.

Ltd.

German

y)InclusionwithHyd

roxy

prop

yl-β-

cyclod

extrin

Inclusionrate

30.4%

Starting

deco

mpo

sition

tempe

rature

at50

°CGoo

dwater

solubilityat

50mg/

mof

astaxa

nthin

conc

entrationev

enafter12

days.

Stab

lestorag

eun

derda

rkan

dlig

htco

nditions

at4,

25,3

7,an

d50

°Cwithin26

0h

Com

plex

show

edaDPP

Hradicalactivity

lower

than

astaxa

nthinat

sameco

ncen

trations

HighFe

3+

toFe

2+

redu

cedpo

wer

capa

bilities

than

pure

astaxa

nthinun

derthesame

conc

entration

Lockwoo

det

al.,

2003

[156

]Astax

anthin

Sigm

alot71

K15

40Inclusionin

captisol

(Sulfobu

tylEthe

rb-Cyclode

xtrin)

Increasedtheap

parent

water

solubility

approx

imately71

-fold,

toaco

ncen

trationof

2mg/

mL

Not

performed

Kim

etal.,20

10[157

]Astax

anthin

(purch

ased

from

Sigm

aChe

mical

Co·St.Lo

uis,

MO,U

SA).

Solven

t:Dichlorom

etha

nean

dAcetone

Inclusionwithβ-cyclod

extrin

Enha

nced

thewater

solubilityup

to11

0-fold

atpH

6.5an

d25

°C.

Improv

edstab

ility

againsth

eat,lig

ht,a

ndox

idation

byov

er7–

9folds

Thermal

stab

ility

even

at10

0°C.

Not

performed

Che

net

al.,20

07[158

]Astax

anthin

prep

ared

from

smallshrim

psSo

lven

t:Dichlorom

etha

ne/acetone

1:1v/

v

Inclusionwithβ-cyclod

extrin

Improv

edwater

solubility<

0.5mg/

mLthermal

(57°C)an

dlig

htstab

ility

after6da

ysInclusionrate

of48

.96%

Not

performed

Nalaw

adeet

Gajjar

2015

[159

]Astax

anthin

(from

Shan

gyuNHU

Bio-

Che

m,C

hina

)So

lubiliz

edin

Hyd

ro-alcoh

olic

solution

(2:1

v/v)

Inclusionwithmethy

lated-β-

cyclod

extrin

usingspraydrying

tech

niqu

e

Solubilityen

hanc

ed54

times

over

astaxa

nthin

alon

e.A

dissolutionrate

of85

%ov

er45

min

Increasedbio-accessibility

onHep

G2celllin

e.

Not

performed

M. Zuluaga et al. Chemico-Biological Interactions 279 (2018) 145–158

153

Wu et al. [118] reviewed astaxanthin's effect to act as a potentialneuroprotective agent, based on its anti-oxidative, anti-inflammatory,and anti-apoptotic effects. In particular, Grimmig et al. studied [119]the potential of astaxanthin to attenuate the neurodegeneration processinduced in Parkinson's disease, while Lobos et al. [120] showed as-taxanthin capacity to protect neurons from the oligomers' noxious ef-fects on mitochondrial ROS production on primary hippocampal cul-tures in vitro.

3.3. Encapsulation carrier's systems

Despite the positive results described above there is still a lack in theunderstanding of astaxanthin's therapeutic mode of action, uptake,distribution, pharmacokinetics, and metabolism. Furthermore, solvingastaxanthin stability drawbacks and parenteral administration pro-blems are of crucial interest for the development of astaxanthin-basedtherapies to prevent and treat oxidative stress-induced cardiovascularpathologies. Consequently, the implementation of new biomaterials toact as astaxanthin vectors in vivo is of vital interest. Tables 2–4 sum-marize some carrier's systems successfully developed for overcomingastaxanthin delivery challenges. These systems were divided into threegroups including polymeric systems, lipid-based carriers, and inclusioncomplex using cyclodextrins (Fig. 3).

3.3.1. Polymeric systemsThe microencapsulation process with polymeric systems consists in

the formation of a polymeric matrix or coating layer around a particularcompound to provide a physical barrier between the core material andenvironmental conditions. These types of systems protect the com-pound's biological activity and enhance its physicochemical stability.

Natural polymeric systems include polysaccharides like cellulose,starch, gum Arabic, alginate or chitosan [121] and the use of proteinslike albumin, gelatin or soy proteins [122] (Table 2). Microencapsula-tion with polymer matrices controls the molecule release, reducing thecore reactivity with environmental factors and facilitating molecularhandling [123,124].

In polymeric nanosystems the absorption profile of the loaded mo-lecules is driven by the particle size, shape and surface properties of thenanoparticles [54], which could be useful to control the drug releaserate during oral administration until it reaches the systemic circulation[122]. Indeed, chitosan-alginate complexes have been shown to de-grade slowly in phosphate buffer, avoiding the initial release of drugsoccurring when using uncoated microspheres [31]. Conversely, poly-meric micelles improve their steric stabilization and ability to interactwith cells due to their hydrophilic shell [122].

Table 2, presents nine different studies using polymeric matrixes toimprove astaxanthin solubility properties. Natural polysaccharides suchas chitosan and alginate are currently being studied for astaxanthinmicroencapsulation due to their biocompatibility and biodegradabilityproperties. For instance, chitosan showed to improve astaxanthin sto-rage conditions and to preserve its antioxidant scavenging abilities, asconfirmed by ABTS chemical method. A high astaxanthin loading effi-ciency was reported in studies using calcium-alginate, however mi-crocapsules size distribution varied from 5.6 to 2041 μm between thestudies. The preservation of the lipid peroxidation inhibitory activitywas confirmed by two of four studies using the TBA method and onlyone study evaluated the in vitro cytotoxicity of the system. In general,all polymeric methods improved astaxanthin solubility but lack in thecomplete evaluation and verification of the antioxidant activity pre-servation after encapsulation.

Fig. 3. Different strategies explored for astaxanthin encapsulation and posterior delivery.

M. Zuluaga et al. Chemico-Biological Interactions 279 (2018) 145–158

154

3.3.2. Lipid based carriersLipid based carriers include micelles, solid lipid nanoparticles

(SLN), nanostructured lipid carriers (NLC), nanoemulsions and micro-emulsions. These systems had been used to encapsulate, protect, anddeliver lipophilic bioactive components by enhancing their long-termstability while increasing bioavailability [132]. The system stabilizationis achieved by surface charge or by surface adsorption of a layer ofsurfactant or polymer, or the combination of both methods [122].

O/W microemulsions and nanoemulsions differ according to theirstability. Microemulsions are thermodynamically stable colloidal dis-persions consisting of small spheroid particles dispersed within anaqueous medium, O/W nanoemulsions refer to a thermodynamicallyunstable colloidal dispersion consisting of two immiscible liquids, beingone of the liquids dispersed in the other liquid [132]. SLN are a mix ofO/W nano/micro-emulsions in which the lipid phase is fully crystal-lized and has a highly-ordered crystalline structure at room/bodytemperature [133]. The low drug loading capacity and drug releaseafter polymorphic transition of the lipid core during storage representsa disadvantage for the use of SLN [133,134]. Nanostructured lipidcarriers (NLC) are modified SLN consisting of a lipid phase of a bio-compatible mixture of solid and liquid lipids in a less-ordered crystal-line structure [135,136]. The incorporation of oil into the core of a solidlipid leads to a higher loading capacity and controlled drug release.Here, the drug is dissolved in the oil and simultaneously encapsulatedin the solid lipid [137]. Lipid based carriers size can range from around10 nm for micelles to hundreds of nanometers for other systems [132].

