Sobrevia & Gonzalez 2009

Embed Size (px)

Citation preview

  • 8/10/2019 Sobrevia & Gonzalez 2009

    1/8

    I

    A Role for Insulin on L-Arginine Transport in Fetal Endothelial Dysfunc-tion in Hyperglycaemia

    I Cellular and Molecular Physiology LaboratOl:r (CI"fPL) and Perinatology Research LaboratOlY (PRL), Department of

    Obstetrics and Gynaecology, Medical Research Centre (Cn1), School o f M edicine, Faculty of Medicine, Pontificia Uni-

    versidad Catolica de Chile, P.O. Box 114-D, Santiago, Chile; 1Department of Physiology, Faculty of Biological Sci-

    ences, Universidad de Concepcion, Chile

    Abstract: Endothelial cells are keyin the regulationofvascular tone throughthe release of vasoactive molecules,includ-

    ingnitric oxide (NO). NO is a gassynthesized from thecationic amino acid L-arginine via the endothelial NO synthase

    (eNOS). The semi-essentialamino acid L-arginine is a taken up byendothelial cells via systems y+ and y+L in primary

    cultures of human umbilical vein endothelialcells (HUVEC). Systemy+isa familyof membranetransportersincluding at

    least five transport systems for cationic amino acids (CAT) of which HUVEC express human CAT-I (hCAT-I) and

    hCAT-2B. Exposure ofHUVECtohigh extracellularconcentrationsofD-glucose increasesL-argininetransport,hCAT-lmRNA expressionand eNOS activity.These phenomena are also relatedwith increased production of reactive oxygen

    species (ROS), thus supporting the possibility that changes in L-argininelNO signalling pathway result from elevated

    ROS. It has been shown that insulin blocks D-glucose-increased L-arginine transport and cGMP accumulation in

    HUVEC,whereasin this cell type insulin alsomodulateshigh D-glucoseeffectsby activatingthetranscriptional factors

    Spl andNFKB.Thesetranscriptionfactorshaveresponse elements in SLC7A] (for hCAT-I) genepromoterregion,thus

    representing2 possible targetsforregulationoftheexpressionof thistransporter byD-glucose and/or insulin inthis cell

    type. Recent evidencessuggest that insulin blocksthestimulatoryeffectof D-gJucoseon L-arginine transport byreducing

    thetranscriptionalactivity ofSLC7A] via Spl-, NFKB-and ROS-dependent mechanisms. Thus, a role for these transcrip-

    tionfactorsin responseto insulin is proposed infetalendothelialcellsexposed tohyperglycaemia.

    Keywords: Glucose, hyperglycaemia, diabetes, L-arginine, transport, human, endothelium.

    Endothelial cells are of mesenchymal ongm forming a

    lane epithelium named endothelium. For decades the endo-

    elium was considered as a simply barrier between the

    lood and other body tissues. However, this early vision has

    now radically changed and currently holds the endothelium

    a real organ, made up of approximately ten trillion cells,

    ,-hich fulfils multiple roles in the physiology and patho-

    .hysiology of vascular tone, with autocrine, paracrine and

    c:J.docrine actions and involved in the processes of coagula-

    :::on and fibrinolysis [1, 2]. Endothelial cells are involved in

    :-egulation of vascular tone through the release of a number

    f vasoactive substances, such as prostacyclin (PGlz) [3],

    dotelin-l [4] and nitric oxide (NO) [3, 5]. Endothelium-

    -=crived NO spreads to the underlying smooth muscle cell

    ~yer in vascular vessels where increases cGMP levels to

    bsequently induce relaxation of the muscle leading to

    odilatation [3,5,6]. Both in vivo and in vitro experiments

    -c:w demonstrated that NO synthesis in endothelial cells is a

    __cess that could be stimulated by various molecules, in-

    ~ding insulin [7-9], D-glucose [10] and adenosine [6].

    ~ilarly, NO synthesis and/or its bioavailability are lower

    .".: ress correspondence to this author at the Cellular and M olecular

    _"iology Laboratory (CMPL), Department of Obstetrics and Gynaecol

    :..' 'vIedical Research Centre (CIM), School of Medicine, Faculty of Medi-

    Pontificia Universidad Cat6lica de Chile, P.O. Box 114-D, Santiago,

    - ~: Tel: 562-3548116; Fax: 562-6321924; E-mail: [email protected]

    in pathological conditions such as intrauterine growth restric-

    tion (IUGR) [11], diabetes mellitus [9,12] or atherosclerosis

    [13].

    The signalling mechanisms involved in NO synthesis

    have been studied in several cell types including human um-

    bilical vein endothelial cells (HUVEC). The gas NO is syn-

    thesized from the cationic, semi-essential amino acid L-

    arginine in a metabolic reaction leading to equimolar forma-

    tion of L-citrulline and NO [6, 14]. This reaction requires the

    activity of endothelial NO synthases (NOS), a group of en-

    zymes conformed by at least three isoforms i.e. neuronal

    NOS (nNOS or Type I), inducible NOS (iNOS or type II)

    and endothelial NOS (eNOS or Type III) [15]. In fact, there

    is evidence that NOS activity may depend on the ability of

    endothelial cells to take up its specific substrate L-arginine

    via a variety of membrane transport systems [6, 10, 11, 14-

    16]. L-Arginine is taken up by endothelial cells via mem-

    brane transport systems grouped as systems l,y+L, bO,+andBO.+[6, 16-18]. It is well known that system y+ conforms a

    family of proteins known as cationic amino acid transporters

    (CATs) family (hereafter referred as 'CATs family'), con-

    formed by CAT-I, CAT-2A, CAT-2B, CAT-3 and CAT-4

    [17, 19]. CAT-l is ubiquitously expressed, while CAT-2A

    and CAT -3 are constitutively expressed in liver and brain,

    respectively, and CAT-2B is induced in a variety of celltypes in response to bacterial endotoxins and pro-

    inflammatory cytokines [6,17, 19-21]. CAT-4 derives from

  • 8/10/2019 Sobrevia & Gonzalez 2009

    2/8

    a sequence of cDNA that is41-42% identical to other mem-

    bers of CATs family, but no transport activity has been yet

    described [6,17]. CAT-I, CAT-2B and CAT-3 are function-

    ally characterized as high affinity (Km ~100-400 f.lM),Na+..

    independent transporters, while CAT-2A has arelatively low

    affinity for cationic amino acids (Km ~2-5 mM). Interest-

    ingly, only 2 members of the CATs family have been func-tionally characterized in primary cultures of HUVEC i.e.

    human CAT-l (hCAT-l) and hCAT-2B, while hCAT-2A

    seems not to be expressed inthis cell type [6, 10, 11, 17, 18].

