Effects of Antioxidant Supplements on the Survival and...

17
Review Article Effects of Antioxidant Supplements on the Survival and Differentiation of Stem Cells Sara Shaban, 1 Mostafa Wanees Ahmed El-Husseny, 1,2 Abdelrahman Ibrahim Abushouk, 2,3,4 Amr Muhammad Abdo Salem, 2,3 Mediana Mamdouh, 1 and Mohamed M. Abdel-Daim 5,6 1 Faculty of Medicine, Fayoum University, Cairo, Egypt 2 NovaMed Medical Research Association, Cairo, Egypt 3 Faculty of Medicine, Ain Shams University, Cairo, Egypt 4 Medical Research Group of Egypt, Cairo, Egypt 5 Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt 6 Department of Ophthalmology and Micro-Technology, Yokohama City University, Yokohama, Japan Correspondence should be addressed to Mohamed M. Abdel-Daim; [email protected] Received 25 February 2017; Accepted 31 May 2017; Published 9 July 2017 Academic Editor: Andreas Daiber Copyright © 2017 Sara Shaban et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Although physiological levels of reactive oxygen species (ROS) are required to maintain the self-renewal capacity of stem cells, elevated ROS levels can induce chromosomal aberrations, mitochondrial DNA damage, and defective stem cell dierentiation. Over the past decade, several studies have shown that antioxidants can not only mitigate oxidative stress and improve stem cell survival but also aect the potency and dierentiation of these cells. Further benecial eects of antioxidants include increasing genomic stability, improving the adhesion of stem cells to culture media, and enabling researchers to manipulate stem cell proliferation by using dierent doses of antioxidants. These ndings can have several clinical implications, such as improving neurogenesis in patients with stroke and neurodegenerative diseases, as well as improving the regeneration of infarcted myocardial tissue and the banking of spermatogonial stem cells. This article reviews the cellular and molecular eects of antioxidant supplementation to cultured or transplanted stem cells and draws up recommendations for further research in this area. 1. Introduction Stem cells are undierentiated cells, characterized by self- renewal and the ability to dierentiate into several cell types (potency) [1]. They can be totipotent (dierentiating into embryonic and extraembryonic cell types), pluripotent (dierentiating into cells of the three germ layers), or mul- tipotent (dierentiating into cells of a closely related family) [2]. Stem cell research runs with an incredible speed and its applications are under investigation in dierent medical elds [3, 4]. There are two main types of stem cells: embry- onic stem cells (ESCs) (present in the inner cell mass of the blastocyst) and adult stem cells (present in dierent mature tissues to replace dead cells) [5, 6]. Induced pluripotent stem cells (iPSCs) are adult cells, genetically reprogrammed to express genes and factors, required for maintaining the properties of ESCs. However, the reprogramming process itself results in oxidative stress by generating high levels of reactive oxygen species (ROS) [7, 8], which cause damage to DNA, RNA, and cell proteins and may induce apoptosis [911]. However, ROS are required in physiological levels to maintain the self-renewal capacity of stem cells and to ght invading microbes [1114]. Antioxidants are biochemical supplements that protect cellular constituents from oxidative stress by neutralizing free radicals and terminating the oxidative reaction chain in the mitochondrial membrane [15]. They can be classi- ed into enzymatic and nonenzymatic, endogenous and Hindawi Oxidative Medicine and Cellular Longevity Volume 2017, Article ID 5032102, 16 pages https://doi.org/10.1155/2017/5032102

Transcript of Effects of Antioxidant Supplements on the Survival and...

Review ArticleEffects of Antioxidant Supplements on the Survival andDifferentiation of Stem Cells

Sara Shaban,1 Mostafa Wanees Ahmed El-Husseny,1,2 Abdelrahman Ibrahim Abushouk,2,3,4

Amr Muhammad Abdo Salem,2,3 Mediana Mamdouh,1 and Mohamed M. Abdel-Daim5,6

1Faculty of Medicine, Fayoum University, Cairo, Egypt2NovaMed Medical Research Association, Cairo, Egypt3Faculty of Medicine, Ain Shams University, Cairo, Egypt4Medical Research Group of Egypt, Cairo, Egypt5Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt6Department of Ophthalmology and Micro-Technology, Yokohama City University, Yokohama, Japan

Correspondence should be addressed to Mohamed M. Abdel-Daim; [email protected]

Received 25 February 2017; Accepted 31 May 2017; Published 9 July 2017

Academic Editor: Andreas Daiber

Copyright © 2017 Sara Shaban et al. This is an open access article distributed under the Creative Commons Attribution License,which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Although physiological levels of reactive oxygen species (ROS) are required to maintain the self-renewal capacity of stem cells,elevated ROS levels can induce chromosomal aberrations, mitochondrial DNA damage, and defective stem cell differentiation.Over the past decade, several studies have shown that antioxidants can not only mitigate oxidative stress and improve stem cellsurvival but also affect the potency and differentiation of these cells. Further beneficial effects of antioxidants include increasinggenomic stability, improving the adhesion of stem cells to culture media, and enabling researchers to manipulate stem cellproliferation by using different doses of antioxidants. These findings can have several clinical implications, such as improvingneurogenesis in patients with stroke and neurodegenerative diseases, as well as improving the regeneration of infarctedmyocardial tissue and the banking of spermatogonial stem cells. This article reviews the cellular and molecular effects ofantioxidant supplementation to cultured or transplanted stem cells and draws up recommendations for further research in this area.

1. Introduction

Stem cells are undifferentiated cells, characterized by self-renewal and the ability to differentiate into several celltypes (potency) [1]. They can be totipotent (differentiatinginto embryonic and extraembryonic cell types), pluripotent(differentiating into cells of the three germ layers), or mul-tipotent (differentiating into cells of a closely related family)[2]. Stem cell research runs with an incredible speed and itsapplications are under investigation in different medicalfields [3, 4]. There are two main types of stem cells: embry-onic stem cells (ESCs) (present in the inner cell mass of theblastocyst) and adult stem cells (present in different maturetissues to replace dead cells) [5, 6].

Induced pluripotent stem cells (iPSCs) are adult cells,genetically reprogrammed to express genes and factors,required for maintaining the properties of ESCs. However,the reprogramming process itself results in oxidative stressby generating high levels of reactive oxygen species (ROS)[7, 8], which cause damage to DNA, RNA, and cell proteinsand may induce apoptosis [9–11]. However, ROS arerequired in physiological levels to maintain the self-renewalcapacity of stem cells and to fight invading microbes [11–14].

Antioxidants are biochemical supplements that protectcellular constituents from oxidative stress by neutralizingfree radicals and terminating the oxidative reaction chainin the mitochondrial membrane [15]. They can be classi-fied into enzymatic and nonenzymatic, endogenous and

HindawiOxidative Medicine and Cellular LongevityVolume 2017, Article ID 5032102, 16 pageshttps://doi.org/10.1155/2017/5032102

exogenous [16], and water-soluble (reacting with oxidantsin the cytosol or plasma) and lipid-soluble antioxidants(preventing lipid peroxidation of cell membranes) [17].

Over the past decade, several studies have shown thatantioxidants can not only mitigate oxidative stress andimprove stem cell survival but also affect the potency and dif-ferentiation of these cells. In our article, we reviewed theresults of preclinical studies that investigated the effects ofantioxidants on cultured or transplanted stem cells in anattempt to draw up recommendations for further researchin this area.

2. Induced Pluripotent Stem Cells (iPSCs)

As highlighted earlier, the reprogramming of iPSCs is associ-ated with generation of high ROS levels. Several reportsshowed that, in comparison to somatic precursor cells, iPSCsexhibit the following criteria: (1) marked protection againstnuclear and mitochondrial DNA (mtDNA) damage and (2)significantly lower levels of ROS due to upregulation ofintrinsic antioxidant enzymes [18, 19]. Dannenmann et al.found a 10-fold decrease in ROS level and a fourfold increaseof glutathione (GSH) and glutathione reductase (GR) levelsin iPSCs, compared to fibroblasts [18]. In another study bythe same authors, they showed that several glutathione S-transferases (GSTs), which act as antioxidant and detoxifyingenzymes, were upregulated in iPSCs, compared to theirsomatic precursor cells [19].

Ji and colleagues reported that mitigation of oxidativestress during cellular reprogramming by antioxidant sup-plementation protects the genome of reprogramming cellsagainst DNA damage and leads to iPSCs with fewer geno-mic aberrations [20]. In the same vein, Luo and colleagues[21] found that iPSCs grew well and “stemness” was pre-served for up to two months after the addition of a low-dose antioxidant supplement. Moreover, using comparativegenomic hybridization (CGH) analysis, they showed thatantioxidant supplementation lowered the levels of geneticaberrations in cultured iPSCs [21].

Hämäläinen and colleagues showed that the repro-gramming and self-renewal abilities of iPSCs were dimin-ished after subtle increases in ROS levels, originating frommtDNA mutagenesis. However, the addition of two differentantioxidants [N-acetyl-L-cysteine (NAC) and mitochondria-targeted ubiquinone (MitoQ)] efficiently rescued these abili-ties in mutator iPSCs [22]. N-acetyl-L-cysteine raises cellularGSH pool and promotes the processing of H2O2 in the cyto-sol [23], whereas MitoQ acts upstream to prevent superoxideproduction within the mitochondria before H2O2 generation[24]. Of note, Hämäläinen et al. highlighted that the thera-peutic window of MitoQ for iPSCs is narrow, while highconcentrations of NAC were not associated with toxic effectson iPSCs [22].

Interestingly, other reports showed no effect of antiox-idant supplementation on the expression of 53BP1 andATM proteins (two molecules involved in DNA repairpathways) [25–27]. Recently, it has been found thathigh-dose antioxidants downregulates DNA repair-relatedkinases, which conversely results in genomic instability of

iPSCs [21]. Therefore, adjusting the dose of supplementaryantioxidants is critical.