Lipid based carrier systems presented in Table 3 showed the en-hancement of astaxanthin properties. Five of twelve studies evaluatedthe in vitro interaction of the system after cellular supplementationwithout reporting toxicity problems. The in vitro antioxidant capacity ofthe system was assessed in four studies reporting the reduction of ROSlevels and the attenuation of cellular inflammatory markers. Moreover,one human trial study revealed the improvement of astaxanthin bioa-vailability after incorporating into a lipid based formulation [143].

3.3.3. Inclusion complex using cyclodextrinCyclodextrins have been used extensively as additives to increase

the solubility of poorly water-soluble organic compounds [122]. Cy-clodextrins are natural macrocyclic oligosaccharides well known forhaving toroid-shaped structures with rigid lipophilic cavities and ahydrophilic outer surface. They are able to enclose highly hydrophobicmolecules inside their hydrophobic cavity, constituting a true mole-cular encapsulation [151]. The resulting non-covalent inclusions orhost–guest complexes are of current scientific and technological interestfor their particular physical, chemical and biological properties. Thesenon-covalent associations can improve the guests water solubility,bioavailability and stability [152], while regulating the release of theguest molecules [153,154].

Cyclodextrin systems highly increased the astaxanthin water solu-bility and its stability against heat, light and oxygen. Two of the sixsystems presented in Table 4 evaluated the chemical antioxidant ac-tivity of the inclusion system using the DPPH, the reduction powerassay and the hydroxyl radical scavenging test. Moreover, one studyconfirmed the system bio-accessibility on HepG2 cell line.

Almost all reviewed studies were focused on chemical formulationsand stabilization parameters. Polymeric, lipid and cyclodextrin systemsshowed an improvement on astaxanthin solubility and stability prop-erties. However, very few studies evaluated astaxanthin antioxidantcapacities after the encapsulation process. Some studies that used thelipid based formulations assessed the in vitro potential of the system.Inclusion process using cyclodextrin represent a practical option tostudy different in vivo delivery pathways due to the higher water so-lubility of the obtained system. Regardless of the results reported usingthe different techniques, all of them require a deeper chemical andbiological characterization to confirm their potential to be used as as-taxanthin carrier systems in order to consider for future evaluations in

clinical applications for the prevention and treatment of cardiovasculardiseases.

4. Conclusion

Despite the influence of ROS to destabilize membrane and cellhomeostasis, a regular production of these radicals is essential in themaintaining of redox signaling. Thus, antioxidant systems oversee ROSregulation without completely eliminating them. All antioxidants havedifferent ways of action; their biological activity may also be condi-tioned by the cellular structure in which they act. Astaxanthin hasshown potent antioxidant actions to stabilize ROS influx during oxi-dative stress related diseases such as I/R injury, as presented here.Indeed, astaxanthin showed a strong ability to reduce lipid oxidationthanks to its polar end groups which extend toward the polar regions ofthe membrane bilayer, thus contributing to the inhibition of thrombusand atherosclerotic plaque formation. However, a drawback of astax-anthin's action has also been attributed to its structure, which renders itprone to oxidation and lowers its bioavailability. Protective en-capsulation systems have been studied to solve these drawbacks.Additionally, new delivery systems may also contribute to limit po-tential untoward effects of in vivo antioxidant therapy that have beenlimited by antioxidant appropriate doses. Finally, the studies reviewedhere show the interesting properties and potential medical use of as-taxanthin to treat oxidative stress related pathologies, particularly incardiovascular diseases such as I/R injury.

Transparency document

Transparency document related to this article can be found online athttp://dx.doi.org/10.1016/j.cbi.2017.11.012.

References

[1] B. Halliwell, Reactive oxygen species in living systems: source, biochemistry, androle in human disease, Am. J. Med. 91 (1991) 14S–22S, http://dx.doi.org/10.1016/0002-9343(91)90279-7.

[2] S. Nemoto, K. Takeda, Z.X. Yu, V.J. Ferrans, T. Finkel, Role for mitochondrialoxidants as regulators of cellular metabolism, Mol. Cell. Biol. 20 (2000)7311–7318, http://dx.doi.org/10.1128/MCB.20.19.7311-7318.2000.

[3] T. Finkel, Oxygen radicals and signaling, Curr. Opin. Cell Biol. 10 (1998) 248–253,http://dx.doi.org/10.1016/S0955-0674(98)80147-6.

[4] M. Schieber, N.S. Chandel, ROS function in redox signaling and oxidative stress,Curr. Biol. 24 (2014) 453–462, http://dx.doi.org/10.1124/dmd.107.016501.CYP3A4-Mediated.

[5] A. Bhattacharyya, R. Chattopadhyay, S. Mitra, S.E. Crowe, Oxidative stress: anessential factor in the pathogenesis of gastrointestinal mucosal diseases, Physiol.Rev. 94 (2014) 329–354, http://dx.doi.org/10.1152/physrev.00040.2012.

[6] H. Li, U. Förstermann, Uncoupling of endothelial NO synthase in atherosclerosisand vascular disease, Curr. Opin. Pharmacol. 13 (2013) 161–167, http://dx.doi.org/10.1016/j.coph.2013.01.006.

[7] U. Landmesser, S. Dikalov, S.R. Price, L. McCann, T. Fukai, S.M. Holland,W.E. Mitch, D.G. Harrison, Oxidation of tetrahydrobiopterin leads to uncouplingof endothelial cell nitric oxide synthase in hypertension, J. Clin. Invest. 111 (2003)1201–1209, http://dx.doi.org/10.1172/JCI200314172.

[8] J.M. McCord, Oxygen-derived free radicals in postischemic tissue injury, N. Engl.J. Med. 312 (1985) 159–163, http://dx.doi.org/10.1056/NEJM198501173120305.

[9] G. Vogiatzi, D. Tousoulis, C. Stefanadis, Role of oxidative stress in atherosclerosis,Hell. J. Cardiol. 50 (2009) 402–409, http://dx.doi.org/10.1016/S0002-9149(02)03144-2.

[10] H.E.P.S. Souza, L.I.C.S. Souza, V. Anastacio, A. Pereira, M. de L. Junqueira,J.E. Krieger, P.L. Da luz, O. Augusto, F.R.M. Laurindo, Vascular oxidant stressearly after balloon injury: evidence for increased NAD(P)H oxidoreductase ac-tivity, Free Radic. Biol. Med. 28 (2000) 1232–1242.

[11] F.Z. Meerson, V.E. Kagan, Y.P. Kozlov, L.M. Belkina, Y.V. Arkhipenko, The role oflipid peroxidation in pathogenesis of ischemic damage and the antioxidant pro-tection of the heart, Basic Res. Cardiol. 77 (1982) 465–485.

[12] T. Finkel, N.J. Holbrook, Oxidants, oxidative stress and the biology of ageing,Nature 408 (2000) 239–247.

[13] M. Zuluaga, V. Gueguen, G. Pavon-Djavid, D. Letourneur, Carotenoids from mi-croalgae to block oxidative stress, BioImpacts 7 (2017) 1–3, http://dx.doi.org/10.15171/bi.2017.01.