    In addition, hCAT-l protein and mRNA, and only hCAT-2B

    mRNA have been detected in this cell type [17].

    Phylogenetically, the SLC7 ('solute carrier') family con-

    sists of 2 subfamilies formed by CATs and the amino acid

    transporters associated to glycoproteins (HATs, heterodi-

    meric amino acid transporters). CATs family members are

    encoded by genes SLC7Ai, 2, 3 and 4, whose protein prod-

    ucts contain 14 tr ansmembrane domains [19]. Specifically,

    the gene encoding hCAT-1 protein i.e. SLC7 Ai (located onchromosome 13q 12-q 14), consists of an open reading fr ame

    with 11 exons and 10 introns. However, towards the 5'-end

    two additional untr anslatable exons (-2 and -1) have been

    described r..-:;.Th -.. ~. now suggested that SLC7Ai couldbe formed by I xo -. \yhere transcription start site (ATG)

    is located in exon -_ [_ ]. \- e have recently cloned a fr ag-

    ment of -6"0' u ~ the ATG of SLC7Ai in primary

    cultures of HC\"EC and detected that this fragment of the

    potential promo er of SCL Al contains several consensus

    sequences for transcription factors that may be critical for theregulation of its expression by insulin and/or D-glucose [23,

    24] (Fig. 1).

    INSULIN AND D-GL 'COSE EFFECT ON ENDOTHE-

    LIAL L-ARGININE/NO PATHWAY

    L-Arginine Transport

    Incubation of HUVEC primary cultures in an euglycae-

    mic culture medium (i.e. containing -5 m M D-glucose) with

    physiological r esting concentrations of insulin (-0.1 nM)

    increased the maximal transport velocity (Vmax) without sig-

    nificant changes in the apparent Km for L-arginine transport

    [23]. This phenomenon was seen as a higher maximal tr ans-

    port capacity (i.e. ~naJKm) [17, 20, 25, 26] of the involved

    membrane transporters for cationic amino acids [23]. The

    -650 GTTAAGGTGAACTGCGCTCCAGCCTGGGCAATAGAGTAACACCCTGTCTCTAAAATGAAAAGAAAACAG- - .-581 TTTAAACCTTTTAAGTGCATACCAAATCTTTTATTTTGGAGAAGGAAAACTGGTCTCGAGTTCCGTGTG

    P53

    -512 AGCTCCCTGGGGCCCGCCGGGAGGGGGTTGGCACGGCCGGACCTGCAGCACTAGTTCTGGCCAGGGCGC

    -443 TGTGGGATCTGCAGGGGACCACAGGATGCTGTGGCGCGGTGCGCTCAGATTGGCGGAGAAACGGCCACA

    EGR-1 NFKB (p50)

    -374 CGCCTACGGAGCTACTGAGAAGGCGAGCGGAGGCGCAGCCCGCCCGCCCGCCGCGGGAACCCCAGGTTG

    -305 GGGCGCTGGGCGCGCGAAGACTCAGCCGCCCCGCCCACCAAGGGCGCGTCGGTCCCCGGCCGCAGCCTC

    CREB Sp1

    -236 TGGGCTGGCAGCCGCCGCCGCGCCGCGCTCCCATTGGTGCCCGGCGGTGACGCGGCCGAGCGGGCCGGG

    Sp1 Sp1 Sp1

    -167 GCTGCCTGGTCCGGGGGCGGGCGTGGGGCGCGGGGCGCGGAGCGCGAGGGGCGGGGGCCGGGCGCACTG

    Elk- 1

    -98 CTGATGAAACCTGGCGCCGGAACCCGCCAGCCCTCGGCGCCCATTCAGTCCGCGCAGGCAGGTGTGAGC

    *

    +109 GAGCGCGTCCGACAGTCTGTCTGTTCGCGATCCTGCCGGAGCCCCGCCGCCGCCGGCTTG~

    Fig. (1). Scheme of the -650 bp region from proposed transcription start site at SLC7AI. Thesequenceofthe region -650 bpfromthe

    proposed transcriptionstartsite(*)ofthe potential promoter ofthe geneS LC7 A I (forhuman cationic amino acid transporter 1, hCAT-1) that

    hasbeencloned is shown.Proposed firstexon ofthetranscript extendsbetween -650to+ 142bp. Thelocation ofprimersusedtoamplifYthi

    region of thepromoter isindicatedbythe arrowsandcorrespondedto:sense ACGCGTTAA GGTGAA CTGCGC TCC, antisense CCATGGATC GCG AAC AGA CAG ACT.Underlinedare bindingsitesfor transcriptionfactors such asstimulatoryprotein 1(Spl), nuclear

    factorkappaB (NFKB), p53 tumor repressor factor,early growth responsefactor1 (EGR-l), cyclic AMP responseelement-binding (CREB)

    and the transcriptionfactorwith ETS domain (Elk- J) .

  • 8/10/2019 Sobrevia & Gonzalez 2009

    3/8

    (NT1 ONOO.

    L-Arginine

    hCAT-1 protein

    " heAT' mRNA

    Insulin ~

    ! -- /..ROSj~.....

    SplNF,B

    Fig. (2). Proposed regulatory mechanisms of hCAT-I expression and activity by high D-glucose and insulin in HUVEC. Exposure of

    HUVEC to an elevated extracellular level of D-glucose (High D-glucose) increases the intracellular level of reactive oxygen species (ROS)

    likely derived from NAD(P)H oxidase. ROS formation may stimulate via Sp I and NFkB transcription factors the SLC7AI gene promoter

    activity leading to increased hCAT-1 mRNA expression and protein abundance. These events result in an increased L-arginine tr ansport via

    heAT-I and nitric oxide (NO) and L-citrulline synthesis via the endothelial NO synthase (eNOS). The NO could rapidly react with ROS to

    fonn peroxynitrite (ONOO) contributing to endothelial dysfunction. Insulin isproposed to protect the endothelial cell by blocking the effect

    of D-glucose on the generation of intracellular ROS, reducing the effect of these molecules on SLC7Al expression and L-arginine transport.