3.BoneMarrow-DerivedMesenchymal (BMSCs)and Hematopoietic Stem Cells (HSCs)

Several studies showed that the ex vivo expansion of ESCsand mesenchymal stem cells (MSCs) [28–31] and thein vitro expansion of HSCs [32] may cause genomic insta-bility. Through a serial transplantation assay, Jang andcolleagues showed that elevated ROS levels reduce theself-renewal ability of HSCs [33]. Therefore, decreasingO2 concentrations to physiological levels or adding properdosages of antioxidants can reduce in vitro, culture-stimulated aneuploidy, providing potential methods tolimit genomic alterations when expanding HSCs in vitro[32, 34, 35]. Hamid et al. conducted an in vitro study toevaluate the antioxidant effects of Hibiscus sabdariffa L.(roselle) on bone marrow-derived HSCs. They showed thatroselle supplementation increased superoxide dismutase(SOD) expression (at 125, 500, and 1000 ng/mL) andHSCs survival (at 500 and 1000ng/mL) and protectedagainst H2O2-induced DNA damage [36].

In another study by Halabian et al., treatment of BMSCswith Lipocalin-2 (Lcn2), a natural cytoprotective factor gen-erated upon exposure to stressful conditions, increased cellu-lar resistance against oxidative, hypoxic, and serumdeprivation stresses. Moreover, Lcn2-treated cells showedSOD gene upregulation, increased proliferation, maintainedpluripotency, and improved cellular adhesion to culturemedia upon H2O2 exposure, in comparison to untreated cells[37]. Similarly, Fan and colleagues studied different methodsfor isolation of BMSCs, aiming at reducing the number ofchromosomal abnormalities in isolated cells. They reportedthat culturing isolated BMSCs at a low O2 concentration(2%) or with antioxidant (NAC) supplementation increasedcellular proliferation and genomic stability, in comparisonto cultured cells at normoxic concentrations (20% O2) [38].

Another study by Choi et al. demonstrated that addingascorbic acid 2-phosphate (AAP) at different concentrationscan influence the fate of BMSCs, that is, AAP significantlyincreased osteogenic differentiation at 50mM concentration,while a significant induction of adipogenic differentiationwith oil droplet formation was noted at concentrations of250mM and higher [39].

4. Cardiomyoblasts and Vascular Progenitor Cells

According to Li and colleagues, culturing cardiac stem cellswith antioxidant increased the number and severity of cyto-genic abnormalities. This could be explained by the excessivedecrease in ROS to subphysiological levels, which may down-regulate DNA repair enzymes [40]. In another study byRodriguez-Porcel et al., modulation of the microenviron-ment, using antioxidants, leads to a higher rate of cardiomyo-blast survival, early after transplantation to the myocardiumof small animals [41]. Therefore, oxidative stress blockademay provide a favorable microenvironment for stem cells’engraftment and survival in the heart [42].

2 Oxidative Medicine and Cellular Longevity

Song and colleagues reported increased ROS productionduring differentiation of human ESCs into vascular progeni-tor cells (CD34+ cells) due to increased activity of NADPHoxidase-4 (Nox4) enzyme. They found that moderate ROSscavenging, using selenium, enhanced the vascular differenti-ation of human ESCs, while complete ROS scavenging, usingNAC, totally inhibited the vascular differentiation of thesecells. This confirms that a minimal level of ROS is requiredfor vascular stem cell differentiation to occur [43].

5. Neural Stem Cells (NSCs)

Neural stem cells are multipotent stem cells that have beensuggested as a therapeutic agent to enhance the recovery ofinjured tissues in neuroinflammatory diseases [44]. Parkand colleagues tested the effects of GV1001, a novel antioxi-dant agent, derived from human telomerase reverse tran-scriptase, on in vitro-cultured mouse NSCs. They showedthat GV1001 treatment attenuated the effects of H2O2 expo-sure, reduced lipid peroxidation and mtDNA mutation, andinduced the expression of survival-related proteins [45].Hachem et al. reported that treatment of NSCs, isolated fromthe spinal cords of transgenic mice, with brain-derived neu-rotrophic factor improved cell viability by increasing thelevels of GR and SOD enzymes; however, it had no effecton cellular proliferation [46].

Nitric oxide (NO) and nitric oxide synthase (NOS)-dependent signaling pathways have been implicated in differ-ent neurodegenerative diseases [47, 48]. Moreover, NO levelswere linked to neural precursor cell (NPC) survival and cellfate determination [49], that is, elevated levels of NO sup-press NSC proliferation and enhance differentiation of NPCsinto astrocytes [50, 51]. Melatonin is a hormone synthesizedin the pineal gland [52] with indirect antioxidant abilitiesthrough induction of antioxidant enzymes [53] and inhibit-ing NO production in glial cultures through p38 inhibition[54]. It has been shown to protect NSCs against lipopolysac-charide- (LPS-) induced inflammation [52]. Moreover, Negiet al. demonstrated that melatonin mitigates neuroinflamma-tion and oxidative stress via upregulating nuclear factor(erythroid-derived 2) (Nrf2) [55], a transcription factorwhich stimulates the PI3K-Akt survival signaling pathway[56, 57] and increases the expression of the antioxidantenzyme heme oxygenase-1 (HO-1) [55].

To test the effects of in vitro antioxidant supplementa-tion, Petro et al. divided male rats with experimentallyinduced thromboembolic stroke into four groups: normalrats, untreated rats with stroke, rats receiving tissue plasmin-ogen activator (tPA) only, and rats receiving tPA+CAT/SOD (loaded on nanoparticles) at three hours post stroke.Two days later, brain tissue samples were harvested foranalysis. Brain sections from the untreated group showedevidence of NSC migration through the rostral migratorystream (through detection of NSCs markers, such as nes-tin, GFAP, and SOX2), confirming the occurrence of neu-rogenesis following stroke. However, brain tissue samplesfrom the tPA-alone group showed reduction in NSCsmigration, indicating that tPA treatment suppresses neuro-genesis, either directly or through reperfusion-induced

ROS generation injury. Interestingly, tPA+Nano-CAT/SOD treatment restored and significantly increased NSCsmigration [58].

6. Human Adipose-Derived Stem Cells (ADSCs)

Adipose-derived stem cells are multipotent stem cells thatcan be isolated from the human adipose tissue and are capa-ble of in vitro expansion. Sun and colleagues reported thatboth hypoxia and antioxidants promoted ADSCs prolifera-tion by raising the number of cells in the S phase, but themaximal increase in cell number was produced in the pres-ence of antioxidants [59]. Hypoxia is believed to influencethe secretion of several growth factors [60, 61], such asinsulin-like growth factor and hepatocyte growth factor[62], while antioxidants increase the expression of stemnessgenes (CDK2, CDK4, and CDC2) and the differentiationpotential of ADSCs [59]. Another study by Higuchi et al.found that lentivirus-mediated NADPH oxidase-4 (Nox-4)overexpression did not increase ROS production in insulin,dexamethasone, indomethacin, and 3-isobutyl-1-methylxan-thine (IDII)-stimulated ADSCs [63]. This finding was laterexplained by the increased expression of endogenous antiox-idants, such as SOD and CAT during adipogenesis [63, 64].

Yang et al. showed that treatment of ADSCs with fullerol(a polyhydroxylated fullerene) potentiated the expression ofthe transcription factor FoxO1 and its downstream genes,such as Runx2 and SOD2. Moreover, it enhanced the osteo-genic activity of ADSCs, as evidenced by increased minerali-zation and expression of osteogenic markers (Runx2, OCN,and alkaline phosphatase) [65]. Wang and colleagues showedthat pretreatment of ADSCs with NAC (3mM) or AAP(0.2mM) for 20 hours suppressed advanced glycosylationend product- (AGE-) induced apoptosis via a microRNA-dependent mechanism by inhibiting AGE-induced overex-pression of miRNA-223: a key modulator of intracellularapoptotic signaling [66].

7. HumanPeriodontal Ligament Cells (hPDLCs)

In a recent study, Chung and colleagues showed that treatinghPDLCs with deferoxamine (DFO), an iron chelator, resultsin a dose-dependent elevation in ROS levels, 24 hours aftertreatment [67]. The same finding was reported in rabbit car-diomyocytes [68] and normal human hepatocytes [69]. How-ever, DFO has the ability to act on Nrf2, increasing its nucleartranslocation and the expression of its target genes, includingGST and glutamate cysteine ligase (GCL) [67]. Therefore,DFO has both beneficial (Nrf2-mediated antioxidant effect)and cytotoxic (increased ROS levels) effects. GSH depletion,using buthionine sulfoximine (BSO) and diethyl maleate(DEM), was shown to inhibit DFO-stimulated hPDLC differ-entiation into osteoblasts [67]. Moreover, GSH depletion wasalso reported to repress myogenic differentiation of murineskeletal muscle (C2C12) cells [70] and phorbol-12-myris-tate-13-acetate- (PMA-) stimulated differentiation of humanmyeloid cell line (HL-60) [71].

3Oxidative Medicine and Cellular Longevity

Table1:Summaryof

theresults

ofin

vitroandin

vivo

stud

ieson

treatm

entof

stem

cells

withantioxidantsupp

lements.

Stud

yID

Antioxidant

(dose)

Stem

celltype

(sou

rce)

Find

ings

Possiblemechanism

s

Jietal.[20]

N-A

cetyl-L-cysteine

(NAC)andvitamin

C.

Indu

cedpluripotentstem

cells

(iPSC

s)generatedfrom

human

neon

atalforeskin

fibroblasts.

Incells,infectedwithreprogramming

factors(retrovirusesencoding

human

OCT4,SO

X2,KLF

4,and

c-MYC),supp

lementation

ofthe

cultu

remediawithNACsignificantly

increasediPSC

ssurvivalandredu

ced

ROSgeneration

andthenu

mber

ofDNAdo

uble-strandedbreaks

inthereprogrammed

cells.

Antioxidantssignificantlyredu

ced

ROSgeneration

andthenu

mber

ofcopy-num

bervariations

(CNVs:

anindication

ofgeno

mic

aberration

s)in

treatediPSC

s,comparedto

theun

treatedcontrol

grou

p(p

<002).Treatmentwith

NAChadno

effecton

transgene

expression

,silencing,andviral

transduction

efficiency.

Luoetal.[21]

Hom

emadeantioxidant

cocktail[ascorbate,

glutathion

e,and

α-tocop

herolat20

mM,

4mM,and

1mM,resp.].

Twohu

man

celllin

esof

iPSC

s(201B7and253G

1).