[14] G. Riccioni, N. D'Orazio, S. Franceschelli, L. Speranza, Marine carotenoids andcardiovascular risk markers, Mar. Drugs 9 (2011) 1166–1175, http://dx.doi.org/

M. Zuluaga et al. Chemico-Biological Interactions 279 (2018) 145–158

155

10.3390/md9071166.[15] F.J. Pashkow, Oxidative stress and inflammation in heart disease: do antioxidants

have a role in treatment and/or prevention? Int. J. Inflam. 2011 (2011) 514623,http://dx.doi.org/10.4061/2011/514623.

[16] H. Tapiero, D.M. Townsend, K.D. Tew, The role of carotenoids in the prevention ofhuman pathologies, Biomed. Pharmacother. 58 (2004) 100–110, http://dx.doi.org/10.1016/j.biopha.2003.12.006.

[17] J. Fiedor, K. Burda, Potential role of carotenoids as antioxidants in human healthand disease, Nutrients 6 (2014) 466–488, http://dx.doi.org/10.3390/nu6020466.

[18] X.-L. Xue, X.-D. Han, Y. Li, X.-F. Chu, W.-M. Miao, J.-L. Zhang, S.-J. Fan,Astaxanthin attenuates total body irradiation-induced hematopoietic system in-jury in mice via inhibition of oxidative stress and apoptosis, Stem Cell Res. Ther. 8(2017) 7, http://dx.doi.org/10.1186/s13287-016-0464-3.

[19] S. Hama, S. Uenishi, A. Yamada, T. Ohgita, H. Tsuchiya, E. Yamashita, K. Kogure,Scavenging of hydroxyl radicals in aqueous solution by astaxanthin encapsulatedin liposomes, Biol. Pharm. Bull. 35 (2012) 2238–2242, http://dx.doi.org/10.1248/bpb.b12-00715.

[20] S. Hama, K. Takahashi, Y. Inai, K. Shiota, R. Sakamoto, A. Yamada, H. Tsuchiya,K. Kanamura, E. Yamashita, K. Kogure, Protective effects of topical application of apoorly soluble antioxidant astaxanthin liposomal formulation on ultraviolet-in-duced skin damage, J. Pharm. Sci. 101 (2012) 2909–2916, http://dx.doi.org/10.1002/jps.

[21] M. Kurashige, E. Okimasu, M. Inoue, K. Utsumi, Inhibition of oxidative injury ofbiological membranes by astaxanthin, Physiol. Chem. Phys. Med. NMR 22 (1990)27–38.

[22] L.S. Jacobsson, X.M. Yuan, B. Ziedén, A.G. Olsson, Effects of α-tocopherol andastaxanthin on LDL oxidation and atherosclerosis in WHHL rabbits,Atherosclerosis 173 (2004) 231–237, http://dx.doi.org/10.1016/j.atherosclerosis.2004.01.003.

[23] Y. Kishimoto, H. Yoshida, K. Kondo, Potential anti-atherosclerotic properties ofastaxanthin, Mar. Drugs 14 (2016) 1–13, http://dx.doi.org/10.3390/md14020035.

[24] G. Hussein, M. Nakamura, Q. Zhao, T. Iguchi, H. Goto, U. Sankawa, H. Watanabe,Antihypertensive and neuroprotective effects of astaxanthin in experimental ani-mals, Biol. Pharm. Bull. 28 (2005) 47–52, http://dx.doi.org/10.1248/bpb.28.47.

[25] R.G. Fassett, J.S. Coombes, Astaxanthin: a potential therapeutic agent in cardio-vascular disease, Mar. Drugs 9 (2011) 447–465, http://dx.doi.org/10.3390/md9030447.

[26] I. Higuera-Ciapara, L. Félix-Valenzuela, F.M. Goycoolea, Astaxanthin: a review ofits chemistry and applications, Crit. Rev. Food Sci. Nutr. 46 (2006) 185–196,http://dx.doi.org/10.1080/10408690590957188.

[27] R.R. Ambati, S.M. Phang, S. Ravi, R.G. Aswathanarayana, P.S. Moi, S. Ravi,R.G. Aswathanarayana, Astaxanthin: sources, extraction, stability, biological ac-tivities and its commercial applications - a review, Mar. Drugs 12 (2014) 128–152,http://dx.doi.org/10.3390/md12010128.

[28] F. Visioli, C. Artaria, Astaxanthin in cardiovascular health and disease: mechan-isms of action, therapeutic merits, and knowledge gaps, (2016), http://dx.doi.org/10.1039/c6fo01721e.

[29] C. Caballo, E.M. Costi, M.D. Sicilia, S. Rubio, Determination of supplementalfeeding needs for astaxanthin and canthaxanthin in salmonids by supramolecularsolvent-based microextraction and liquid chromatography-UV/VIS spectroscopy,Food Chem. 134 (2012) 1244–1249, http://dx.doi.org/10.1016/j.foodchem.2012.03.051.

[30] A. Nagao, Absorption and metabolism of dietary carotenoids, BioFactors 37 (2011)83–87, http://dx.doi.org/10.1002/biof.151.

[31] G. Sandmann, Carotenoid biosynthesis in microorganisms and plants, Eur. J.Biochem. 223 (1994) 7–24, http://dx.doi.org/10.1111/j.1432-1033.1994.tb18961.x.

[32] R. Sarada, U. Tripathi, G. Ravishankar, Influence of stress on astaxanthin pro-duction in Haematococcus pluvialis grown under different culture conditions,Process Biochem. 37 (2002) 623–627, http://dx.doi.org/10.1016/S0032-9592(01)00246-1.

[33] G.R. Lorenz, R.T. Cysewski, Commercial potential for Haematococcus microalgaeas a natural source of astaxanthin, Trends Biotechnol. 18 (2000) 160–167.

[34] Y. Yang, B. Kim, J. Lee, Astaxanthin structure, metabolism, and health benefits, J.Hum. Nutr. Food Sci. 1 (2013) 1–11 http://www.j-scimedcentral.com/Nutrition/Articles/nutrition-1-1003.pdf.

[35] M.M.R. Shah, Y. Liang, J.J. Cheng, M. Daroch, Astaxanthin-Producing green mi-croalga haematococcus pluvialis: from single cell to high value commercial pro-ducts, Front. Plant Sci. 7 (2016) 531, http://dx.doi.org/10.3389/fpls.2016.00531.

[36] K.D.K. Nguyen, Astaxanthin: a comparative case of synthetic vs. natural produc-tion, Chem. Biomol. Eng. Publ. Other Work 1 (2013) 1–11.

[37] M. Rodríguez-Sáiz, J.L. De La Fuente, J.L. Barredo, Xanthophyllomyces den-drorhous for the industrial production of astaxanthin, Appl. Microbiol. Biotechnol.88 (2010) 645–658, http://dx.doi.org/10.1007/s00253-010-2814-x.

[38] A. Tsubokura, H. Yoneda, H. Mizuta, Aerobic Gram-negative astaxanthin-produ-cing bacteri urn, Int. J. Sytematic Bacteriol. 49 (1999) 277–282.

[39] B. Capelli, G. Cysewski, ASTAXANTHIN Natural Astaxanthin : King of theCarotenoids, Published by Cyanotech Corporation, 2007.

[40] L. Fuji, Chemical Indusctry Co, New dietary ingredient notification for astaxanthinextracted from haematococcus algae, U. S. Food Frug Adm. 1 (2004) 1–6.

[41] G. Hussein, U. Sankawa, H. Goto, K. Matsumoto, H. Watanabe, Astaxantin, acarotenoid with potentian un human health and nutrition, J. Nat. Prod. 69 (2006)443–449, http://dx.doi.org/10.1021/np050354.