    Insulin under hyperglycaemia could also act as a repressor of D-glucose activated ROS generation protecting the endothelial cells from this

    abnormal environment.

    reported magnitude of the insulin stimulatory effect on L-

    arginine transport (~5 fold) was comparable to the trans-

    -rimulatory effect of L-lysine on L-arginine transport meas-

    ured under zero-trans conditions for L-lysine in this cell type

    [II, 23] (see Fig. (2. These results are complemented and

    supported by parallel studies showing that insulin increased

    dlehCAT-I mRNA number of copies [23] and protein abun-

    ance (Gonzalez M, Sobrevia L, unpublished data). These

    -rudies suggest that insulin-induced L-arginine transport is

    most likely due to increased expression and activity of

    ,CAT-! membrane transporters in HUVEC. In addition,

    :l1Sulineffect on L-arginine transport was proposed as a phe-

    :lOmenon resulting from a sequential activation of cell sig-

    :1alling molecules, including phosphatidylinositol 3 kinase

    PI3k), protein kinase C (PKC), NOS and mitogen-activated

    rotein kinases p42 and p44 (p42/44mapk), which are known

    to be involved also in the modulation of several other mem-

    rane transport systems in mammalian cells [6, 17].

    It has been reported that incubation ofprimary cultures of

    ~VEC with increasing concentrations of extracellular D-

    glucose for a period of 2 minutes (acute effect) increased L-

    c:rginine transport, reaching a maximal effect at 25 mM D-

    _ucose [10]. The increase in L-arginine transport induced by

    ute exposure of HUVEC to high D-glucose has been ex-

    lained as an increase in the activity ofhCAT-! transporters.-;tis phenomenon is more likely due to plasma membrane

    yperpolarization due to increased K+ efflux through ATP-

    : nsitive K+ channels ([K +]ATP)[10]. It has also been re-

    ported that longer incubation periods (24 h) with high con-

    centrations of extracellular D-glucose (~25 mM) increases

    the ~nax of the L-arginine transport, an effect blocked by

    inhibition of protein synthesis or when cells are pre-

    incubated with 1 nM insulin for 8h [16]. The increase of L-

    arginine transport induced by chronic incubation with D-

    glucose was also associated with increased hCAT-l mRNA

    level and L_[3H] citrulline synthesis from L-eH] arginine

    [24]. In preliminary assays, we have been able to detect a

    potential effect of D-glucose at a transcriptional level modu-

    lating hCAT-! expression, a phenomenon that is also

    blocked by i nsulin (Gonzalez M, Sobrevia L, unpublished

    data). Thus, it is feasible that insulin could be triggering sig-

    nalling pathways that will potentially interfere with an in-

    creased SLC7Ai transcriptional activity in response to a high

    D-glucose environment.

    eNOS Expression and Activity

    In endothelial cells from human aorta, long incubation

    periods (7 days) with 25 mM D-glucose decreases eNOS

    activity, protein abundance and mRNA level, effects that

    was associated with r educed activity of eNOS promoter [27].

    In HUVEC, chronic incubation with 25 m M D-glucose (up

    to 24 h) increases eNOS protein abundance, an effect that

    seems involve P13k and protein kinase B (PKB)/Akt activity[24, 28]. Several studies report that in an early hyperglycae-

    mic state, activation of these signalling molecules would lead

    to cell survival, but after 2 4 h it will increase cell death by

  • 8/10/2019 Sobrevia & Gonzalez 2009

    4/8

    apoptosis, 'an effect regulated by activation of the nuclear

    factor kappa B (NFKB) [28]. This differential response of

    cells to states of hyperglycaemia could result f rom accumu-

    lation of reactive oxygen species (ROS), whose generation is

    elevated in cells exposed to high extracellular concentrations

    of D-glucose [28, 29]. On the other hand, in endothelial cells

    from mouse lungs it has been reported that insulin could pro-tect in a NO-dependent manner against endothelial dysfunc-

    tion (estimated by variations of the electrical trans-

    endothelial resistance) induced by hydrogen peroxide (HzOz)

    [30]. However, endothelial cells from bovine aorta (BAEC)

    exhibit less NO synthesis induced by insulin when cells were

    incubated with high concentrations of extracellular D-

    glucose, an effect that appears to depend on a signalling

    pathway involving insulin receptor type 1 (lR-I), PI3k and

    the inhibitor of the kinase subunit of nuclear factor kappa 13

    (IKKB) [31]. In studies performed in primary cultures of

    HUVEC, high D-glucose (25 mM, 24 h) increased cGMP

    synthesis, an effect blocked by 1 nM insulin (8 h) [17]. In the

    same cell type it has been shown that at this concentrationand time of exposure to insulin is enough to block the high

    D-glucose-dependent inhibition of the membrane transport

    of adenosine, an endogenous nucleoside that induces NO-

    depehdent vasodilatation in most vascular beds [32, 33]. All

    together these results indicate that deleterious effects of

    chronic exposure to high concentrations of extracellular D-

    glucose could be mediated by increased NO synthesis in

    human umbilical vein endothelium. Whether the effect of

    high D-glucose and the potential role of insulin as a protec-

    tive factor in this phenomenon is either at a transcriptional

    and/or post-transcriptional level has not yet been described[9, 17,33].

    INSULIN AND D-GLUCOSE EFFECT ON ROS

    SYNTESIS

    Cell death by apoptosis induced by high concentrations

    of extracellular D-glucose has been linked to the increased

    production of ROS. This is reflected as increased nitroty-

    rosine formation and PKC activity in HUVEC incubated in

    the presence of high concentrations of extracellular D-

    glucose, either under permanent (20 mM, 14 days) or inter-

    mittent (repeated periods of 24 hours with concentrations of

    5 or 20 mM D-glucose for 14 days) incubation periods [34].