(i)Measurementof

cellu

larROSlevels

show

eddiminishedROSlevelsin

cells,

cultu

redwithantioxidants,com

pared

totheun

treatedgrou

p.(ii)Moreover,theaddition

ofho

mem

ade

antioxidantcocktailredu

cedthenu

mber

ofgeno

micaberration

sin

treatedcells.

(i)The

compo

nentsof

homem

ade

cocktailexertedafree-radical

scavenging

activity

toneutralize

ROSin

treatedcells.

(ii)Fo

rtwomon

thsof

cultu

ring

withlowdo

sesof

antioxidants,

iPSC

smaintainedtheexpression

ofstem

ness-related

genes(O

ct3/4,

Nanog,SSE

A-4,and

ALP

).

Ham

idetal.[36]

Roselle(H

ibiscussabdariffaL.)

at125,500,or

1000

ng/m

L.

Bon

emarrow-derived

hematop

oieticstem

cells

(HSC

s)from

murinebone

marrow.

(i)Add

ingroselle

(at500and1000

ng/m

L)significantlyincreasedthesurvivalof

HSC

sandprotectedthem

against

H2O

2-indu

cedDNAdamage.

(ii)Rosellesupp

lementation

was

geno

protective,asevidencedby

the

nonrem

arkabledifference

onthe

percentage

oftailDNA,com

paredto

thecontrolgroup

(untreated

BMSC

s).

Com

paredto

thecontrolgroup

,roselle

enhanced

theactivity

ofSO

Din

HSC

s(at125,500,and

1000

ng/m

L)withasignificant

increase

inGSH

level(p<005).

How

ever,there

was

nodifference

inROSlevelsbetweenroselle-

treatedandcontrolgroup

s.

Ikedaetal.[77]

Poly(ethylene

glycol)-b-po

ly[4-(2,2,6,6-tetram

ethylpiperidine-

1-oxyl)amino-methylstyrene]

(PEG-b-PMNT).

Hem

atop

oieticstem

cells

(HSC

s)from

micefetalliver

cells.

Ikedaetal.d

esignedabiocom

patible

cellcultu

resurfacethat

canbe

used

during

exvivo

cultu

ring

and

expansionof

HSC

s.Thisnew

surfacehasseveraladvantages,

comparedto

thecurrently

used

oneinclud

inglowmolecular

weight

andantioxidantsupp

lementation

.ItdecreasedROSprod

uction

,inhibitedapop

tosis,andincreased

thepu

rity

ofseparatedcells.

The

antioxidantcultu

resurface

(PEG-b-PMNT)scavengednitric

oxideradicalsandredu

ced

oxidativemem

branedamage

witho

utchanging

the

mitocho

ndrialmem

brane

potentialb

ecause

itisno

tinternalized

withinthecellas

theconvention

alLM

Wsystem

s.

4 Oxidative Medicine and Cellular Longevity

Table1:Con

tinu

ed.

Stud

yID

Antioxidant

(dose)

Stem

celltype

(sou

rce)

Find

ings

Possiblemechanism

s

Liuetal.[32]

N-A

cetyl-L-cysteine

(NAC)at

0.1to

1μM.

LSKcells

(Lin−Sca-1+

c-Kit+,a

popu

lation

enriched

withHSC

s).

(i)Aneup

loidy/chromosom

alinstability

during

invitroexpansionof

HSC

sis

ROS-mediatedandcanbe

minim

ized

bymaintaining

ahypo

xiccond

ition

(3%

O2)du

ring

cellcultu

ring.

(ii)Similarly,NACadministration

significantlyredu

cedthepercentage

ofaneuploidy

inHSC

s,cultu

red

underno

rmoxiccond

itions,onlyat

lowdo

sages(0.1μM).

(i)Atop

timum

concentrations,

NACsignificantlyredu

ced

oxidativedamagedu

eto

itsROS-

scavenging

activity.

(ii)Moreover,hypo

xiccond

itions

andNACredu

cedthepercentage

ofaneuploidy

inboth

youn

gandoldaged

stem

cells.

Halabianetal.[37]

Lipo

calin

-2(Lcn2),a

natural

cytoprotective

factor,generated

withinthecellup

onexpo

sure

tostressfulcon

dition

s.

Bon

emarrow-derived

stem

cells

(BMSC

s)from

ratbone

marrow

(4–6

weeks

old).

(i)Lcn2

-expressingBMSC

sshow

eda

morepo

tent

defenseagainstH

2O2,

hypo

xia,andserum

deprivation

stresses,com

paredto

controlM

SCs

(ii)Moreover,Lcn2

expression

inMSC

sincreasedcellproliferation

and

adhesion

tocultu

remediaby

45%

onH

2O2expo

sure,com

paredto

controlM

SCs.

(iii)

Lcn2

-expressingBMSC

sshow

edno

rmalmultipo

tencyinto

different

celllin

eage

withmild

potentiation

ofadipogeniclin

eage

comparedto

thecontrolgroup

.

(i)The

antioxidanteffectof

Lcn2

expression

isdu

eto

ROS

scavenging

activity,associated

withup

regulation

ofantioxidant

enzymes’genes,suchas

SOD.

(ii)Thisantioxidanteffectis

associated

withan

antiapop

totic

prop

erty

asindicatedby

the

diminishednu

mberof

apop

totic

cells

onoxidativestressexpo

sure,

comparedto

thecontrolgroup

.

Fanetal.[38]

Alpha-phenyl-t-bu

tyln

itrone

(PBN)at

800μM

andNAC

at5mM.

Mesenchym

alstem

cells

(MSC

s)from

miceem

bryos.

(i)CulturedMSC

swithextracellular

matrixfrom

mou

seem

bryonic

fibroblast(M

EF-ECM)un

derhypo

xic

cond

itions

(2%

O2)show

edgreater

proliferation

,low

ergeneration

ofROS,andincreasedchromosom

alstability

comparedto

thecontrol

grou

p,cultu

redon

plasticplates

underno

rmoxiccond

itions.

(ii)Tofurtherdiminishchromosom

alinstability

andbasedon

theincreased

DNAdamageon

H2O

2expo

sure,

antioxidantsupp

lementation

tothe

cultu

remediasignificantlyredu

ced

thenu

mberof

DNAmicronu

cleiand

karyotypingabno

rmalities.The

use

ofboth

(antioxidantsandMEF-ECM)

intheinitialisolation

ofcells

from

the

Altho

ughauthorsdidno

tinvestigate

theun

derlying

mechanism

sfor

antioxidants’effects,theysuggested

that

theirfind

ings

canbe

attributed

totheability

ofboth

PBN

andNAC

totrap

free

radicals.M

oreover,NAC

serves

asaprecursorforglutathion

e,an

intracellularantioxidantmolecule.

5Oxidative Medicine and Cellular Longevity

Table1:Con

tinu

ed.

Stud

yID

Antioxidant

(dose)

Stem

celltype

(sou

rce)

Find

ings

Possiblemechanism

s

marrowincreasedpu

rity

andno

rmal

cellkaryotyping.

Wangetal.[78]

2-Vinyl-8-hydroxyqu

inoline

derivatives.

Mesenchym

alstem

cells

(MSC

s)from

ratbone

marrow.

Ingeneral,2-vinyl-8-hydroxyquino

line

derivativeshadapo

sitive

effecton

MSC

sproliferation

inado

se-

depend

entmanner.

2-Vinyl-8-hydroxyqu

inoline

derivativesareph

enol

compo

unds

that

perform

theirantioxidant

activity

throughreaction

oftheir

hydroxylgrou

pwithfree

radicals.

Cho

ietal.[39]and

Mekalaetal.[79]

Ascorbicacid-2-pho

sphate

(AAP)at

0,5,50

250,500mM.

Mesenchym

alstem

cells

(MSC

s)from

adulthu

man

bone

marrow

[39]

andhu

man

umbilicalcord

blood-derivedstem

cells

(hUCB-SCs)from

umbilical

vein

[79].

(i)Ascorbicacid

significantlyincreasedthe

proliferation

ofMSC

s/hU

CB-SCs,compared

tothecontrolgroup

(withthehighest

proliferation

rateat

250mM).Ithadno

effecton

cellu

larantigenicexpression

and

differentiation.

(ii)Moreover,AAPsignificantlyincreased

osteogenicdifferentiationat

50mM

(highest

calcium

depo

sition

atthisconcentration).In

contrast,a

significant

indu

ctionof

adipogenic

differentiationwithoild

ropletsform

ation

was

notedat

250mM

andhigher.

(i)AAPim

proved

theam

ount

ofcollagenprod

uction

percelland

increasedtheam

ount

ofcalcium

(at50

mM)andoild

eposition

(at≥2

50mM),enhancingMSC

s/hU

CB-SCsdifferentiation.

(ii)The

positive

effectof

AAP

oncellu

larproliferation

isdo

se-

depend

ent(highestat

250mM

anddecreaseswithhigher

doses

dueto

inhibitory

effecton

glycosam

inoglycanform

ation).

Koetal.[80]

PEG-catalase(200

μg/mL)

andNAC(1mM).

Hum

anum

bilicalcord

blood-

derivedstem

cells

(hUCB-SCs)

from

umbilicalvein.

(i)Exposureto

geno

toxicstress(H

2O2)in

cultu

remediacaused

amoresignificant

redu

ctionin

cellu

larproliferation

and

DNAsynthesisin

hUCB-SCs,compared

tocontrolcells(cancercells

andhu

man

prim

aryfibroblasts).

(ii)Moreover,hU

CB-SCsshow

edlow

resistance

tooxidativestresswithcellu

lar

senescence

andapop

tosisat

H2O

2levels

muchlower

than

thoseof

controlgroup

s.

Measuring

thecellu

larantioxidant

capacity

show

edthat

hUCB-SCs

hadalower

antioxidantcapacity

than

controlcells.T

oconfi

rmthat,

antioxidantsupp

lementation

increasedthiscapacity

and

diminishedcellu

lardamageup

onexpo

sure

tooxidativestress.

Zengetal.[81]

Edaravone

(10μM),a

clinicallyapproved

drug.

Hum

anum

bilicalcord

blood-

derivedstem

cells

(hUCB-SCs)

from

umbilicalvein.

(i)Unlikethepro-oxidant(diethyl

maleate),edaravon

esignificantly

redu

cedlip

opolysaccharide(LPS)/H

2O2-

indu

ceddamageandincreasedstem

cell

viability

(p<005).