[42] M. Guerin, M.E. Huntley, M. Olaizola, Haematococcus astaxanthin: applicationsfor human health and nutrition, Trends Biotechnol. 21 (2003) 210–216, http://dx.

doi.org/10.1016/S0167-7799(03)00078-7.[43] (European Food Safety Authority) EFSA, Opinion of the scientific panel on ad-

ditives and products or substances used in animal feed on the request from thecommission on the safety of use of colouring agents in animal nutrition, EFSA J.291 (2005) 1–40.

[44] R. Lorenz, A Technical Review of Haematococcus Algae, NatuRoseTM Tech. Bull.#060,Cyanotech Corp, Kailua-Kona, HI, USA, 1999, pp. 1–12.

[45] P. Régnier, J. Bastias, V. Rodriguez-Ruiz, N. Caballero-Casero, C. Caballo,D. Sicilia, A. Fuentes, M. Maire, M. Crepin, D. Letourneur, V. Gueguen, S. Rubio,G. Pavon-Djavid, Astaxanthin from haematococcus pluvialis prevents oxidativestress on human endothelial cells without toxicity, Mar. Drugs 13 (2015)2857–2874, http://dx.doi.org/10.3390/md13052857.

[46] W. Zhang, J. Wang, J. Wang, T. Liu, Attached cultivation of Haematococcus plu-vialis for astaxanthin production, Bioresour. Technol. 158 (2014) 329–335, http://dx.doi.org/10.1016/j.biortech.2014.02.044.

[47] D. Ruen-ngam, A. Shotipruk, P. Pavasant, Comparison of extraction methods forrecovery of astaxanthin from Haematococcus pluvialis, Sep. Sci. Technol. 46 (2010)64–70, http://dx.doi.org/10.1080/01496395.2010.493546.

[48] C.D. Kang, S.J. Sim, Direct extraction of astaxanthin from Haematococcus cultureusing vegetable oils, Biotechnol. Lett. 30 (2008) 441–444, http://dx.doi.org/10.1007/s10529-007-9578-0.

[49] M. Kobayashi, Y. Kurimura, Y. Sakamoto, Y. Tsuji, Selective extraction of astax-anthin and chlorophyll from the green alga Haematococcus pluvialis, Biotechnol.Tech. 11 (1997) 657–660, http://dx.doi.org/10.1023/A:1018455209445.

[50] S. Machmudah, A. Shotipruk, M. Goto, M. Sasaki, T. Hirose, Extraction of astax-anthin from Haematococcus p luvialis using supercritical CO 2 and ethanol as en-trainer, Ind. Eng. Chem. Res. 45 (2006) 3652–3657, http://dx.doi.org/10.1021/ie051357k.

[51] J.O. Valderrama, M. Perrut, W. Majewski, Extraction of astaxantine and phyco-cyanine from microalgae with supercritical carbon dioxide, J. Chem. Eng. Data 48(2003) 827.

[52] F.A. Reyes, J.A. Mendiola, E. Ibañez, J.M. Del Valle, Astaxanthin extraction fromHaematococcus pluvialis using CO 2-expanded ethanol, J. Supercrit. Fluids 92(2014) 75–83, http://dx.doi.org/10.1016/j.supflu.2014.05.013.

[53] A. Ballesteros-Gomez, M.D. Sicilia, S. Rubio, Supramolecular solvents in the ex-traction of organic compounds. A review, Anal. Chim. Acta 677 (2010) 108–130,http://dx.doi.org/10.1016/j.aca.2010.07.027.

[54] A.R. Bilia, B. Isacchi, C. Righeschi, C. Guccione, M.C. Bergonzi, Flavonoids loadedin Nanocarriers : an opportunity to increase oral bioavailability and bioefficacy,Food Nutr. Sci. 5 (2014) 1212–1227, http://dx.doi.org/10.4236/fns.2014.513132.

[55] D.M. Small, S.A. Penkett, D. Chapman, Studies on simple and mixed bile saltmicelles by nuclear magnetic resonance spectroscopy, Biochem. Biophys. Acta 176(1969) 178–189.

[56] Y. Okada, M. Ishikura, T. Maoka, Bioavailability of astaxanthin in haematococcusalgal extract: the effects of timing of diet and smoking habits, Biosci. Biotechnol.Biochem. 73 (2009) 1928–1932, http://dx.doi.org/10.1271/bbb.90078.

[57] K. Nagaoka, T. Matoba, Y. Mao, Y. Nakano, G. Ikeda, S. Egusa, M. Tokutome,R. Nagahama, K. Nakano, K. Sunagawa, K. Egashira, A new therapeutic modalityfor acute myocardial infarction: nanoparticle-mediated delivery of pitavastatininduces cardioprotection from ischemia-reperfusion injury via activation of PI3K/Akt pathway and anti-inflammation in a rat model, PLoS One 10 (2015) 1–23,http://dx.doi.org/10.1371/journal.pone.0132451.

[58] G.N. Coral-Hinostroza, T. Ytrestøyl, B. Ruyter, B. Bjerkeng, Plasma appearance ofunesterified astaxanthin geometrical E/Z and optical R/S isomers in men givensingle doses of a mixture of optical 3 and 3′R/S isomers of astaxanthin fatty acyldiesters, Comp. Biochem. Physiol. - C Toxicol. Pharmacol. 139 (2004) 99–110,http://dx.doi.org/10.1016/j.cca.2004.09.011.

[59] A. Ranga Rao, R.L. Raghunath Reddy, V. Baskaran, R. Sarada, G.A. Ravishankar,Characterization of microalgal carotenoids by mass spectrometry and their bioa-vailability and antioxidant properties elucidated in rat model, J. Agric. FoodChem. 58 (2010) 8553–8559, http://dx.doi.org/10.1021/jf101187k.

[60] J.S. Coombes, J.E. Sharman, R.G. Fassett, Astaxanthin has no effect on arterialstiffness, oxidative stress, or inflammation in renal transplant recipients: a ran-domized controlled trial (the XANTHIN trial), Am. J. Clin. Nutr. 103 (2016)283–289, http://dx.doi.org/10.3945/ajcn.115.115477.

[61] M. Osterlie, B. Bjerkeng, S. Liaaen-Jensen, Accumulation of astaxanthin all- e, 9 Zand 13 Z geometrical isomers and 3 and 3′ RS optical isomers in rainbow trout (Oncorhynchus mykiss ) is selective, Nutr. Metab. (1999) 391–398.

[62] H.D. Choi, H.E. Kang, S.H. Yang, M.G. Lee, W.G. Shin, Pharmacokinetics and first-pass metabolism of astaxanthin in rats, Br. J. Nutr. 105 (2011) 220–227, http://dx.doi.org/10.1017/S0007114510003454.

[63] N. Anarjan, C.P. Tan, Developing a three component stabilizer system for produ-cing astaxanthin nanodispersions, Food Hydrocoll. 30 (2013) 437–447, http://dx.doi.org/10.1016/j.foodhyd.2012.07.002.

[64] J.S. Stewart, Å. Lignell, A. Pettersson, E. Elfving, M.G. Soni, Safety assessment ofastaxanthin-rich microalgae biomass: acute and subchronic toxicity studies in rats,Food Chem. Toxicol. 46 (2008) 3030–3036, http://dx.doi.org/10.1016/j.fct.2008.05.038.