    In BAEC, ithas been shown that incubation with 25 mM D-

    glucose for 3 h increases ROS synthesis dependent of

    NAD(P)H oxidase [35]. This study also shows that incuba-

    tion with physiological concentrations of insulin for 3 h in-

    duces an even greater increase in the synthesis of ROS, al-

    though during the first hour of incubation with this hormone

    is possible to detect a decline in the synthesis of ROS in-

    duced by D-glucose [35]. Thus, it is likely that an acute ef-

    fect of insulin could be acting to protect from the damage

    induced by overproduction of ROS in response to high con-

    centrations of extracellular D-glucose. This effect of insulin

    could probably be due to activation of NOS and increased

    NO bioavailability in the endothelium. The mechanisms by

    which ROS induces its effect include regulation of gene ex-

    pression and post-translational mod ifications [36]. One of

    the most studied transcription factor regulated by ROS is

    NFKB, which is found in most cells at rest, and that rapidly

    translocates to the nucleus inducing gene transcription after

    its a 'y ::0 ::1 . _'hBis also involved in the d evelopment of

    insulin res' .one of the first events that occur before the

    establ"-hm t 0 diabetes mellitus [37]. Although we know

    that imer D-glu 0e or insulin increase L-arginine transport

    and:\O . 'llth -is. a haracterization of the mechanisms be-

    hind the r ulting in reased ROS production and activation

    of transcription fac ors, including d1e zinc finger promoter-selectiYe transcription factor specific protein 1(Spl), in re-

    sponse to these molecules is not well established. Unveiling

    the intrinsic mechanisms behind modulation of gene tran-

    scriptional activity will give us clues to understand a poten-

    tial protective effect of insulin before endothelial damage is

    triggered bypathological states of hyperglycaemia.

    D-GLUCOSE AND I SULIN EFFECT ON Spl ACTI-

    VI TY

    Within aregion of~4000 bp upstream from the transcrip-

    tion start site of SLC7AI, multiple consensus sites of various

    transcription factors have been identified by insilica analysis

    of sequence. Within d1ese factors, it is possible to identify

    consensus sites for Smad-4, NFKB and Spl, among others,

    which are regulated by insulin or D-glucose. Within this re-

    gion d1ere are at least four consensus sites for Spl close to

    the transcription start site ofSLC7 AI. These sites could play

    key roles as regulators of the transcription of this gene since

    it lacks of TATA box [22]. Also, there are 2 consensus sites

    for NFKB around -3200 and -600 bp r egion from the tran-

    scription start site, which are potentially regulated by ROS.

    Since there is experimental evidence suggesting that Spl is

    involved in gene-specific responses to a variety of cellular

    signals independent of the interaction with other inducible

    transcription factors, and since this transcription factor is

    apparently involved in the regulation of membrane transport-

    ers expression, such as SLC29AI for the human equilibrative

    nucleoside transporters type 1(hENTl) in HUVEC [38], in

    preliminary experiments we have studied a region of 650 bp

    upstream of the transcription start site in SLC7AI [39]. Spl

    is activated by phosphorylation of serine and threonine resi-

    dues, a phenomenon altered in response to various stimuli

    that activate different signalling pathways. The amino acid

    sequence of Spl contains consensus phosphorylation sites

    for numerous protein kinases, including calmodulin kinases

    (CamKs), casein kinases (CK) 1 and 2, protein kinase A

    (PKA), PKC, and p42/44mapk [40]. Interestingly, it has been

    reported that increase in the tr anscriptional activity of plas-

    minogen activator inhibitor-l (PAl-I) induced by insulin

    (100 nM, 16 hours) in HepG2 cells transfected with PAI-l

    promoter, involves activity of PI3k, PKC and p42/44map\

    among other signalling molecules [41]. In PAI-I gene at

    least three consensus sites for transcription factors induced

    by insulin has been identified, all of which co-localize with

    putative consensus sites for SpI [41]. Thus, a potential role

    for Sp 1 as modulator of PAl-1 gene expression is suggested.

    In addition, in the same cell type, D-glucose incubation (~23

    mM, 24 and 48 h) decreases the level of apolipoprotein A 1

    (apoA-I) mRNA, an effect that was blocked by insulin. This

    phenomenon was dependent on a region of the promoter

    where an insulin response core element (IRCE) is present

    [42]. SpI binds to this element and the signallinp pathwaytriggered by insulin appears to involve p42/44map and PKC

    activity, and Spl phosphorylation [43].

  • 8/10/2019 Sobrevia & Gonzalez 2009

    5/8

    I

    There is also evidence that SpI is essential for basal tran-

    scription of calmodulin gene and for the increase of tran-

    scriptional activity of this gene induced by insulin [44]. In

    addition, insulin increases the o-glycosylation and phos-

    phorylation of SpI in the HAllE hepatoma cell line, which

    is reduced when cells are from streptozotocin-induced dia-betic rats [45]. Recently, it has been shown that insulin (100

    ) increases PKC8 expression, aphenomenon that is under

    ::cgulation by PKCa in the skeletal muscle cell line L6 via a

    :;:nechanismrequiring Sp 1and NFKB activation [46]. Since in

    rimary cultures of HUVEC it has been reported that insulin

    srimulation of hCAT- I -mediated L-arginine transport de-

    _ nds on PI3k, PKC and p42/44mapk activity [23], it is likely

    :hat SpI would be acting as akey transcription factor modu-

    ~ -ng SLC7Ai expression by insulin or high extracellular

    :uncentrations of D-glucose in HUVEC (Table 1).

    Preliminary experiments performed inHUVEC incubated

    'm 25 mM D-glucose (24 hours) and I nM insulin (for last

    h of the 24 h period of incubation with high D-glucose)-' ow increased SpI protein abundance in nuclear protein

    _ :::-acts(Gonzalez M, Sobrevia L, unpublished data). Co-

    _ -ubation of HUVEC monolayers with D-glucose and insu-

    lin produced a greater increase of S p I n uclear abundance,

    which would possibly indicates that both insulin and D-

    glucose activated diff erent signalling pathways to increase

    Sp I activity in this cell type. Although ithas not been possi-

    ble to determine whether it also leads to an increase in phos-

    phorylation or glycosy lation of Sp 1,we found increased S p 1

    binding to a region of the SLC7 Ai promoter containing four

    consensus sites for SpI in tandem. This finding indicates that

    at least part of the D-glucose- and insulin-increased hCAT-I

    mRNA level could be due to increased expression and activ-

    ity of Sp I on SLC7 Ai. Sp I activity seems to regulate not

    only hCA T-1 expression, but could also participate in the

    transcriptional regulation of eNOS. The promoter region of

    eNOS has been characterized and is known to exhibit con-

    sensus sites for transcription factors such as activator pro-

    tein-I (AP-I), GATA binding protein 2 (GATA-2), ETS

    binding domain (Ets) family members, and members of the

    Sp family (Spl and Sp3), among others [47]. On this back-

    ground it is tempting to propose that the signalling pathwaysinduced by D-glucose and insulin might include activation of

    Spl inducing expression of genes relevant to the endothelial

    L-arginine/NO pathway.