(ii)In

diabeticmicewithsevere

combined

immun

odeficiency,onlythreemicedied

inthegrou

p,injected

with(G

al/LPSand

hUCB-SCs),com

paredto

50%

lossin

the

grou

p,injected

withGal/LPSon

ly.

(iii)

Pretreatm

entwithedaravon

erescuedall

micewithpo

tentiation

ofthehepaticcell

regenerative

power.F

urthermore,it

(i)LP

S/H

2O2challengeindu

ced

apop

tosisby

augm

entin

goxidative

stressandincreasing

Bax/Bcl2

ratio

.How

ever,pretreatm

entwith

edaravon

eabolishedthesechanges.

(ii)Moreover,edaravon

eincreased

theexpression

ofendo

geno

usantio

xidant

enzymes

(sup

eroxide

dism

utase,catalase).

6 Oxidative Medicine and Cellular Longevity

Table1:Con

tinu

ed.

Stud

yID

Antioxidant

(dose)

Stem

celltype

(sou

rce)

Find

ings

Possiblemechanism

s

diminishedthelevelsof

cellu

larinjury

and

proinfl

ammatorymarkers

intreatedmice,

comparedto

thecontrolgroup

.

Rod

riguez-Porcel

etal.[41]

Tem

pol(SO

Dmim

etic)at

0to

10mm/L

concentration.

Rat

cardiomyoblasts,transfected

byabiolum

inescencerepo

rter

gene

forin

vivo

detectionand

transplanted

into

themyocar-

dium

,guided

byhigh-resolution

ultrasou

nd.

(i)Cells,exposed

tohypo

xic/oxidativestress

cond

itions

during

invitroculturing,show

eddecreasedcellviability

withincreasedROS

prod

uction

andNADPH-oxidase-1

expression

,com

paredto

thecontrolgroup

.These

effectsweresignificantlyredu

cedafter

adding

antioxidantsin

ado

se-dependent

manner.

(ii)After

transplantationinto

ratmyocardium,

antioxidant-treatedcells

show

edsignificantly

higher

cellviabilitywithinthefirstthree

days

oftransplantation,

comparedto

untreated

cells.

Hypoxiaindu

cesoxidativestress

byincreasing

theexpression

ofNAD(P)H

oxidaseenzyme.

Interestingly,adding

antioxidant

didno

tredu

ceNAD(P)H

expression

,suggestingthat

tempo

lredu

cesoxidativestressby

neutralizingfree

radicalsrather

than

decreasing

theirprod

uction

.

Lietal.[40]

Hom

emadeantioxidant

cocktailconsisting

of100ML-ascorbate,L-glutathion

e,andα-tocop

herolacetate.

Cardiac

stem

cells

(CSC

s)from

theendo

myocardialtissueof

apatientun

dergoing

acardiacprocedure.

(i)Cells,culturedun

derhypo

xic

cond

itions

(5%

O2)hadalower

number

ofchromosom

alabno

rmalities,compared

tocells,culturedun

derno

rmoxic

cond

itions

(20%

O2).

(ii)Unexpectedly,increasedcytogenic

abno

rmalitiesin

numberandseverity

were

recorded

incells,culturedwithantioxidant

supp

lementation

.(iii)

Antioxidant

supp

lementation

,if

excessive,may

hind

ertheph

ysiological

rolesof

ROSin

stem

cells’p

roliferation

anddifferentiation,

which

raises

anew

concept[reductive

stress].

(i)Measuring

c-H2A

Xfoci

(amarkerof

DNAbreaks)show

edabiph

asicrelation

ship

between

ROSlevelsandfrequencyof

DNA

breaks,thatis,increased

DNA

damageoccursatlowantioxidant/

high

ROSlevels,w

hileexcessive

supp

ressionofROSlevelsincreases

DNAdamage.

(ii)There

isan

optim

allevelo

fROSin

stem

cells,above

which

geno

micinstability

occursdu

eto

ROS-indu

cedDNAdamageand

belowwhich

DNArepairenzymes

areno

tactivated

tomaintainthe

DNAstability.

Takahashi

etal.[82]

Ascorbicacid

(104

M/L)

incubation

for12

days.

Hum

anem

bryonicstem

cells

(ESC

s).

(i)Ascorbicacid

significantlyincreased

ESC

sdifferentiationinto

cardiacmyocytes

inado

se-dependent

manner,as

evidenced

byincreasedexpression

ofthecardiac

specificgene

(myosinheavychain(M

HC)).

(ii)Ascorbicacid,ind

ependent

ofits

antioxidativeprop

erty,ind

uced

ESC

sdifferentiationinto

cardiacmyocytes.

Thiswas

evidencedby

theabsenceof

this

effectwithotherantioxidants,suchas

NAC,T

iron

,and

vitamin

E.

Ascorbicacid

increasedthe

expression

ofcardiacmuscle

genes,such

asGATA4,Nkx2.5,

α-M

HC,β

-MHC,and

atrial

natriureticfactor

(ANF),w

ith

subsequent

cardiac-specific

proteinprod

uction

.

7Oxidative Medicine and Cellular Longevity

Table1:Con

tinu

ed.

Stud

yID

Antioxidant

(dose)

Stem

celltype

(sou

rce)

Find

ings

Possiblemechanism

s

Song

etal.[43]

Selenium

(20or

50ng/m

L)andNAC(100

μM).

Hum

anem

bryonicstem

cell-

(ESC

-)derivedvascular

progenitors.

(i)Fo

llowingvascular

differentiation

ofESC

sinto

vascular

progenitor

cells

(CD34+cells),aqu

iescentstateof

cellu

larproliferation

developedwith

41%

ofthecells

intheG0ph

ase,

upregulation

oftheG1checkp

oint

inhibitor(p21)proteinand

downregulationof

mitosis-related

genes.

(ii)Selenium

increasedcellu

larproliferation

,redu

cedp21expression

,and

decreased

thenu

mberof

cells

inG0ph

aseof

cell

cycle.Moreover,itprom

oted

thevascular

differentiationof

ESC

swithno

similar

effecton

endo

derm

alor

ectoderm

alpo

tentiality.

(iii)

Using

NACtotally

inhibitedthe

vascular

differentiationof

ESC

s.

(i)Physiologically,R

OSare

prod

uced

during

vascular

differentiationmainlyby

NADPH

oxidaseandis

respon

sibleforthisqu

iescent

state.

(ii)Selenium

throughincreasing

glutathion

eandthioredo

xin

activity

mod

eratelydiminished

ROSlevels.

(iii)

How

ever,N

AC,throu

ghcompletescavenging

ofROS,

abolishedtheirph

ysiological

rolein

vascular

differentiation.

Parketal.[45]

GV1001

[derived

from

human

telomerasereverse

transcriptasefrom

0to

100μM.

Neuralstem

cells

(NSC

s)from

miceem

bryonicbrain

(corticaltissue).

GV1001

significantlyredu

cedH

2O2

effectson

NSC

sinclud

ingdiminished

cellu

larproliferation

,migration

and

increasedapop

tosis.Interestingly,

GV1001

itselfhadno

effecton

norm

alun

treatedcells.

(i)GV1001

hasan

ROS-

scavenging

activity,p

reventing

lipid

peroxidation

andDNA

damage.

(ii)Atthemolecular

level,

GV1001

indu

cedtheexpression

ofsurvival-related

proteins

and

diminishedtheexpression

ofapop

tosis-relatedproteins.

Hachem

etal.[46]

CyclosporineA(CsA

),brain-derivedneurotroph

icfactor

(BDNF),and

thyrotropin-

releasingho

rmon

e(TRH).

Neuralstem

cells

(NSC

s)from

thespinalcord

oftransgenicadultfemale

rats(spinalcordinjury

mod

el).

(i)Pretreatm

entwithBDNFfor48

hours

(beforeH

2O2expo

sure)significantly

increasedNSC

sviability

anddecreased

intracellularROSaccumulation,

compared

tothecontrolgroup

.How

ever,C

sAand

TRH-treated

cells

show

edno

significant

changesfrom

thecontrolgroup

.(ii)Interestingly,BDNF-treatedcells

show

edno

changesin

cellu

larproliferation

and

differentiationcomparedto

thecontrol

grou

p.

The

neurop

rotectiveeffectof

BDNFisexertedthroughits

ROS-scavenging

activity

and

indu

ctionof

antioxidantenzymes,

such

asGRandSO

D.M

oreover,

significant

redu

ctions

inapop

totic

features

wereno

tedin

BDNF-

treatedcells,com

paredto

the

controlgroup

.

Song

etal.[52]

Melaton

in(100

nM).

Neuralstem

cells

(NSC

s)from

miceem

bryonic

corticaltissue.

(i)Melaton

insignificantlyredu

cedLP

S-indu

cedtoxicity

andapop

tosisof

NSC

sthroughredu

cing

nitricoxide(N

O)

prod

uction

andindu

cing

antioxidant

enzymes.

(i)Melaton

inincreasedthe

expression

ofmultiple

transcriptionalfactors,involved

inNSC

sproliferatio

n,self-renewal,

anddifferentiatio

n,such

asorph

an

8 Oxidative Medicine and Cellular Longevity

Table1:Con

tinu

ed.

Stud

yID

Antioxidant

(dose)

Stem

celltype

(sou

rce)

Find

ings

Possiblemechanism

s

(ii)Fu

rtherm

ore,itmaintainedthe

neurosph

eresize

inNSC

s,treatedwithLP

S.(iii)

Melaton

inincreasedcellsurvival

byactivating

PI3K/A

ktpathway.T

hiswas

confi

rmed

bytheaddition

ofwortm

annin

(aPI3Kinhibitor),w

hich

inhibitedNrf2

expression

andsubsequent

antioxidant

activities.

nuclearreceptor

TLX

andfibroblast

grow

thfactor

receptor-2.

(ii)Melaton

inup

regulatedthe

expression

ofnu

clearfactor-

erythroid2-relatedfactor

2(N

rf2),

respon

siblefordo

wnstream

activationof

antio

xidant

enzymes’

genes.

Sunetal.[59]

N-A

cetyl-Lcysteine

(NAC)

at2mM

andascorbicacid-

2-ph

osph

ate(A

AP)at

0.2mM

incomparisonto

theeffectof

hypo

xia.