[65] K. Vega, J. Edwards, P. Beilstein, Subchronic (13-week) toxicity and prenataldevelopmental toxicity studies of dietary astaxanthin in rats, Regul. Toxicol.Pharmacol. 73 (2015) 819–828, http://dx.doi.org/10.1016/j.yrtph.2015.10.013.

[66] S. Schneider, W. Mellert, S. Schulte, B. van Ravenzwaay, A developmental toxicitystudy of 3S, 3′S-Astaxanthin in New Zealand white rabbits, Food Chem. Toxicol.90 (2016) 95–101, http://dx.doi.org/10.1016/j.fct.2016.02.001.

[67] T. Katsumata, T. Ishibashi, D. Kyle, A sub-chronic toxicity evaluation of a natural

M. Zuluaga et al. Chemico-Biological Interactions 279 (2018) 145–158

156

astaxanthin-rich carotenoid extract of Paracoccus carotinifaciens in rats, Toxicol.Rep. 1 (2014) 582–588, http://dx.doi.org/10.1016/j.toxrep.2014.08.008.

[68] A. Satoh, S. Tsuji, Y. Okada, N. Murakami, M. Urami, K. Nakagawa, M. Ishikura,M. Katagiri, Y. Koga, T. Shirasawa, Preliminary clinical evaluation of toxicity andEfficacy of A New astaxanthin-rich haematococcus pluvialis extract, J. Clin.Biochem. Nutr. 44 (2009) 280–284, http://dx.doi.org/10.3164/jcbn.08-238.

[69] European Food Safety Authority, Scientific Opinion on the safety of astaxanthin-rich ingredients ( AstaREAL A1010 and AstaREAL L10 ) as novel food ingredients1, EFSA J. 12 (2014) 1–35, http://dx.doi.org/10.2903/j.efsa.2014.3757.

[70] D.N. Granger, P.R. Kvietys, Reperfusion injury and reactive oxygen species: theevolution of a concept, Redox Biol. 6 (2015) 524–551, http://dx.doi.org/10.1016/j.redox.2015.08.020.

[71] N.R. Madamanchi, A. Vendrov, M.S. Runge, Oxidative stress and vascular disease,Arterioscler. Thromb. Vasc. Biol. 25 (2005) 29–38, http://dx.doi.org/10.1161/01.ATV.0000150649.39934.13.

[72] D.M. Yellon, D.J. Hausenloy, Myocardial reperfusion injury, N. Engl. J. Med. 357(2007) 1121–1135, http://dx.doi.org/10.1056/NEJMra071667.

[73] H.K. Eltzschig, T. Eckle, Ischemia and reperfusion–from mechanism to translation,Nat. Med. 17 (2011) 1391–1401, http://dx.doi.org/10.1038/nm.2507.

[74] R.S. Hotchkiss, A. Strasser, J.E. Mcdunn, P.E. Swanson, Cell death in disease:mechanisms and emerging therapeutic concepts, N. Engl. J. Med. 361 (2009)1570–1583, http://dx.doi.org/10.1056/NEJMra0901217.Cell.

[75] S.W. Park, M. Kim, K.M. Brown, V.D. D'Agati, H.T. Lee, Paneth cell-derived IL-17Acauses multi-organ dysfunction after hepatic ischemia and reperfusion injury,Hepatology 53 (2011) 1662–1675, http://dx.doi.org/10.1002/hep.24253.

[76] M.R. Elliott, F.B. Chekeni, P.C. Trampont, E.R. Lazarowski, A. Kadl, S.F. Walk,D. Park, R.I. Woodson, P. Sharma, J.J. Lysiak, T.K. Harden, K.S. Ravichandran,Nucleotides released by apoptotic cells act as a find-me signal for phagocyticclearance, Nature 461 (2010) 282–286, http://dx.doi.org/10.1038/nature08296.Nucleotides.

[77] F.B. Chekeni, M.R. Elliott, J.K. Sandilos, S.F. Walk, M. Kinchen, E.R. Lazarowski,A.J. Armstrong, S. Penuela, W. Laird, G.S. Salvesen, B.E. Isakson, D. a Bayliss,S. Kodi, Pannexin 1 channels mediate “find–me” signal release and membranepermeability during apoptosis, Nature 467 (2010) 863–867, http://dx.doi.org/10.1038/nature09413.

[78] E.P. Cummins, E. Berra, K.M. Comerford, A. Ginouves, K.T. Fitzgerald,F. Seeballuck, C. Godson, J.E. Nielsen, P. Moynagh, J. Pouyssegur, C.T. Taylor,Prolyl hydroxylase-1 negatively regulates I B kinase-beta, giving insight into hy-poxia-induced NF B activity, Proc. Natl. Acad. Sci. 103 (2006) 18154–18159,http://dx.doi.org/10.1073/pnas.0602235103.

[79] G. Yilmaz, T.V. Arumugam, K.Y. Stokes, D.N. Granger, Role of T lymphocytes andinterferon-gamma in ischemic stroke, Circulation 113 (2006) 2105–2112, http://dx.doi.org/10.1161/CIRCULATIONAHA.105.593046.

[80] R. Jin, G. Yang, G. Li, Inflammatory mechanisms in ischemic stroke: role of in-flammatory cells, J. Leukoc. Biol. 87 (2010) 779–789.

[81] J.E. Freedman, Oxidative stress and platelets, Arterioscler. Thromb. Vasc. Biol. 28(2008), http://dx.doi.org/10.1161/ATVBAHA.107.159178.

[82] M.Y. Mok, C.S. Lau, The burden and measurement of cardiovascular disease in SSc,Nat. Rev. Rheumatol. 6 (2010) 430–434, http://dx.doi.org/10.1038/nrrheum.2010.65.

[83] D.D. Wagner, P.S. Frenette, The vessel wall and its interactions, Blood 111 (2008)5271–5281, http://dx.doi.org/10.1182/blood-2008-01-078204.

[84] G.Y. Chen, G. Nuñez, Sterile inflammation: sensing and reacting to damage, Nat.Rev. Immunol. 10 (2010) 826–837, http://dx.doi.org/10.1038/nri2873.

[85] M. Le Brocq, S.J. Leslie, P. Milliken, I.L. Megson, Endothelial dysfunction: frommolecular mechanisms to measurement, clinical implications, and therapeuticopportunities, Antioxid. Redox Signal 10 (2008) 1631–1674, http://dx.doi.org/10.1089/ars.2007.2013.

[86] S. Ogawa, H. Gerlach, C. Esposito, A. Pasagian-Macaulay, J. Brett, D. Stern,Hypoxia modulates the barrier and coagulant function of cultured bovine en-dothelium. Increased monolayer permeability and induction of procoagulantproperties, J. Clin. Invest. 85 (1990) 1090–1098, http://dx.doi.org/10.1172/JCI114540.

[87] S. Karbach, P. Wenzel, A. Waisman, T. Munzel, A. Daiber, eNOS uncoupling incardiovascular diseases–the role of oxidative stress and inflammation, Curr.Pharm. Des. 20 (2014) 3579–3594, http://dx.doi.org/10.1155/2014/615312.

[88] C. Ladecola, J. Anrathner, The immunology of stroke: from mechanism to trans-lation, Nat. Med 17 (2012) 796–808, http://dx.doi.org/10.1038/nm.2399.The.

[89] G. Douglas, K.M. Channon, The pathogenesis of atherosclerosis, Med. Baltim. 38(2010) 397–402, http://dx.doi.org/10.1016/j.mpmed.2010.05.002.