    I I Stimuli Gene Blank Cell Type Promoter Mechanism ReferencesActivity

    Insulin PAl-l Human hepatoblastoma Increased Increased Sp1binding to DNA [41]

    HepG2

    Insulin Apo-Al Human hepatoblastoma Increased Increased Spl binding to IRCE motifs [42]

    HepG2

    Insulin Apo-Al Human hepatoblastoma Increased Phpsphorylation of Sp 1 a nd increase of [43]

    HepG2 IRCE binding

    Insulin Calmodulin Rat hepatoma H-4 lIE Increased Promotor without Sp I binding sites, cannot [44]

    I I be stimulated by insulin

    Insulin PKC15 Skeletal muscle L6 Increased Inhibition of expression and activity of Sp I [46]

    blocked the effect of insulin on PKCo

    promoter

    J-Glucose TGF-j3l PAl-l Rat mesangial cells Increased Inhibition of expression and activity of SpI [61]

    blocked the effect of insulin on TGF-fil

    and PAI-I promoter

    J-Glucose lR-l Rat hepatocytes Increased Effect ofD-glucose on promoter was [62]

    blocked when 3 or 4 binding sites of SpI

    were mutated

    ._ ose Insulin Leptin Adipocytes and hepa- Increased Mutation ofSpl response element, blocked [63]

    tocytes from rat the D-glucose/insulin effect

    =>-Glucose Acetyl-CoA SL2 from Drosophila Increased Mutation of Sp 1binding sites, blocked the [64]

    effect of D-glucose

    H,O, Spl Nuclear extracts f rom n.d. H,O, decreased the Spl binding to DNA [65]

    rat kidney

    H,O, Aldolase Pyruvate Rat timocytes n.d. H,O, decreased the Sp I binding to DNA [66]

    kinase

  • 8/10/2019 Sobrevia & Gonzalez 2009

    6/8

    INSULIN ~D D-GLUCOSE EFFECT ON NFKB

    Since it was described in 1986, the transcription f actor

    NFKB has been the subject of many studies due to its impor-

    tant role in inflammatory processes and other pathologies. In

    the beginning NFKB was described as a specific protein of B

    cells, but it is now known to be an ubiquitous transcription

    factor [48]. NFKB activation is induced and independent of

    protein synthesis, requiring post-translational changes for its

    migration to the nucleus. Initially, NFKB was described as a

    transcription factor that could be stimulated by several im-

    munological stimuli, such as tumour necrosis factor (TN F),

    lipopolysaccharide (LPS) [49, 50], interleukin-I (IL-I) [51],

    or as a f actor that activates T cells [52]. However, more re-

    cently it has been shown that the transcriptional activity of

    several genes is increased via NFKB in response to a series of

    stimuli that are unrelated with the immune response, such as

    ultraviolet radiation, growth factors [53] or viral infections

    [54]. The latent nature of NFKB is in relation with it associa-

    tion with the inhibitory protein kappa B inhibitor (IKB). The

    release of this complex allows the migration of NFKB to the

    nucleus [55]. Interestingly, it has been reported that incuba-

    tion of BAEC in culture medium containing 30 mM D-

    glucose increases NFKB translocation toward the nucleus

    [56]. In addition, in HUVEC it has been shown that hyper-

    glycaemia also increased NFKB protein abundance in the

    nucleus, and that this increase depends on the activity of

    PI3kJAkt [57], signalling molecules that ar e determinant for

    L-arginine transport and NO synthesis in response to insulin

    in this cell type [17, 23]. Other studies also show that incuba-

    tion of BAEC and human aortic endothelial cells (HAEC) in

    30 mM D-glucose (16 hours) increases NFKB binding to the

    DNA [58]. Thus, it seems clear that high D-glucose effect on

    endothelial cells involves NFKB activation in a variety of

    endothelia indicating that this phenomenon is not limited to a

    specific type of endothelium. On the other side, insulin has

    been shOKl1 to either reduce NFKB binding to the DNA in

    HAEC [59] or increase )!FKB-induced transcriptional activ-

    ity of genes in '-ascular smooth muscle cells from bovine

    aorta (YSMC) transfected with a vector containing response

    elements to NFKB [60]. Thus, literature regarding the poten-

    tial involvement of NFKB in cell response to insulin is wide

    open and specific mechanisms are not well understood (Ta-

    ble 2).

    CONCLUDING REMARKS

    It is proposed that insulin modulates L-arginine transport

    and NO synthesis in human fetal endothelium via cell signal-

    ling mechanisms that respond to increased activity of Sp I

    andNFKB transcription factors. The gene SLC7 Al coding for

    hCAT -I membrane transporters express consensus se-

    quences for binding of t hese transcription factors, particu-

    larly S p I, suggesting that increased expression of hCA T-I in

    response to elevated D-glucose and insulin could result from

    increased expression ofSLC7 AI. Nothing is known regard-

    ing ROS and the transcriptional activity of SLC7AI in re-sponse to elevated D-glucose or in response to insulin, even

    when it is accepted that the pathological condition of hyper-

    glycaemia leads to generation of abnormal levels of ROS in

    HUVEC. These fmdings could be d eterminant in the dy-

    namic of the regulation of expression and activity of L-

    arginine membrane transporters (particularly hCA T-I) and

    eNOS in human fetal endothelium d erived from pregnancies

    where abnormally elevated plasma D-glucose levels could be

    found such as in gestational diabetes.

    Supported by Fondo Nacional de Desarrollo Cientifico y

    Tecnol6gico (FONDECYT 1070865, 1080534, 7070249)

    Stimuli Gene blank Cell type Effect on Mechanism References

    mRNA level

    D-Glucose NFJd3 BAEC n.d. Increased nuclear localization ofNFK.B [56]

    D-Glucose COX-2 HUVEC n.d. Increased NFKB DNA binding [57]

    D-Glucose NFKB HAEC n.d. Increased NFKB DNA binding [58]

    D-Glucose Fibronectin HUVEC Increased Increased NFKB DNA binding [67]

    D-Glucose VCAM-l HAEC Increased Increased NFKB DNA binding [68]

    D-Glucose Fibronectin HUVEC Increased Increased NFKB DNA binding [69]

    Insulin ?KCa L6 skeletal muscle Increased Inhibition ofNFKB activity blocked the [46]

    increase ofmRNA

    Insulin MC?-! HAEC Decreased Decreased NfKB DNA binding [59]

    ROS NFKB HUVEC n.d. Inhibition ofROS synthesis blocked ROS- [70]

    induced NFKB activity

    ROS NFKB HMEC.l n.d. LPS-increased ROS increased NfKB DNA [71]

    binding

    BAEC: bovine artery endothelialcells, HUVEC: humanumbilicalvein endothelialcells, HAEC: human arteryendotbelialcells, HMEC.I: human dermal microvascularendotheli2l

    cells,LPS: lipopolysaccharide,ROS: reactive oxygen species,n.d.: not determined.