Adipo

se-derived

stem

cells

(ADSC

s)from

human

adiposetissue.

ADSC

s,grow

nin

media,

supp

lementedby

antioxidantsor

underhypo

xiccond

itions

(5%

po2),

show

edamoresignificant

increase

incellproliferation

andadecrease

indo

ublin

gtimethan

thecontrolgroup

,supp

lementedby

fibroblastgrow

thfactor-2.M

oreover,cytometric

analysisshow

edthat

cells,cultured

inantioxidant-supp

lementedand

hypo

xicmedia,h

adagreater

prop

ortion

ofcells

inS1

phase

ofthecellcyclewithdiminished

G0/G1ph

asecells,com

pared

tothecontrolgroup

.

Inantioxidant-supp

lementedmedia,

PCRshow

eddiminishedlevelsof

cyclin-dependent

kinase

inhibitors

(CDK:impo

rtantcellcycle

regulatorsthat

controlenteringS1

phase),w

ithenhanced

expression

ofstem

ness-related

genes,

comparedto

thecontrolgroup

.

Lyub

linskayaetal.

[11]

Tem

pol(1-2mM),NAC

(5–20mM),andresveratrol

(20–40

μM).

End

ometrialstem

cells,

isolated

from

desquamated

endo

metrium

ofmenstrual

bloodandADSC

sfrom

adiposetissue.

(i)Reactiveoxygen

speciesare

impo

rtantregulatorsof

stem

cellself-

renewalandproliferation

upon

exit

ofthequ

iescentstage.

(ii)Using

asynchron

ized

cellin

G0

phase,therewas

atransientincrease

inROSlevelsup

onstim

ulationof

cell

proliferation

anddu

ring

initial

stages

ofDNAsynthesis.

(iii)

Add

ingantioxidantsto

the

medium

afterproliferation

indu

ction

andbefore

initiation

ofS1

phase

blockedS1

transition

.Antioxidant

didno

thave

thesameeffect

whenaddedafterS1

initiation

.

(i)Cells,treated

withantioxidants,

show

edexpression

ofthe

proliferative

marker(K

i-67),

which

isabsent

inthenu

cleusof

quiescentcells,ind

icatingthat

the

cellleftthequ

iescentstateand

was

arrested

inG1ph

ase.

(ii)Antioxidants,throughdo

se-

depend

entredu

ctionof

ROS

levels,can

beused

tocontrol

cellu

larproliferation

during

invitroculturing.

Yangetal.[65]

Fullerol(apo

lyhydroxylated

fullerene)at

0.1,0.3,1,3,

and10

μM.

Hum

anadipose-derived

stem

cells

(ADSC

s).

(i)Fu

llerolenh

ancedtheosteogenic

differentiationof

ADSC

s,as

indicated

byincreasedexpression

ofosteogenic

markers(Run

x2,O

CN,and

alkalin

e

Fullerolexerted

anantioxidant

effecton

ADSC

sthrough

potentiating

theexpression

ofthetranscriptionfactor

FoxO

1

9Oxidative Medicine and Cellular Longevity

Table1:Con

tinu

ed.

Stud

yID

Antioxidant

(dose)

Stem

celltype

(sou

rce)

Find

ings

Possiblemechanism

s

phosph

atase)

andmineralization.

(ii)Moreover,fullerol(at

all

concentrations)redu

cedROSlevelsin

theosteogeniccultu

remedia.

anditsdo

wnstream

genes

(Run

x2andSO

D2),w

hich

prom

oteROSscavenging

and

osteoblasticdifferentiation.

Yuetal.[83]

L-Ascorbicacid

2-ph

osph

ate

(AAP)at

250μM.

Adipo

se-derived

stem

cells

(ADSC

s)from

thesubcutaneous

adiposetissue

from

afemale

patient,un

dergoing

abdo

minop

lasty.

(i)Ascorbicacid

significantlyincreased

ADSC

sproliferation

,preserved

cellu

lar

stem

nessandincreasedthepo

tentiality

foradipogenic,h

epatic,n

euraland

osteogenicdifferentiation.

(ii)In

AAP-ind

uced

cellsheet,there

was

asignificant

increase

inthe

geneticexpression

andsecretionof

collagen,

laminin,and

fibron

ectin

proteins.

(iii)

The

AAP-ind

uced

cellsheet

improved

wou

ndhealingin

amurine

wou

ndmod

el,w

hich

indicatesthe

possibility

ofADSC

sdifferentiation

inano

n-mesenchym

allin

eage.

(i)Add

ingAAPto

ADSC

sincreased

theexpression

ofstem

ness-related

proteins.

(ii)Using

otherantioxidants,such

asNACdidno

tshow

anincrease

ofstem

ness

markers.Incontrast,

adding

acollagensynthesis

inhibitorabolished

AAP-ind

uced

overexpression

ofstem

ness

proteins,

indicating

that

theinvolved

mechanism

inAAPaction

iscollagensynthesis,no

tROS

scavenging.

Wangetal.[66]

NACandAAPat

3mM

and

0.2mM,respectively

(for

20ho

urs).

Hum

anADSC

sfrom

10differenthu

man

patients.

(i)PretreatedMSC

swithantioxidants

show

edless

apop

tosisandlower

caspase-3levelsup

onexpo

sure

toadvanced

glycosylationend-prod

ucts

(AGE),comparedto

thecontrolgroup

.(ii)The

effectsof

NACandAAPwere

significantlyam

plified

afterthe

addition

ofmiRNA-223

mim

etics

andweresignificantlyabolishedby

miRNA-223

inhibitors.

Antioxidantsredu

cedROS

generation

andapop

tosis,indu

ced

byAGE.T

hiscanbe

explainedby

theeffectof

both

onmiR-223

(aregulatorof

intracellularapop

totic

singlin

gthroughmod

ulationof

fibroblast-likegrow

thfactor

receptor-2

proteinlevels.

Drowleyetal.[72]

N-A

cetyl-Lcysteine

at10

mM

incomparisonto

the

pro-oxidant(diethylmaleate)

at50

μM.

Muscle-derivedstem

cells

(MDSC

s)from

theskeletal

muscleof

3-week-old

femalemice.

(i)In

comparisonto

controlcells,

NAC-treated

cells

show

edincreased

survivalanddifferentiationinto

myotubesup

onexpo

sure

tooxidative

(H2O

2)or

inflam

matorystress

(tum

ornecrosisfactor).

(ii)In

failedmicehearts,injection

withNAC-treated

cells

significantly

improved

systolicanddiastolic

function

onecho

cardiographicassessment,

comparedto

injectionwithun

treated

andDEM-treated

cells.M

oreover,

scar

tissue

form

ationwas

significantly

(i)IncreasedcellsurvivalafterNAC

treatm

entisprobablyrelatedto

stim

ulationof

mitogen-activated

proteinkinases(M

APK)and

extracellularsignal-regulated

kinase

(ERK),kinase

families

involved

incellu

larsurvivaland

proliferation

.(ii)Interestingly,asimilarincrease

inCD31+endothelialcellsinNAC-

treatedandun

treatedMDSC

swas

observed.

10 Oxidative Medicine and Cellular Longevity

Table1:Con

tinu

ed.

Stud

yID

Antioxidant

(dose)

Stem

celltype

(sou

rce)

Find

ings

Possiblemechanism

s

lower

inNAC-treated

cells

than

untreatedcells.

Aliakbarietal.[74]

Catalase(40mL)

and

α-tocop

herol(200mL).

Spermatogon

ialstem

cells

(SSC

s)from

neon

atal

malemicetestis.

Antioxidant

supp

lementation

ofcryopreservedSSCsredu

cedoxidative

damageto

mem

branes

andorganelles

andincreasedcellsurvivalin

ado

se-

depend

entmanner.

Catalaseandα-tocop

herolreduced

ROSgeneration

intreatedcells,

comparedto

controlcells.

Moreover,antioxidant-treated

cells

show

edan

increased

expression

oftheanti-apo

ptotic

BcL-2

gene

withdecreased

expression

ofthepro-apop

totic

BAXgene,com

paredto

the

controlgroup

.

ADSC

s:adipose-derivedstem

cells;C

AT:catalase;DEM:diethylmaleate;G

SH:glutathione;H

SCs:hematop

oieticstem

cells;iPSC

s:indu

cedpluripotentstem

cells;M

DSC

s:muscle-derivedstem

cells;N

AC:N

-acetyl

cysteine;N

SCs:neuralstem

cells;SCC:sperm

atogon

ialstem

cells;SOD:sup

eroxidedism

utase;ROS:reactive

oxygen

species.

11Oxidative Medicine and Cellular Longevity

8. Muscle-Derived Stem Cells (MDSCs)

According to Drowley and colleagues, injection of injuredskeletal muscles with NAC-treated MDSCs significantlyincreased muscle regeneration, compared to muscles injectedwith untreated or DEM-treated MDSCs. The direction ofscar tissue formation was opposite the direction of the hostmuscle regeneration [72]. Additionally, they showed animproved survival of NAC-treated MDSCs, probably due tostimulation of extracellular signal-regulated kinase (ERK)pathway, as evidenced by decreased survival of NAC treatedcells after inhibition of the ERK pathway [72, 73].

Moreover, they demonstrated that experimentallyinfarcted hearts, injected with NAC-treated MDSCs, showeda more significant reduction in the percentage area of collag-enous scar tissue than hearts injected with either untreated,DEM-treated, or phosphate buffered saline- (PBS-) treatedMDSCs. There was no difference in myocardial scar forma-tion between hearts injected with DEM-treated MDSCs andthose injected with PBS [72].

9. Spermatogonia Stem Cells (SSCs)

Cryopreservation of spermatogonial stem cells, in the pres-ence of catalase (CAT) and α-tocopherol (α-TCP), promotedcell viability and suppressed apoptosis through inducing theexpression of the antiapoptotic BcL-2 gene and inhibiting

the expression of the proapoptotic BAX gene [74]. In otherstudies, cryopreservation with antioxidants could promotecell enrichment and increase the efficiency of colony forma-tion in isolated SSCs [75, 76]. Spermatogonia-derived colo-nies showed increased SSC marker activity, enhancedexpression of self-renewal genes, such as promyelocytic leu-kemia zinc finger (Plzf) protein and DNA-binding proteininhibitor ID4, and suppressed expression of the proto-oncogene (c-kit) in both CAT and α-TCP treated groups[74]. This technique can increase the possibility of SSCsbanking for men with malignant diseases and promote theresumption of spermatogenesis in SCCs recipients. A sum-mary of the design and main findings of included studies isillustrated in Table 1.