[90] R. Ross, Atherosclerosis-An inflammatory disease, N. Engl. J. Med. 340 (1999)115–126.

[91] A. Ranga Rao, V. Baskaran, R. Sarada, G.A. Ravishankar, In vivo bioavailabilityand antioxidant activity of carotenoids from microalgal biomass - a repeated dosestudy, Food Res. Int. 54 (2013) 711–717, http://dx.doi.org/10.1016/j.foodres.2013.07.067.

[92] H. McNulty, R.F. Jacob, R.P. Mason, Biologic activity of carotenoids related todistinct membrane physicochemical interactions, Am. J. Cardiol. 101 (2008),http://dx.doi.org/10.1016/j.amjcard.2008.02.004.

[93] W.I. Gruszecki, K. Strzałka, Carotenoids as modulators of lipid membrane physicalproperties, Biochim. Biophys. Acta 1740 (2005) 108–115, http://dx.doi.org/10.1016/j.bbadis.2004.11.015.

[94] H.P. McNulty, J. Byun, S.F. Lockwood, R.F. Jacob, R.P. Mason, Differential effectsof carotenoids on lipid peroxidation due to membrane interactions: X-ray dif-fraction analysis, Biochim. Biophys. Acta - Biomembr. 1768 (2007) 167–174,http://dx.doi.org/10.1016/j.bbamem.2006.09.010.

[95] F.J. Pashkow, D.G. Watumull, C.L. Campbell, Astaxanthin: a novel potentialtreatment for oxidative stress and inflammation in cardiovascular disease, Am. J.Cardiol. 101 (2008) 58D–68D, http://dx.doi.org/10.1016/j.amjcard.2008.02.010.

[96] Z. Deng, W.-G. Shan, S. Wang, M.-M. Hu, Y. Chen, Effects of astaxanthin on bloodcoagulation, fibrinolysis and platelet aggregation in hyperlipidemic rats, Pharm.Biol. 55 (2017) 663–672, http://dx.doi.org/10.1080/13880209.2016.1261905.

[97] S.F. Lockwood, G.J. Gross, Disodium disuccinate astaxanthin (Cardax): anti-oxidant and antiinflammatory cardioprotection, Cardiovasc. Drug Rev. 23 (2005)199–216.

[98] G.J. Gross, S.F. Lockwood, Cardioprotection and myocardial salvage by a dis-odium disuccinate astaxanthin derivative (Cardax™), Life Sci. 75 (2004) 215–224,http://dx.doi.org/10.1016/j.lfs.2003.12.006.

[99] G.J. Gross, S.F. Lockwood, Acute and chronic administration of disodium dis-uccinate astaxanthin (Cardax™) produces marked cardioprotection in dog hearts,Mol. Cell. Biochem. 272 (2005) 221–227, http://dx.doi.org/10.1007/s11010-005-7555-2.

[100] D.A. Lauver, S.F. Lockwood, B.R. Lucchesi, Disodium Disuccinate Astaxanthin(Cardax) attenuates complement activation and reduces myocardial injury fol-lowing ischemia/reperfusion, J. Pharmacol. Exp. Ther. 314 (2005) 686–692,http://dx.doi.org/10.1124/jpet.105.087114.

[101] G.J. Gross, S.L. Hazen, S.F. Lockwood, Seven day oral supplementation withCardaxTM (disodium disuccinate astaxanthin) provides significant cardioprotec-tion and reduces oxidative stress in rats, Mol. Cell. Biochem. 283 (2006) 23–30,http://dx.doi.org/10.1007/s11010-006-2217-6.

[102] R.S. Adluri, M. Thirunavukkarasu, L. Zhan, N. Maulik, K. Svennevig, M. Bagchi,G. Maulik, Cardioprotective efficacy of a novel antioxidant mix VitaePro against exvivo myocardial ischemia-reperfusion injury, Cell Biochem. Biophys. 67 (2013)281–286, http://dx.doi.org/10.1007/s12013-011-9300-7.

[103] Y.P. Lu, S.Y. Liu, H. Sun, X.M. Wu, J.J. Li, L. Zhu, Neuroprotective effect of as-taxanthin on H2O2-induced neurotoxicity in vitro and on focal cerebral ischemiain vivo, Brain Res. 1360 (2010) 40–48, http://dx.doi.org/10.1016/j.brainres.2010.09.016.

[104] D.-H. Lee, Y.J. Lee, K.H. Kwon, Neuroprotective effects of astaxanthin in oxygen-glucose deprivation in SH-SY5Y cells and global cerebral ischemia in rat, J. Clin.Biochem. Nutr. 47 (2010) 121–129, http://dx.doi.org/10.3164/jcbn.10-29.

[105] J. Li, F. Wang, Y. Xia, W. Dai, K. Chen, S. Li, T. Liu, Y. Zheng, J. Wang, W. Lu,Y. Zhou, Q. Yin, J. Lu, Y. Zhou, C. Guo, Astaxanthin pretreatment attenuates he-patic ischemia reperfusion-induced apoptosis and autophagy via the ROS/MAPKpathway in mice, Mar. Drugs 13 (2015) 3368–3387, http://dx.doi.org/10.3390/md13063368.

[106] G.D. Curek, A. Cort, G. Yucel, N. Demir, S. Ozturk, G.O. Elpek, B. Savas, M. Aslan,Effect of astaxanthin on hepatocellular injury following ischemia/reperfusion,Toxicology 267 (2010) 147–153, http://dx.doi.org/10.1016/j.tox.2009.11.003.

[107] X. Qiu, K. Fu, X. Zhao, Y. Zhang, Y. Yuan, S. Zhang, X. Gu, H. Guo, Protectiveeffects of astaxanthin against ischemia/reperfusion induced renal injury in mice, J.Transl. Med. 13 (2015) 1–9, http://dx.doi.org/10.1186/s12967-015-0388-1.

[108] T. Iwamoto, K. Hosoda, R. Hirano, H. Kurata, A. Matsumoto, W. Miki,M. Kamiyama, H. Itakura, S. Yamamoto, K. Kondo, Inhibition of low-density li-poprotein oxidation by astaxanthin, J. Atheroscler. Thromb. 7 (2000) 216–222.

[109] S. Ursoniu, A. Sahebkar, M.C. Serban, M. Banach, Lipid profile and glucosechanges after supplementation with astaxanthin: a systematic review and meta-analysis of randomized controlled trials, Arch. Med. Sci. 11 (2015) 253–266,http://dx.doi.org/10.5114/aoms.2015.50960.

[110] H. Yoshida, H. Yanai, K. Ito, Y. Tomono, T. Koikeda, H. Tsukahara, N. Tada,Administration of natural astaxanthin increases serum HDL-cholesterol and adi-ponectin in subjects with mild hyperlipidemia, Atherosclerosis 209 (2010)520–523, http://dx.doi.org/10.1016/j.atherosclerosis.2009.10.012.

[111] P.-T. Yeh, H.-W. Huang, C.-M. Yang, W.-S. Yang, C.-H. Yang, Astaxanthin inhibitsexpression of retinal oxidative stress and inflammatory mediators in streptozo-tocin-induced diabetic rats, PLoS One 11 (2016) e0146438, , http://dx.doi.org/10.1371/journal.pone.0146438.