  • 8/10/2019 Sobrevia & Gonzalez 2009

    7/8

    #

    dComisi6n Nacional de Ciencia y Tecnologia (CONICYT

    -~070213), Chile. M. Gonzalez holds a CONICYT-PhD

    ~!Jowship (Chile). We thank the researchers at the Cellular

    Molecular Physiology Laboratory (CMPL) and Perina-

    _ogyResearch Laboratory (PRL) of the Pontificia Univer-

    d Cat6lica de Chile (puq for their contribution in the__ duction of the experimental data that has been cited

    :=oughout the text. Authors also thank the personnel of the

    o pital Clinico Pontificia Universidad Cat6lica de Chile-,' urward for supply of umbilical cords.

    Fishman AP. Endothelium: A distributed organ of diverse capabili-

    ties. Ann NY Acad Sci 1982; 401: 1-8.

    Galley H, Webster N. Physiology of the endothelium. Br J Anaesth

    2004; 93: 105-13.

    Moncada S, P almer R MJ, Higgs EA. The discovery of nitric oxide

    as the endogenous nitrovasodilator. Hypertension 1988; 12: 365-

    72.

    Yanagisawa M, Kurihara H, Kimura S, Tomobe Y. A novel potent

    vasoconstrictor peptide produced by vascular endotelial cells.

    Nature 1988; 332: 411-5.

    Ignarro LJ, Buga GM, Wood KS, Byrns RE, Chaudhuri G. Endo-

    thelium-derived relaxing factor produced and released from artery

    and vein is nitric oxide. Proc Natl Acad Sci 1987; 84: 9265-9.

    Mann GE, Yudilevich DL, Sobrevia L. Rel,'Ulation of amino a cid

    and glucose transporters in endothelial and smooth muscle cells.

    Physiol Rev 2003; 83: 183-252.

    Steinberg HO, Baron AD. Vascular function, insulin resistance and

    fatty acids. Diabetologia 2002; 45: 623-34.

    Kuboki K, Jiang ZY, Takahara N, Ha SW, Igarashi M, Yamauchi

    T, et af. Regulation of endothelial constitutive nitric oxide synthase

    geneexpression in endothelial cells and in vivo: a specific vascular

    action of insulin. Circulation 2000; 101: 676-81.

    Desoye G, Hauguel-de Mouzon S. The human placenta in gesta-

    tional diabetes mellitus. The insulin and cytokine network. Diabe-

    tes Care 2007; 30: 120-6.

    Flores C, Rojas S, Aguayo C, Parodi J, Mann G, Pearson JD, et al.

    Rapid stimulation of L-arginine transport by D-glucose involves

    p42/44mapk and nitric oxide in human umbilical vein endothelium.

    Cir Res 2003; 92: 64-72.

    Casanello P, Sobrevia L. Intrauterine growth retardation is associ-

    ated w ith reduced activity and expression of the cationic amino

    acid transport systems y+/hCA T-l and y+/hCAT-2B and lower ac-

    tivity of nitric oxide synthase in human umbilical vein endothelial

    cells. Circ Res 2002; 91: 127-34.

    Durante W, Sen AK, Sunahara FA. Impairment of endothelium-

    dependent relaxation in aortae from spontaneously diabetic rats. Br

    JPharmacol 1988; 94: 463-8.

    Bossaller C, Habib GB, Yamamoto H, Williams C, Wells S, Henry

    PD. Impaired muscarinic endothelium-dependent relaxation and

    cyclic guanosine 5'-monophosphate formation in atherosclerotic

    human coronary artery and rabbit aorta. J Clin Invest 1987; 79:

    170-4.

    Guyao W, Morris SM. Arginine metabolism: nitric oxide and be-

    yond. Biochem J 1998336: 1-17.

    Alderton WK, Cooper CE, Knowles RG. Nitric oxide synthases:

    structure, function and inhibition. Biochem J200I; 357: 593-615.

    Sobrevia L, Mann GE. Dysfunction of the nitric oxide signalling

    pathway in diabetes and hyperglycaemia. Exp Physiol 1997; 82:

    423-52.

    Casanello P, Escudero C, Sobrevia L. Equilibrative Nucleoside

    (ENTs) and Cationic Amino acid (CATs) Transporters: implica-

    tions in foetal endothelial dysfunction in human pregnancy dis-

    eases. Curr Vasc Pharmacol2007; 5: 6 9-84.

    Arancibia-Garavilla Y, Toledo F, Casanello P, Sobrevia L. Nitric

    oxide synthesis requires activity of the cationic and neutral amino

    acid transport system y+L in human umbilical vein endothelium.

    Exp Physiol2003; 88: 699-710.

    Verrey F, Closs EI, Wagner CA, Palacin M, Endou H, Kanai Y.

    CATs and HATs: the SLC7 family of amino acid transporters. Eur

    J Physiol2003; 447: 532-42.

    [20] Deves R, Boyd CAR. Transporters for cationic amino acids in

    animal cells: Discovery, structure and function. Physiol Rev 1998;

    78: 487-545.

    [21] Palacin M, Estevez R, Bertran J, Zorzano A. Molecular biolog y of

    mammalian plasma membrane amino acid transporters. Physiol

    Rev 1998; 78: 969-1054.

    [22] Hammermann R, Brunn G, R acke K. Analysis of the genomic

    organization of the human cationic amino acid transporters CA T-l,

    CAT-2 and CAT-4. Amino Acids 2001; 21: 211-9.

    [23] Gonzalez M, Flores C, Pearson JD, Casanello P, Sobrevia L. Cell

    signalling-mediating insulin increase ofmRNA expression for cati-

    onic amino acid transporters-l and -2 and membrane hyperpolari-

    zation in human umbilical vein endothelial cells. Eur J Physiol

    2004; 448: 383-94.