10. Discussion

Our review highlights that antioxidants can influence stemcell activities by [1] mitigating oxidative stress through neu-tralization of free radicals and increasing the expression ofantioxidant enzymes and [2] influencing the differentiationfate of precursor stem cells. Further beneficial effects of anti-oxidant treatment include increasing genomic stability,improving the adhesion of stem cells to culture media, andenabling researchers to manipulate stem cell proliferationby using different doses of antioxidants. Figure 1 summarizesthe effects of antioxidants on different types of stem cells.

Induced pluripotent stem cells:mitigation of oxidative stressduring reprogramming and

reduction of genetic aberrations

Hematopoietic stem cells:increase cellular proliferation,

genomic stability, and SODcellular concentration and

in�uence di�erentiation fate

Cardiomyoblasts: increaseearly survival of

cardiomyoblasts andin�uence vascular

di�erentiation of embryonicstem cells

Neural stem cells: mitigateneuroin�ammation andoxidative stress, reducelipid peroxidation andmtDNA mutation, and

increase stem cellsmigration

Human periodontalligament cells: increase

GSH and GSTconcentrations through

Nrf2 activation andenhance osteoplastic

di�erentiation

Spermatogonial stemcells: enhance self-

renewal and suppressapoptosis by increasing

Bcl2/Bax ratio

Muscle-derived stemcells: improved survival by

stimulating MAPK/ERKpathway and decrease

collagen scar formation ininjured muscle

Adipose-derived stemcells: increase stemness

genes’ expression,reduce apoptosis, and

increase osteogenicactivity

Figure 1: Summarizes the effects of antioxidants on different types of stem cells.

12 Oxidative Medicine and Cellular Longevity

We also discussed that a physiological level of ROS (oxi-dative optimum) is needed for proper differentiation of stemcells, especially for proper cardiogenesis and vasculogenesis[40]. These findings can have several clinical applications,such as improving neurogenesis in patients with stroke andneurodegenerative diseases, as well as improving the regener-ation of infarcted myocardial tissue and the banking of SCCs.

Antioxidants are prevalent supplements worldwide.However, little is known about their cell-type-specificactions. It has been shown that a therapeutic dose may varybetween different cell types: a dose that rescues a pathologyin one tissue may roughly challenge the function of another[22]. Therefore, there is a need for dose-effect studies on anti-oxidants to confirm their safety as nutritional supplements ortherapeutic agents—particularly in the case of antioxidantsaccumulating in the mitochondria. Our review also showedthe potential of some endogenous molecules, such as melato-nin, BDNF, and the adipokine (lipocalin-2) in preservingstem cell viability and differentiation potential. Whetherthese compounds can be used in future clinical applicationsof stem cells and whether other endogenous molecules withproven antioxidant activities, such as adiponectin [84], canbe useful in this regard require further investigation.

11. Recommendations

(1) Multiplicity of stem cell sources within the body (dif-ferent home environments) and their variable ROSscavenging capacity make them susceptible to oxida-tive stress at different thresholds. Therefore, we triedto review each stem cell type as a separate entity andwe believe that clearing those differences on themolecular and genetic levels will optimize the clinicalapplication of stem cells in different medical fields.

(2) Most of stem cell characteristics are establishedwithinin vitro culturing environments. More in vivo studiesare required to define their interactions within thebody. Furthermore, few in vivo studies have focusedon the long-term survival of transplanted stem cells;therefore, this should be the interest of future studies.

(3) The effect of ROS level and redox state on the long-term oncogenicity of stem cells should be furtherinvestigated prior to in vivo clinical trials.

12. Conclusion

Using antioxidants can improve the viability and self-renewalcapacity of stem cells and affect their differentiation poten-tial. More research is needed on the dose-effect associationand cell-type-specific actions of antioxidant before applyingthese findings in human therapeutic trials.

Abbreviations

ADSCs: Adipose-derived stem cellsBMSCs: Bone marrow-derived mesenchymal stem cells

CAT: CatalaseDEM: DiethylmaleateDFO: DeferoxamineGSH: GlutathionehPDLCs: Human periodontal ligament cellsHSCs: Hematopoietic stem cellsiPSCs: Induced pluripotent stem cellsMDSCs: Muscle-derived stem cellsNAC: N-acetyl cysteineNSCs: Neural stem cellsPBP: Phosphate buffered salineSCCs: Spermatogonial stem cellsSOD: Superoxide dismutaseROS: Reactive oxygen species.

Conflicts of Interest

The authors declare that they have no conflicts of interest.

Authors’ Contributions

Sara Shaban, Mostafa Wanees Ahmed El-Husseny, andAbdelrahman Ibrahim Abushouk contributed equally tothis work.

References

[1] J. Thomson and J. Odorico, “Human embryonic stem cell andembryonic germ cell lines,” Trends in Biotechnology, vol. 18,no. 2, pp. 53–57, 2000.

[2] N. Knoepffler, D. Schipanski, and S. L. Sorgner,Humanbiotech-nology as Social Challenge: An Interdisciplinary Introduction toBioethics, Ashgate, Farnham, Surrey, United Kingdom, 2007.

[3] B. Vastag, “Stem cells step closer to the clinic,” Jama, vol. 285,no. 13, p. 1691, 2001, American Medical Association.

[4] M. Izumikawa, R. Minoda, K. Kawamoto et al., “Auditory haircell replacement and hearing improvement by Atoh1 genetherapy in deaf mammals,” Nature Medicine, vol. 11, no. 3,pp. 271–276, 2005.

[5] S. D. Narasipura, J. C. Wojciechowski, N. Charles, J. L.Liesveld, and M. R. King, “P-selectin–coated microtubefor enrichment of CD34+ hematopoietic stem and progen-itor cells from human bone marrow,” Clinical Chemistry,vol. 54, no. 1, pp. 77–85, 2007.

[6] M. Witkowska-Zimny and K. Walenko, “Stem cells from adi-pose tissue,” Cellular and Molecular Biology Letters, vol. 16,no. 2, pp. 236–257, 2011.

[7] M. A. Esteban, T. Wang, B. Qin et al., “Vitamin C enhances thegeneration of mouse and human induced pluripotent stemcells,” Cell Stem Cell, vol. 6, no. 1, pp. 71–79, 2010.

[8] M. Stadtfeld, E. Apostolou, F. Ferrari, J. Choi, and R. Walsh,“Ascorbic acid prevents loss of Dlk1-Dio3 imprinting andfacilitates generation of all-iPS cell mice from terminally differ-entiated B cells,” Nature, vol. 44, no. 4, pp. 398–405, 2012.

[9] W. Neeley and J. Essigmann, “Mechanisms of formation, geno-toxicity, and mutation of guanine oxidation products,” Chemi-cal Research in Toxicology, vol. 19, no. 4, pp. 491–505, 2006.

[10] J. Silva andO.Coutinho, “Free radicals in the regulationof dam-age and cell death—basic mechanisms and prevention,” DrugDiscoveries & Therapeutics, vol. 4, no. 3, pp. 144–167, 2010.

13Oxidative Medicine and Cellular Longevity

[11] O. G. Lyublinskaya, Y. G. Borisov, N. A. Pugovkina et al.,“Reactive oxygen species are required for human mesenchy-mal stem cells to initiate proliferation after the quiescenceexit,” Oxidative Medicine and Cellular Longevity, vol. 2015,Article ID 502105, 8 pages, 2015.

[12] B. J. Le, N. Orozco, A. Paucar, and J. Saxe, “Proliferative neuralstem cells have high endogenous ROS levels that regulate self-renewal and neurogenesis in a PI3K/Akt-dependant manner,”Cell Stem Cell, vol. 8, no. 1, pp. 59–71, 2011.

[13] C. Kobayashi and T. Suda, “Regulation of reactive oxygen spe-cies in stem cells and cancer stem cells,” Journal of CellularPhysiology, vol. 227, no. 2, pp. 421–430, 2012.

[14] C. Pérez Estrada, R. Covacu, S. R. Sankavaram, M. Svensson,and L. Brundin, “Oxidative stress increases neurogenesis andoligodendrogenesis in adult neural progenitor cells,” StemCells and Development, vol. 23, no. 19, pp. 2311–2327, 2014.

[15] S. Kothari, A. Thompson, A. Agarwal, and S. S. du Plessis,“Free Radicals: Their Beneficial and Detrimental Effects onSperm Function,” Indian Journal of Experimental Biology,vol. 48, no. 5, pp. 425–435, 2010.

[16] S. Vertuani, A. Angusti, and S. Manfredini, “The antioxidantsand pro-antioxidants network: an overview,” Current Pharma-ceutical Design, vol. 10, no. 14, pp. 1677–1694, 2004.

[17] H. Sies, “Oxidative stress: oxidants and antioxidants,” Experi-mental Physiology, vol. 82, no. 2, pp. 291–295, 1997.

[18] B. Dannenmann, S. Lehle, F. Essmann, and K. Schulze-Osthoff,“Genome surveillance in pluripotent stem cells: low apoptosisthreshold and efficient antioxidant defense,”Molecular&Cellu-lar Oncology, vol. 3, no. 2, article e1052183, 2016, Taylor &Francis.

[19] B. Dannenmann, S. Lehle, D. G. Hildebrand et al., “High gluta-thione and glutathione peroxidase-2 levels mediate cell-type-specificDNAdamageprotection in human inducedpluripotentstem cells,” Stem Cell Reports, vol. 4, no. 5, pp. 886–898, 2015.

[20] J. Ji, V. Sharma, S. Qi et al., “Antioxidant supplementationreduces genomic aberrations in human induced pluripotentstem cells,” Stem Cell Reports, vol. 2, no. 1, pp. 44–51, 2014.

[21] L. Luo, M. Kawakatsu, C.-W. Guo et al., “Effects of antioxi-dants on the quality and genomic stability of induced pluripo-tent stem cells,” Scientific Reports, vol. 4, p. 3779, 2014, NaturePublishing Group.