[112] A.M. Mimoun-benarroch, J. Lallement, H. Younes, F. Depeint, Free form astax-anthin from yeast Phaffia rhodozyma fermentation reduces plasmatic triglyceridesin a pre-obesity diet-induced dyslipidaemia mouse model, J. Food Compos. Anal.(2017), http://dx.doi.org/10.1016/J.JFCA.2017.07.023.

[113] M.S.M. Al-bulish, C. Xue, M.I. Waly, J. Xu, Y. Wang, Q. Tang, The defensive role ofantioxidants astaxanthin against oxidative damage in diabetic rats injected withstreptozotocin, J. Food Nutr. Res. 5 (2017) 191–196, http://dx.doi.org/10.12691/jfnr-5-3-9.

[114] H.D. Choi, J.H. Kim, M.J. Chang, Y. Kyu-Youn, W.G. Shin, Effects of astaxanthinon oxidative stress in overweight and obese adults, Phyther. Res. 25 (2011)1813–1818, http://dx.doi.org/10.1002/ptr.3494.

[115] G.H. Kim, J.E. Kim, S.J. Rhie, S. Yoon, The role of oxidative stress in neurode-generative diseases, Exp. Neurobiol. 24 (2015) 325–340, http://dx.doi.org/10.5607/en.2015.24.4.325.

[116] H. Shen, C.-C. Kuo, J. Chou, A. Delvolve, S.N. Jackson, J. Post, A.S. Woods,B.J. Hoffer, Y. Wang, B.K. Harvey, Astaxanthin reduces ischemic brain injury inadult rats, FASEB J. 23 (2009) 1958–1968, http://dx.doi.org/10.1096/fj.08-123281.

[117] B. Grimmig, S.H. Kim, K. Nash, P.C. Bickford, R. Douglas Shytle, Neuroprotectivemechanisms of astaxanthin: a potential therapeutic role in preserving cognitivefunction in age and neurodegeneration, GeroScience 39 (2017) 19–32, http://dx.doi.org/10.1007/s11357-017-9958-x.

[118] H. Wu, H. Niu, A. Shao, C. Wu, B.J. Dixon, J. Zhang, S. Yang, Y. Wang, Astaxanthinas a potential neuroprotective agent for neurological diseases, Mar. Drugs 13(2015) 5750–5766, http://dx.doi.org/10.3390/md13095750.

M. Zuluaga et al. Chemico-Biological Interactions 279 (2018) 145–158

157

[119] B. Grimmig, J. Morganti, K. Nash, P.C. Bickford, Immunomodulators as ther-apeutic agents in mitigating the progression of parkinson's disease, Brain Sci. 6(2016) 1–12, http://dx.doi.org/10.3390/brainsci6040041.

[120] P. Lobos, B. Bruna, A. Cordova, P. Barattini, J.L. Galaz, T. Adasme, C. Hidalgo,P. Muñoz, A. Paula-Lima, Astaxanthin protects primary hippocampal neuronsagainst noxious effects of AB-oligomers, Neural Plast. 2016 (2016) 1–13, http://dx.doi.org/10.1155/2016/3456783.

[121] I. Higuera-Ciapara, L. Felix-Valenzuela, F.M. Goycoolea, W. Argüelles-Monal,Microencapsulation of astaxanthin in a chitosan matrix, Carbohydr. Polym. 56(2004) 41–45, http://dx.doi.org/10.1016/j.carbpol.2003.11.012.

[122] L. Brannon-Peppas, Recent advances on the use of biodegradable microparticlesand nanoparticles in controlled drug delivery, Int. J. Pharm.. 116(995) 1–9.doi:10.1016/0378-5173(94)00324-X.

[123] S.F. Lin, Y.C. Chen, R.N. Chen, L.C. Chen, H.O. Ho, Y.H. Tsung, M.T. Sheu,D.Z. Liu, Improving the stability of astaxanthin by microencapsulation in calciumalginate beads, PLoS One 11 (2016) 1–10, http://dx.doi.org/10.1371/journal.pone.0153685.

[124] K.G.H. Desai, H. Jin Park, Recent Developments in Microencapsulation of FoodIngredients, (2005), http://dx.doi.org/10.1081/DRT-200063478.

[125] Q. Dai, X. You, L. Che, F. Yu, C. Selomulya, X.D. Chen, An investigation in mi-croencapsulating astaxanthin using a monodisperse droplet spray dryer, Dry.Technol. 31 (2013) 1562–1569, http://dx.doi.org/10.1080/07373937.2013.800877.

[126] X. Zhang, W. Yin, Y. Qi, X. Li, W. Zhang, G. He, Microencapsulation of astaxanthinin alginate using modified emulsion technology: preparation, characterization,and cytostatic activity, Can. J. Chem. Eng. 95 (2017) 412–419, http://dx.doi.org/10.1002/cjce.22712.

[127] P. Kittikaiwan, S. Powthongsook, P. Pavasant, A. Shotipruk, Encapsulation ofHaematococcus pluvialis using chitosan for astaxanthin stability enhancement,Carbohydr. Polym. 70 (2007) 378–385, http://dx.doi.org/10.1016/j.carbpol.2007.04.021.

[128] J.-S. Lee, S.-A. Park, D. Chung, H.G. Lee, Encapsulation of astaxanthin-richXanthophyllomyces dendrorhous for antioxidant delivery, Int. J. Biol. Macromol.49 (2011) 268–273, http://dx.doi.org/10.1016/j.ijbiomac.2011.04.021.

[129] C. Bustos-Garza, J. Yáñez-Fernández, B.E. Barragán-Huerta, Thermal and pH sta-bility of spray-dried encapsulated astaxanthin oleoresin from Haematococcuspluvialis using several encapsulation wall materials, Food Res. Int. 54 (2013)641–649, http://dx.doi.org/10.1016/j.foodres.2013.07.061.

[130] F.R.S. Machado, D.F. Reis, D.L. Boschetto, J.F.M. Burkert, S.R.S. Ferreira,J.V. Oliveira, C.A. V Burkert, Encapsulation of astaxanthin from Haematococcuspluvialis in PHBV by means of SEDS technique using supercritical CO2, Ind. CropsProd. 54 (2014) 17–21, http://dx.doi.org/10.1016/j.indcrop.2014.01.007.

[131] V. Suganya, S.T. Asheeba, Microencapsulation of astaxanthin using ionotropicgelation method isolated from three crab varieties, Int. J. Curr. Pharm. Res. 7(2015) 7–10.

[132] D.J. McClements, Nanoemulsions versus microemulsions: terminology, differ-ences, and similarities, Soft Matter 8 (2012) 1719–1729, http://dx.doi.org/10.1039/C2SM06903B.

[133] W. Mehnert, K. Mader, Solid lipid nanoparticles: production, characterization andapplications, Adv. Drug Deliv. Rev. 64 (2012) 83–101, http://dx.doi.org/10.1016/j.addr.2012.09.021.

[134] L. Müller, K. Fröhlich, V. Böhm, Comparative antioxidant activities of carotenoidsmeasured by ferric reducing antioxidant power (FRAP), ABTS bleaching assay(αTEAC), DPPH assay and peroxyl radical scavenging assay, Food Chem. 129(2011) 139–148, http://dx.doi.org/10.1016/j.foodchem.2011.04.045.

[135] R.H. Müller, M. Radtke, S.A. Wissing, Nanostructured lipid matrices for improvedmicroencapsulation of drugs, Int. J. Pharm. 242 (2002) 121–128, http://dx.doi.org/10.1016/S0378-5173(02)00180-1.