    [24] Vasquez R, Farias M, Lvis VJ, San MR, Vecchiola A, Casanello P,

    et al. D-Glucose stimulation of L-arginine transport and nitric ox-

    ide synthesis results from activation of mitogen-activated protein

    kinases p42/44 and Smad2 requiring functional type II TGF- Re-

    ceptors in human umbilical vein endothelium. J Cell Physiol 2007;

    212: 626-32.

    [25] Escudero C, Casanello P, Sobrevia L. Human equilibrative nucleo-

    side transporters I and 2 may be differentially modulated by A2B

    adenosine receptors in placenta microvascular endothelial cells

    from preeclampsia. Placenta 2008; 29: 816-25.

    [26] Vasquez G, Sanhueza F, Vasquez R, Gonzalez M, San Martin R,

    Casanello P,et al. Role of adenosine transport in gestational diabe-

    tes-induced L-arginine transport and nitric oxide synthesis in hu-

    man umbilical vein endothelium. J Physiol2004; 560: 111-22.

    [27] Srinivasan S, Hatley ME, Bolick DT, Palmer LA, Edelstein D,

    Brownlee M, et al. Hyperglycaemia-induced superoxide production

    decreases eNOS expression via AP-l activation in aortic endothe-

    lial cells. Diabetologia 2004; 47: 1727-34.

    [28] Ho FM, Lin WW, Chen BC, Chao CM, Yang CR, Lin LY, et al.

    High glucose-induced apoptosis in hu man vascular endothelial

    cells is mediated through NF-kappaB and c-Jun NH2-terminal

    kinase pathway and prevented by PI3K/AktieNOS pathway. Cell

    Signal 2006; 18:391-9.

    [29] Hadi HA, Suwaidi JA. Endothelial dysfunction indiabetes mellitus.

    Vasc Health Risk Manag. 2007; 3: 853-76.

    [30] Rath S, Kalogeris T, Mai N, Zibari G, Alexander JS, Lefer D, et af.

    Insulin prevents oxidant-induced endothelial cell barrier dysfunc-

    tion via nitric oxide-dependent pathway. Surgery 2006; 139: 82-91.

    [31] Kim F, T ysseling KA, Rice J, Gallis B, Haji L, Giachelli CM, et af.

    Activation of IKKbeta by glucose isnecessary and sufficient to im-

    pair insulin signaling and nitric oxide production in endothelial

    cells. J Mol Cell Cardiol2005; 39: 327-34.

    [32] Munoz G, San Martin R, Farias M, Cea L, Vecchiola A, Casanello

    P, etal. Insulin r estores glucose inhibition of adenosine transport

    by increasing the expression and activity of the equilibrative nu-

    cleoside transporter 2 in human umbilical vein endothelium. J Cell

    Physiol 2006; 209: 826-35.

    [33] San Martin R, Sobrevia L. Gestational diabetes and t he adeno-

    sinelL-arginine/nitric oxide (ALANO) pathway in human umbilical

    vein endothelium. Placenta 2006; 27: 1-10.

    [34] Quagliaro L, Piconi L, Assaloni R, Martinelli L, Motz E, Ceriello

    A. Intermittent high glucose enhances apoptosis related to oxida-

    tive stress in human umbilical vein endothelial cells: the role of

    protein kinase C and NAD(P)H-oxidase activation. Diabetes 2003;

    52: 2795-804.[35] Yano M, Hasegawa G, Ishii M, Yamasaki M, Fukui M, Nakamura

    N, et al. Short-term exposure of high glucose concentration ind uces

    generation of reactive oxygen species in endothelial cells: implica-

    tIOn for the oxidative stress associated with postprandial hypergly-

    cemia. Redox Rep 2004; 9: 111-6.

    [36] Carnesecchi S, Carpentier JL, Foti M, Szanto 1. Insulin-induced

    vascular endothelial growth factor expression is mediated by the

    NADPH oxidase NOX3. Exp Cell Res 2006; 3 I 2: 3413-24.

    [37] Rutledge AC, Adeli K. Fructose and the metabolic syndrome:

    pathophysiology and molecular mechanisms. Nutr Rev 2007; 65:

    SI3-S23.

    [38] Puebla C, Farias M, Gonzalez M, Vecchiola A, Aguayo C, Krause

    B, et al. High D-glucose reduces SL C29A1 promoter activity and

    adenosine transport involving specific protein I in h uman umbilical

    vein endothelium. J C ell Physiol2008; 215: 645-56.

    [39] Gonzalez M, Vecchiola A, Farias M, Casanello P, Sobrevia LInsulin blockage of D-glucose-stimulation of L-arginine trans

  • 8/10/2019 Sobrevia & Gonzalez 2009

    8/8

    does not involves Spl binding to SLC7Alpromoter region in hu -

    man fetal endothelium. Physiol Mini-Rev 2006; I: 28 (Abstract).

    Samson S, Wong NCW. Role of Spl in insulin regulation of gene

    expression. J Mol Endocrinol 2 002; 29: 265-79.

    Banfi C, Eriksson P, Giandomenico G, Mussoni L, Sironi L, Ham-

    sten A, et al. Transcriptional regulation of plasminogen activator

    inhibitor type I gene by insulin: insights into the signaling path-

    way. Diabetes 2001; 50: 1522-30.

    Murao K, Wada Y, Nakamura T, Taylor AH, Mooradian AD,

    Wong NC. Effects of glucose and insulin on rat apolipoprotein A-Igene expression. J Bioi Chern 1998; 273: 18959-65.

    Lam JK, Matsubara S, Mihara K, Zheng XL, Mooradian AD,

    Wong Ne. Insulin induction of apolipoprotein AI, role of Spi.

    Biochemistry 2003; 42: 2680-90.

    Solomon SS, Palazzolo MR, Takahashi T, Raghow R. Transcrip-

    tion factor SpI is necessary for basal calmodulin gene transcription

    and for its selective stimulation by insulin. Endocrinology 1997;

    138: 5052-64.

    Majumdar G, Harrington A, Hungerford J, Martinez-Hernandez A,

    Gerling IC, Raghow R, et at. Insulin dynamically regulates

    calmodulin gene expression by sequential o-glycosylation and

    phosphorylation of Spl and its subcellular compartmentalization in

    liver cells. J Bioi Chern 2006; 281: 3642-50.

    Horovitz-Fried M, Sampson SR. Involvement of PKCalpha in

    insulin-induced PKCdelta expression: Importance of SP-I and

    NFkappaB transcription factors. Biochem Biophys Res Commun

    2007; 352: 78-83.