[22] R. H. Hämäläinen, K. J. Ahlqvist, P. Ellonen et al., “mtDNAmutagenesis disrupts pluripotent stem cell function by alteringredox signaling,” Cell Reports, vol. 11, no. 10, pp. 1614–1624,2015.

[23] A. M. Sadowska, B. Manuel-y-Keenoy, and W. A. De Backer,“Antioxidant and anti-inflammatory efficacy of NAC in thetreatment of COPD: discordant in vitro and in vivo dose-effects: a review,” Pulmonary Pharmacology & Therapeutics,vol. 20, no. 1, pp. 9–22, 2007.

[24] G. F. Kelso, C. M. Porteous, C. V. Coulter et al., “Selectivetargeting of a redox-active ubiquinone to mitochondriawithin cells: antioxidant and antiapoptotic properties,” TheJournal of Biological Chemistry, vol. 276, no. 7, pp. 4588–4596, 2001, American Society for Biochemistry and Molecu-lar Biology.

[25] T. Kinoshita, G. Nagamatsu, T. Kosaka et al., “Ataxia-telangi-ectasia mutated (ATM) deficiency decreases reprogrammingefficiency and leads to genomic instability in iPS cells,” Bio-chemical and Biophysical Research Communications, vol. 407,no. 2, pp. 321–326, 2011.

[26] R. Kitagawa and M. B. Kastan, “The ATM-dependent DNAdamage signaling pathway,” Cold Spring Harbor Symposia onQuantitative Biology, vol. 70, no. 1, pp. 99–109, 2005, ColdSpring Harbor Laboratory Press.

[27] A. T. Noon and A. A. Goodarzi, “53BP1-mediated DNA dou-ble strand break repair: insert bad pun here,” DNA Repair,vol. 10, no. 10, pp. 1071–1076, 2011.

[28] D. Foudah, S. Redaelli, E. Donzelli et al., “Monitoring thegenomic stability of in vitro cultured rat bone-marrow-derived mesenchymal stem cells,” Chromosome Research,vol. 17, no. 8, pp. 1025–1039, 2009, Springer Netherlands.

[29] Y. F. Zhou, M. Bosch-Marce, H. Okuyama et al., “Spontaneoustransformation of cultured mouse bone marrow–derived stro-mal cells,” Cancer Research, vol. 66, no. 22, pp. 10849–10854,2006.

[30] D. E. C. Baker, N. J. Harrison, E. Maltby et al., “Adaptation toculture of human embryonic stem cells and oncogenesisin vivo,” Nature Biotechnology, vol. 25, no. 2, pp. 207–215,2007, Nature Publishing Group.

[31] A. Maitra, D. E. Arking, N. Shivapurkar et al., “Genomic alter-ations in cultured human embryonic stem cells,” NatureGenetics, vol. 37, no. 10, pp. 1099–1103, 2005, Nature Publish-ing Group.

[32] A. M. Liu, W. W. Qu, X. Liu, and C.-K. Qu, “Chromosomalinstability in in vitro cultured mouse hematopoietic cells asso-ciated with oxidative stress,” American Journal of BloodResearch, vol. 2, no. 1, pp. 71–76, 2012, e-Century PublishingCorporation.

[33] Y.-Y. Jang and S. J. Sharkis, “A low level of reactive oxygen spe-cies selects for primitive hematopoietic stem cells that mayreside in the low-oxygenic niche,” Blood, vol. 110, no. 8,pp. 3056–3063, 2007.

[34] P. Eliasson, M. Rehn, P. Hammar et al., “Hypoxia mediates lowcell-cycle activity and increases the proportion of long-term–reconstituting hematopoietic stem cells during in vitro culture,”Experimental Hematology, vol. 38, no. 4, pp. 301–310.e2, 2010.

[35] G. H. Danet, Y. Pan, J. L. Luongo, D. A. Bonnet, and M. C.Simon, “Expansion of human SCID-repopulating cells underhypoxic conditions,” The Journal of Clinical Investigation,vol. 112, no. 1, pp. 126–135, 2003, American Society for Clin-ical Investigation.

[36] Z. A. Hamid, W. Hii, L. Lin et al., “The role of Hibiscus sabdar-iffa L. (roselle) in maintenance of ex vivo murine bonemarrow-derived hematopoietic stem cells,” The ScientificWorld Journal, vol. 2014, Article ID 258192, 10 pages, 2014.

[37] R. Halabian and H. A. Tehrani, “Lipocalin-2-mediated upreg-ulation of various antioxidants and growth factors protectsbone marrow-derived mesenchymal stem cells against unfa-vorable microenvironments,” Cell Stress and Chaperones,vol. 18, no. 6, pp. 785–800, 2013.

[38] G. Fan, L. Wen, M. Li et al., “Isolation of mouse mesenchymalstem cells with normal ploidy from bone marrows by reducingoxidative stress in combination with extracellular matrix,”BMC Cell Biology, vol. 12, no. 1, p. 30, 2011.

[39] K. Choi, Y. Seo, H. Yoon et al., “Effect of ascorbic acid on bonemarrow-derived mesenchymal stem cell proliferation and dif-ferentiation,” Journal of Bioscience and Bioengineering,vol. 105, no. 6, pp. 586–594, 2008.

[40] T. S. Li and E. Marbán, “Physiological levels of reactive oxygenspecies are required to maintain genomic stability in stemcells,” Stem Cells, vol. 28, no. 7, pp. 1178–1185, 2010.

14 Oxidative Medicine and Cellular Longevity

[41] M. Rodriguez-Porcel, O. Gheysens, R. Paulmurugan et al.,“Antioxidants improve early survival of cardiomyoblasts aftertransplantation to the myocardium,” Molecular Imaging andBiology, vol. 12, no. 3, pp. 325–334, 2010, Springer-Verlag.

[42] C. Napoli, S. Williams-Ignarro, F. de Nigris et al., “Beneficialeffects of concurrent autologous bone marrow cell therapyand metabolic intervention in ischemia-induced angiogenesisin the mouse hindlimb,” Proceedings of the National Academyof Sciences of the United States of America, vol. 102, no. 47,pp. 17202–17206, 2005, National Academy of Sciences.

[43] S. Song, K. Kim, J. J. Park, K. H. Min, and W. Suh, “Reactiveoxygen species regulate the quiescence of CD34-positive cellsderived from human embryonic stem cells,” CardiovascularResearch, vol. 103, no. 1, pp. 1–29, 2014.

[44] M. Song, Y.-J. Kim, Y.-H. Kim, J. Roh, S. U. Kim, and B.-W.Yoon, “Effects of duplicate administration of human neuralstem cell after focal cerebral ischemia in the rat,” InternationalJournal of Neuroscience, vol. 121, no. 8, pp. 457–461, 2011,Taylor & Francis.

[45] H. H. Park, H. J. Yu, K. Sangjae et al., “Neural stem cellsinjured by oxidative stress can be rejuvenated by GV1001, anovel peptide, through scavenging free radicals and enhancingsurvival signals,” Neurotoxicology, vol. 55, pp. 131–141, 2016,Elsevier B.V.

[46] L. D. Hachem, A. J. Mothe, and C. H. Tator, “Effect of BDNFand other potential survival factors in models of in vitro oxida-tive stress on adult spinal cord–derived neural stem/progenitorcells,” BioResearch Open Access, vol. 4, no. 1, pp. 146–159,2015.

[47] A. A. Pieper, S. Blackshaw, E. E. Clements et al., “Poly(ADP-ribosyl)ation basally activated by DNA strand breaks reflectsglutamate-nitric oxide neurotransmission,” Proceedings of theNational Academy of Sciences of the United States of America,vol. 97, no. 4, pp. 1845–1850, 2000, National Academy ofSciences.

[48] V. Calabrese, T. E. Bates, and A. M. G. Stella, “NO synthaseand NO-dependent signal pathways in brain aging andneurodegenerative disorders: the role of oxidant/antioxidantbalance,” Neurochemical Research, vol. 25, no. 9/10, pp. 1315–1341, 2000, Kluwer Academic Publishers-Plenum Publishers.

[49] A. Cheng, S. L. Chan, O. Milhavet, S. Wang, and M. P.Mattson, “p38 MAP kinase mediates nitric oxide-inducedapoptosis of neural progenitor cells,” The Journal of Biolog-ical Chemistry, vol. 276, no. 46, pp. 43320–43327, 2001,American Society for Biochemistry and Molecular Biology.

[50] R. Covacu, A. I. Danilov, B. S. Rasmussen et al., “Nitric oxideexposure diverts neural stem cell fate from neurogenesistowards astrogliogenesis,” Stem Cells, vol. 24, no. 12,pp. 2792–2800, 2006, John Wiley & Sons, Ltd.

[51] A. Torroglosa, M. Murillo-Carretero, C. Romero-Grimaldi, E.R. Matarredona, A. Campos-Caro, and C. Estrada, “Nitricoxide decreases subventricular zone stem cell proliferation byinhibition of epidermal growth factor receptor and phosphoi-nositide-3-kinase/Akt pathway,” Stem Cells, vol. 25, no. 1,pp. 88–97, 2007, John Wiley & Sons, Ltd.

[52] J. Song, S. M. Kang, K. M. Lee, and J. E. Lee, “The protectiveeffect of melatonin on neural stem cell against LPS-inducedinflammation,” BioMed Research International, vol. 2015,Article ID 854359, 13 pages, 2015, Hindawi PublishingCorporation.

[53] C. Tomás-Zapico and A. Coto-Montes, “A proposed mecha-nism to explain the stimulatory effect of melatonin on

antioxidative enzymes,” Journal of Pineal Research, vol. 39,no. 2, pp. 99–104, 2005, Munksgaard International Publishers.

[54] A. Vilar, L. de Lemos, I. Patraca et al., “Melatonin suppressesnitric oxide production in glial cultures by pro-inflammatorycytokines through p38 MAPK inhibition,” Free RadicalResearch, vol. 48, no. 2, pp. 119–128, 2014, Taylor & Francis.

[55] G. Negi, A. Kumar, and S. S. Sharma, “Melatonin modulatesneuroinflammation and oxidative stress in experimental dia-betic neuropathy: effects on NF-κB and Nrf2 cascades,” Jour-nal of Pineal Research, vol. 50, no. 2, pp. 124–131, 2010,Blackwell Publishing Ltd.