[136] C.C. Müller-Goymann, Physicochemical characterization of colloidal drug deliverysystems such as reverse micelles, vesicles, liquid crystals and nanoparticles fortopical administration, Eur. J. Pharm. Biopharm. 58 (2004) 343–356, http://dx.doi.org/10.1016/j.ejpb.2004.03.028.

[137] J. Varshosaz, S. Eskandari, M. Tabakhian, Production and optimization of valproicacid nanostructured lipid carriers by the Taguchi design, Pharm. Dev. Technol. 15(2010) 89–96, http://dx.doi.org/10.3109/10837450903013568.

[138] P. Barros, E. Pinto, P. Colepicolo, M. Pedersen, Astaxanthin and peridinin inhibitoxidative damage in Fe2+-Loaded liposomes: scavenging oxyradicals or changingmembrane permeability, Biochem. Biophys. Res. Commun. 288 (2001) 225–232.

[139] C.H. Peng, C.H. Chang, R.Y. Peng, C.C. Chyau, Improved membrane transport ofastaxanthine by liposomal encapsulation, Eur. J. Pharm. Biopharm. 75 (2010)154–161, http://dx.doi.org/10.1016/j.ejpb.2010.03.004.

[140] F. Acevedo, M. Rubilar, I. Jofré, M. Villarroel, P. Navarrete, M. Esparza,F. Romero, E.A. Vilches, V. Acevedo, C. Shene, Oil bodies as a potential micro-encapsulation carrier for astaxanthin stabilisation and safe delivery, J.Microencapsul. 31 (2014) 488–500, http://dx.doi.org/10.3109/02652048.2013.879931.

[141] C. Kamezaki, A. Nakashima, A. Yamada, S. Uenishi, H. Ishibashi, N. Shibuya,S. Hama, S. Hosoi, E. Yamashita, K. Kogure, Synergistic antioxidative effect ofastaxanthin and tocotrienol by co-encapsulated in liposomes, J. Clin. Biochem.Nutr. 59 (2016) 100–106, http://dx.doi.org/10.3164/jcbn.15-153.

[142] A. Bustamante, L. Masson, J. Velasco, J. Manuel, P. Robert, Microencapsulation ofH. pluvialis oleoresins with different fatty acid composition : kinetic stability ofastaxanthin and alpha-tocopherol, Food Chem. 190 (2016) 1013–1021, http://dx.doi.org/10.1016/j.foodchem.2015.06.062.

[143] J.M. Odeberg, Å. Lignell, A. Pettersson, P. Höglund, Oral bioavailability of theantioxidant astaxanthin in humans is enhanced by incorporation of lipid basedformulations, Eur. J. Pharm. Sci. 19 (2003) 299–304, http://dx.doi.org/10.1016/S0928-0987(03)00135-0.

[144] H.S. Ribeiro, L.G. Rico, G.G. Badolato, H. Schubert, Production of O/W emulsionscontaining astaxanthin by repeated premix membrane emulsification, Food Eng.Phys. Prop. 70 (2005) 117–123.

[145] A. Tachaprutinun, T. Udomsup, C. Luadthong, S. Wanichwecharungruang,Preventing the thermal degradation of astaxanthin through nanoencapsulation,Int. J. Pharm. 374 (2009) 119–124, http://dx.doi.org/10.1016/j.ijpharm.2009.03.001.

[146] N. Anarjan, I.A. Nehdi, C.P. Tan, Influence of astaxanthin, emulsifier and organicphase concentration on physicochemical properties of astaxanthin nanodisper-sions, Chem. Cent. J. 7 (2013) 1–11, http://dx.doi.org/10.1186/1752-153X-7-127.

[147] N. Anarjan, S.I. Al-resayes, C.P. Tan, Effects of homogenization process parameterson physicochemical properties of astaxanthin nanodispersions prepared using asolvent-diffusion technique, Int. J. Nanomedicine 10 (2015) 1109–1118, http://dx.doi.org/10.2147/IJN.S72835.

[148] F. Tamjidi, M. Shahedi, J. Varshosaz, A. Nasirpour, Design and characterization ofastaxanthin-loaded nanostructured lipid carriers, Innov. Food Sci. Emerg. Technol.26 (2014) 366–374, http://dx.doi.org/10.1016/j.ifset.2014.06.012.

[149] M.M.R. Meor Mohd Affandi, T. Julianto, A. Majeed, Development and stabilityevaluation of Astaxanthin nanoemulsion, Asian J. Pharm. Clin. Res. 4 (2011)143–148.

[150] C.H. Chiu, C.C. Chang, S.T. Lin, C.C. Chyau, R.Y. Peng, Improved hepatoprotectiveeffect of liposome-encapsulated astaxanthin in lipopolysaccharide-induced acutehepatotoxicity, Int. J. Mol. Sci. 17 (2016), http://dx.doi.org/10.3390/ijms17071128.

[151] G. Astray, C. Gonzalez-Barreiro, J.C. Mejuto, R. Rial-Otero, J. Simal-Gándara, Areview on the use of cyclodextrins in foods, Food Hydrocoll. 23 (2009)1631–1640, http://dx.doi.org/10.1016/j.foodhyd.2009.01.001.

[152] H. Dodziuk, Modeling of CyDs and their complexes, in: cyclodextrins their com-plexes, Chem. Anal. Methods. Appl. (2006) 333–355, http://dx.doi.org/10.1002/3527608982.ch11.

[153] D. Duchene, D. Wouessidjewe, Uses of cyclodextrins, Drug Dev. Ind. Pharm. 16(1990) 2487–2499.

[154] E.M.M. Del Valle, Cyclodextrins and their uses: a review, Process Biochem. 39(2004) 1033–1046, http://dx.doi.org/10.1016/S0032-9592(03)00258-9.

[155] S. Dong, Y. Huang, R. Zhang, Z. Lian, S. Wang, Y. Liu, Inclusion complexes ofastaxanthin with hydroxypropyl-β-cyclodextrin: parameters optimization, spec-troscopic profiles, and properties, Eur. J. Lipid Sci. Technol. 116 (2014) 978–986,http://dx.doi.org/10.1002/ejlt.201300261.

[156] S.F. Lockwood, S.O. Malley, G.L. Mosher, Improved aqueous solubility of crys-talline astaxanthin (sulfobutyl ether β-cyclodextrin), J. Pharm. Sci. 92 (2003)922–926, http://dx.doi.org/10.1002/jps.10359.

[157] S. Kim, E. Cho, J. Yoo, E. Cho, S. Ju Choi, S.M. Son, J.M. Lee, M.-J. In, D.C. Kim, J.-H. Kim, H.J. Chae, B-CD-mediated encapsulation enhanced stability and solubilityof Astaxanthin, J. Appl. Biol. Chem. 53 (2010) 559–565, http://dx.doi.org/10.3839/jksabc.2010.086.

[158] X. Chen, R. Chen, Z. Guo, C. Li, P. Li, The preparation and stability of the inclusioncomplex of astaxanthin with β-cyclodextrin, Food Chem. 101 (2007) 1580–1584,http://dx.doi.org/10.1016/j.foodchem.2006.04.020.

[159] P. Nalawade, A. Gajjar, Assessment of in-vitro bio accessibility and characteriza-tion of spray dried complex of astaxanthin with methylated betacyclodextrin, J.Incl. Phenom. Macrocycl. Chem. 83 (2015) 63–75, http://dx.doi.org/10.1007/s10847-015-0541-8.

M. Zuluaga et al. Chemico-Biological Interactions 279 (2018) 145–158

158