    Fish JE, Marsden PA. Endothelial nitr ic oxide synthase: insight

    into cell-specific gene regulation in the vascular endothelium. Cell

    Mol Life Sci 2005; 63: 144-62.

    Grimm S, Baeuerle PA. The inducible transcription factor NF-

    kappa B: structure-function relationship of its protein subunits.

    Biochem J 1993; 290: 297-308.

    Crisostomo PR, Wang Y, Markel TA, Wang M, Lahm T, Meldrum

    DR. Human mesenchymal stem cells stimulated by TNF-alpha,

    LPS, or hypoxia produce growth factors by an NF kappa B- but not

    JNK-dependent mechanism. Am J Physiol 2008; 294: C675-C682.

    Nakao S, Oglata Y, Shimizu E, Yamazaki M, Furuyama S, Sugiya

    H. Tumor necrosis factor alpha (TNF-alpha)-induced prostaglandin

    E2 release is mediated by the activation of cyclooxygenase-2

    (COX-2) transcription via NFkappaB in human gingival fibro-blasts. Mol Cell Biochem 2002; 238: 11-8.

    Jung YJ, Isaacs JS, Lee S, Trepel J, Neckers L. IL-I bela-mediated

    up-regulation of HIF-lalpha via an NFkappaB/COX-2 pathway

    identifies HIF-I as a critical l ink between inflammation and onco-

    genesis. FASEB J2003; 17:21 15-7.

    Schaecher K, Goust JM, Banik NL. The effects of calpain inhibi-

    tion on IkB alpha degradation after activation of PBMCs: identifi-

    cation of the calpain cleavage sites. Neurochem Res 2004; 29:

    1443-5 I.

    Chetty A, Cao GJ, Nielsen He. Insulin-like growth factor-I signal-

    ing mechanisms, type I collagen and alpha smooth muscle actin in

    human fetal lung fibroblasts. Pediatr Res 2006; 60: 389-94.

    Kuhnel F, Zender L, Paul Y, Tietze MK, Trautwein C, Manns M, et

    al. NFkappaB mediates apoptosis through transcriptional activation

    of Fas (CD95) in adenoviral hepatitis. J Bioi Chern 2000; 275:

    6421-7.

    Baldwin AS Jr. The NF-kappa B and I kappa B proteins: new di;-

    coveries and insights. Annu Rev Immunol1996; 14: 649-83.

    Nishikawa T, Edelstein D, Du XL, Yamagishi S, Matsumura -

    Kaneda Y, et al. Normalizing mitochondrial superoxide pr oducti

    blocks three pathways of hyperglycaemic damage. Nature 2000

    404: 787-90.

    Sheu ML, Ho FM, Yang RS, Chao KF, Lin WW, Lin-Shiau SY,

    al. High glucose induces human endothelial cell apoptosis thro _

    a phosphoinositide 3-kinase-regulated cyclooxygenase-2 pathwz;

    Arterioscler Thromb Vasc BioI 2005; 25: 539-45.Mohan S, Hamuro M, Koyoma K, Sorescu GP, Jo H, Natarajan . l

    High glucose induced NF-kappaB DNA-binding activity in HAEC

    is maintained under low shear stress but inhibited u nd er high sh=

    stress: role of nitric oxide. Atherosclerosis 2003; 171: 225-34.

    Aljada A, Ghanim H, Saadeh R, Dandona P. Insulin inhibi-

    NFkappaB and MCP-I expression in human aortic endothelia.

    cells. J Clin Endocrinol Metab 2001; 86: 450-63.

    Golovchenko I,Goalstone ML, Watson P, Brown lee M, Draznin B

    Hyperinsulinemia enhances transcriptional activity of nuclear fa -

    tor-kappaB induced by angiotensin II, hyperglycemia, and ad-

    vanced glycosylation end products in vascular smooth muscle cells.

    Circ Res 2000; 87: 722-4.

    Chae YM, Park KK, Magae J, Lee IS, Kim CH, Kim HC, el a~

    Sp I-decoy oligodeoxynucleotide inhibits high glucose-induced me-

    sangial cell proliferation. Biochem Biophys Re s Commun 200!

    319: 550-5.

    Fukuda H, Noguchi T, Iritan i N. Transcriptional regulation of inS1..

    lin receptor gene promoter in rat hepatocytes. Biochem Biophy:

    Res Commun 2001; 280: 1274-8.

    Fukuda H, Iritani N. Transcriptional regulation of leptin gene pro-

    moterinrat. FEBSLett 1999;455: 165-9.

    Daniel S, Kim KH. Spl mediates glucose activation of the acetyl-

    CoA carboxylase promoter. JBioi Chern 1996; 271: 1385-92.

    Ammendola R, Mesuraca M, Russo T, Cimino F. The DNA-

    binding efficiency of Spl is affected by redox changes. Eu r J Bio-

    chern 1994; 225: 483-9.

    Schafer D, Hamm-Kunzelmann B, Herrnfisse U, Brand K. Differ-

    ences in DNA-binding efficiency of SpI to aldolase and pyruva ~

    kinase promoter correlate with altered redox states in resting ane

    proliferating rat thymocytes. FEBS Lett 1996; 391: 35-8.

    Xin X, Khan ZA, Chen S, Chakrabarti S. Glucose-induced AI..,:activation mediates fibronectin synthesis in endothelial cells. Dia-

    betologia 2005; 48: 2428-36.

    Kouroedov A, Eto M, Joch H, Volpe M, Luscher TF, Cosentino F

    Selective inhibition of protein kinase Cbeta2 prevents acute effec

    of high glucose on vascular cell adhesion molecule-I expression -

    human endothelial cells. Circulation 2004; 110: 91-6.

    Chen S, Mukherjee S, Chakraborty C, Chakrabarti S. High gluco-~-

    induced, endothelin-dependent fibronectin synthesis is medial

    via NF-kappa Band AP-l. Am J Physiol Cell Physiol 2003; 2

    C263-72.

    Matsunaga T, Hokari S, Koyama I, Harada T, Komoda T. 1\T-

    kappa B activation in endothelial cells treated with oxidized hig

    density lipoprotein. Biochem Biophys Res Commun 2003; 30:'

    313-9.

    Chan EL, Murphy JT. Reactive oxygen species mediate endotoxID-

    induced human dermal endothelial NF-kappaB activation. J SUl1"

    Res 2003; III: 120-6.