[56] Q. Ma, “Role of nrf2 in oxidative stress and toxicity. Annualreview of pharmacology and toxicology,” NIH Public Access,vol. 53, pp. 401–426, 2013.

[57] Y.-J. Surh, J. K. Kundu, M.-H. Li, H.-K. Na, and Y.-N. Cha,“Role of Nrf2-mediated heme oxygenase-1 upregulation inadaptive survival response to nitrosative stress,” Archives ofPharmacal Research, vol. 32, no. 8, pp. 1163–1176, 2009, Phar-maceutical Society of Korea.

[58] M. Petro, H. Jaffer, J. Yang, S. Kabu, V. B. Morris, and V.Labhasetwar, “Biomaterials tissue plasminogen activatorfollowed by antioxidant-loaded nanoparticle delivery pro-motes activation/mobilization of progenitor cells ininfarcted rat brain,” Biomaterials, vol. 81, pp. 169–180,2016, Elsevier Ltd.

[59] L.-Y. Sun, C.-Y. Pang, D.-K. Li et al., “Antioxidants cause rapidexpansion of human adipose-derived mesenchymal stem cellsvia CDK and CDK inhibitor regulation,” Journal of BiomedicalScience, vol. 20, no. 1, p. 53, 2013, BioMed Central.

[60] K. Tamama, H. Kawasaki, S. S. Kerpedjieva, J. Guan, R. K.Ganju, and C. K. Sen, “Differential roles of hypoxia induciblefactor subunits in multipotential stromal cells under hypoxiccondition,” Journal of Cellular Biochemistry, vol. 112, no. 3,pp. 804–817, 2011, Wiley Subscription Services, Inc., A WileyCompany.

[61] S.-P. Hung, J. H. Ho, and Y.-R. V. ShihT. Lo and O. K. Lee,“Hypoxia promotes proliferation and osteogenic differentia-tion potentials of human mesenchymal stem cells,” Journal ofOrthopaedic Research, vol. 30, no. 2, pp. 260–266, 2012, WileySubscription Services, Inc., A Wiley Company.

[62] I. Martin, A. Muraglia, G. Campanile, R. Cancedda, and R.Quarto, “Fibroblast growth factor-2 supports ex vivo expan-sion and maintenance of osteogenic precursors from humanbone marrow,” Endocrinology, vol. 138, no. 10, pp. 4456–4462, 1997, Endocrine Society.

[63] M. Higuchi, G. J. Dusting, H. Peshavariya et al., “Differentia-tion of human adipose-derived stem cells into fat involvesreactive oxygen species and forkhead box O1 mediated upreg-ulation of antioxidant enzymes,” Stem Cells and Development,vol. 22, no. 6, pp. 878–888, 2013, Mary Ann Liebert, Inc. 140Huguenot Street, 3rd Floor New Rochelle, NY 10801 USA.

[64] T. Kojima, T. Norose, K. Tsuchiya, and K. Sakamoto, “Mouse3T3-L1 cells acquire resistance against oxidative stress as theadipocytes differentiate via the transcription factor FoxO,”Apoptosis, vol. 15, no. 1, pp. 83–93, 2010, Springer US.

[65] X. Yang, C. J. Li, Y. Wan, P. Smith, G. Shang, and Q. Cui,“Antioxidative fullerol promotes osteogenesis of humanadipose-derived stem cells,” International Journal of Nanome-dicine, vol. 9, pp. 4023–4031, 2014.

[66] Z. Wang, H. Li, R. Guo, Q. Wang, and D. Zhang, “Antioxi-dants inhibit advanced glycosylation end-product-inducedapoptosis by downregulation of miR-223 in human adipose

15Oxidative Medicine and Cellular Longevity

tissue-derived stem cells,” Scientific Reports, vol. 6, pp. 1–11,2016, Nature Publishing Group.

[67] J.-H. Chung, Y.-S. Kim, K. Noh, Y.-M. Lee, S.-W. Chang, andE.-C. Kim, “Deferoxamine promotes osteoblastic differentia-tion in human periodontal ligament cells via the nuclear factorerythroid 2-related factor-mediated antioxidant signalingpathway,” Journal of Periodontal Research, vol. 49, no. 5,pp. 563–573, 2014.

[68] S. Philipp, L. Cui, B. Ludolph et al.J. M. Downey et al., “Desfer-oxamine and ethyl-3,4-dihydroxybenzoate protect myocar-dium by activating NOS and generating mitochondrial ROS,”American Journal of Physiology-Heart and Circulatory Physiol-ogy, vol. 290, no. 1, p. H450, 2005.

[69] C.-N. Im, J.-S. Lee, Y. Zheng, and J.-S. Seo, “Iron chelationstudy in a normal human hepatocyte cell line suggests thattumor necrosis factor receptor-associated protein 1 (TRAP1)regulates production of reactive oxygen species,” Journal ofCellular Biochemistry, vol. 100, no. 2, pp. 474–486, 2007, WileySubscription Services, Inc., A Wiley Company.

[70] E. Ardite, J. A. Barbera, J. Roca, and J. C. Fernández-Checa,“Glutathione depletion impairs myogenic differentiation ofmurine skeletal muscle C2C12 cells through sustained NF-κBactivation,” The American Journal of Pathology, vol. 165,no. 3, pp. 719–728, 2004.

[71] F. Esposito, V. Agosti, G. Morrone et al., “Inhibition of the dif-ferentiation of human myeloid cell lines by redox changesinduced through glutathione depletion,” Biochemical Journal,vol. 301, no. 3, pp. 649–653, 1994.

[72] L. Drowley, M. Okada, S. Beckman et al., “Cellular antioxidantlevels influence muscle stem cell therapy,”Molecular Therapy,vol. 18, no. 10, pp. 1865–1873, 2010, Nature Publishing Group.

[73] A. Paranjpe, N. A. Cacalano, W. R. Hume, and A. Jewett, “N-acetylcysteine protects dental pulp stromal cells from HEMA-induced apoptosis by inducing differentiation of the cells,”Free Radical Biology and Medicine, vol. 43, no. 10, pp. 1394–1408, 2007.

[74] F. Aliakbari, M. A. S. Gilani, F. Amidi et al., “Improving theefficacy of cryopreservation of spermatogonia stem cells byantioxidant supplements,” Cellular Reprogramming, vol. 18,no. 2, pp. 87–95, 2016, Mary Ann Liebert, Inc. 140 HuguenotStreet, 3rd Floor New Rochelle, NY 10801 USA.

[75] M. Koruji, M. Koruji, M. Movahedin, S. J. Mowla, and H.Gourabi, “Colony formation ability of frozen thawed sper-matogonial stem cell from adult mouse,” International Journalof Reproductive BioMedicine, vol. 5, no. 3, pp. 109–115, 2012.

[76] T. Mirzapour, M. Movahedin, T. A. Tengku Ibrahim, A. W.Haron, and M. R. Nowroozi, “Evaluation of the effects of cryo-preservation on viability, proliferation and colony formationof human spermatogonial stem cells in vitro culture,” Androlo-gia, vol. 45, no. 1, pp. 26–34, 2013.

[77] Y. Ikeda, T. Yoshinari, H. Miyoshi, and Y. Nagasaki, “Designof antioxidative biointerface for separation of hematopoieticstem cells with high maintenance of undifferentiated pheno-type,” Journal of Biomedical Materials Research Part A,vol. 104, no. 8, pp. 2080–2085, 2016.

[78] T. Wang, G. Zeng, X. Li, and H. Zeng, “In vitro studies on theantioxidant and protective effect of 2-substituted-8-hydroxy-quinoline derivatives against H2O2-induced oxidative stressin BMSCs,” Chemical Biology & Drug Design, vol. 75, no. 2,pp. 214–222, 2010.

[79] N. K. Mekala, R. R. Baadhe, S. R. Parcha, and D. Y. Prameela,“Enhanced proliferation and osteogenic differentiation of

human umbilical cord blood stem cells by L-ascorbic acid,in vitro,” Current Stem Cell Research & Therapy, vol. 8, no. 2,pp. 156–162, 2013.

[80] E. Ko, K. Y. Lee, and D. S. Hwang, “Human umbilical cordblood–derived mesenchymal stem cells undergo cellular senes-cence in response to oxidative stress,” Syem Cells and Develop-ment, vol. 21, no. 11, pp. 1877–1886, 2012.

[81] W. Zeng, J. Xiao, G. Zheng, F. Xing, G. L. Tipoe, and X. Wang,“Antioxidant treatment enhances human mesenchymal stemcell anti-stress ability and therapeutic efficacy in an acute liverfailure model,” Scientific Reports, vol. 5, article 11100, 2015,Nature Publishing Group.

[82] T. Takahashi, B. Lord, P. C. Schulze et al., “Ascorbic acidenhances differentiation of embryonic stem cells into cardiacmyocytes,” Circulation, vol. 107, no. 14, pp. 1912–1917, 2003.

[83] J. Yu, T. Yuan-Kun, and N.-C. C. Yueh-Bih Tang, “Stemnessand transdifferentiation of adipose- derived stem cells usingL-ascorbic acid 2,” Biomaterials, vol. 35, no. 11, pp. 3516–3526, 2014.

[84] M. W. El Husseny, M. Mamdouh, S. Shaban et al., “Adipo-kines: potential therapeutic targets for vascular dysfunctionin type II diabetes mellitus and obesity,” Journal of DiabetesResearch, vol. 2017, Article ID 8095926, 11 pages, 2017.

16 Oxidative Medicine and Cellular Longevity

Submit your manuscripts athttps://www.hindawi.com

Stem CellsInternational

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

MEDIATORSINFLAMMATION

of

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Behavioural Neurology

EndocrinologyInternational Journal of

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Disease Markers

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

BioMed Research International

OncologyJournal of

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Oxidative Medicine and Cellular Longevity

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

PPAR Research

The Scientific World JournalHindawi Publishing Corporation http://www.hindawi.com Volume 2014

Immunology ResearchHindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Journal of

ObesityJournal of

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Computational and Mathematical Methods in Medicine

OphthalmologyJournal of

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Diabetes ResearchJournal of

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Research and TreatmentAIDS

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Gastroenterology Research and Practice

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Parkinson’s Disease

Evidence-Based Complementary and Alternative Medicine

Volume 2014Hindawi Publishing Corporationhttp://www.hindawi.com