Articulo bioquimica

Embed Size (px)

DESCRIPTION

bioquimica

Citation preview

  • The International Journal of Biochemistry & Cell Biology 69 (2015) 5261

    Contents lists available at ScienceDirect

    The International Journal of Biochemistry& Cell Biology

    jo u r n al homep ag e: www.elsev ier .com/ locate /b ioce l

    Inhibition of MEF2A prevents hyperglycemia-indmatrix accumulation by blocking Akt and TGF-1cardiac broblasts

    Xueying Chena, Guoliang Liub, Wei Zhanga, Jianing Zhangc, YuWenqian Donga, Ershun Lianga, Yun Zhanga, Mingxiang Zhanga The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education anQilu Hospital ob Henan Provin ity, Kac School of Fored Department o

    a r t i c l e i n f o

    Article history:Received 15 June 2015Received in re19 SeptemberAccepted 13 O

    Keywords:HyperglycemiDiabetesCardiac brobMyocyte enhaCardiac remod

    a b s t r a c t

    Myocyte enhancer factor 2A (MEF2A) functions in muscle-specic and/or growth factor-related tran-

    1. Introdu

    Diabetesease that ca

    Abbreviatinormal glucosprotein kinasetozotocin; MOtissue inhibitoJun NH2-term8, Cell Countininterventriculend-systolic dshortening; E/matrix.

    CorresponXi Road, Jinan,

    E-mail add

    http://dx.doi.o1357-2725/ vised form 2015ctober 2015

    c

    lastsncer factor 2Aeling

    scription and is involved in cell growth, survival, and apoptosis. To evaluate the role of this transcriptionfactor in cardiac broblasts (CFs) in diabetes mellitus, we performed a series of in vitro and in vivo experi-ments. We used short hairpin RNA (shRNA) to inhibit the expression of MEF2A in CFs in vitro. Inhibition ofMEF2A signicantly reduced hyperglycemia-induced CF proliferation and migration, myobroblast dif-ferentiation, matrix metalloproteinase (MMP) activities, and collagen production. Furthermore, MEF2Ainhibition attenuated HG-induced activation of the mitogen-activated protein kinase (MAPK), Akt, andTGF-1/Smad signaling pathways. For in vivo analysis in a mouse model, type-1 diabetes was inducedby streptozotocinand MEF2A expression was knocked down by myocardial injection with lentiviruscarrying shRNA-MEF2A. Cardiac function was assessed by echocardiography. Total collagen depositionwas assessed by Massons trichrome and Picrosirius red staining. Knockdown of MEF2A ameliorateddiabetes-induced cardiac dysfunction and collagen deposition. Our study suggests that inhibition ofMEF2A could alleviate HG-induced extracellular matrix accumulation by blocking the activation of Aktand TGF-1/Smad signaling pathway in CFs. Thus, inhibition of MEF2A has therapeutic potential in thetreatment of diabetic-induced cardiac remodeling.

    2015 Elsevier Ltd. All rights reserved.

    ction

    is an independent risk factor for cardiovascular dis-n cause cardiac remodeling and heart failure (Ban and

    ons: MEF2A, myocyte enhancer factor 2A; CFs, cardiac broblasts; NG,e; HG, high glucose; OC, osmotic control; MAPK, mitogen-activateds; shRNAs, short-hairpin RNAs; RNAi, RNA interference; STZ, strep-I, multiplicity of infection; MMP, matrix metalloproteinase; TIMP1,r of metalloproteinase; TGF-1, transforming growth factor-1; JNK,inal kinase; ERK1/2, extracellular-regulated protein kinase 1/2; CCK-g Kit-8; PE, phycoerythrin; 7-AAD, 7-amino-actinomycin D; IVS, thear septum; LVEDD, left ventricular end-diastolic dimension; LVESD, LVimension; LVPW, LV posterior wall; EF, ejection fraction; FS, fractionalA, the ratio of early to late mitral inow velocity; ECM, extracellular

    ding author at: Shandong University, Qilu Hospital, No. 107, Wen Hua Shandong 250012, China.ress: [email protected] (M. Zhang).

    Twigg, 2008). Cardiac remodeling is one of the major pathophysi-ological processes contributing to increased myocardial stiffnessand hampered systolic ejection, which eventually lead to heartfailure (Kong et al., 2014). However, no curative treatment for car-diac remodeling has been developed to date. Cardiac broblasts(CFs) are the critical mediators of physiological and pathologicalcardiac remodeling. CFs respond to stimuli in various ways, includ-ing proliferation, migration, myobroblast differentiation, matrixgeneration and degradation, secretion of cytokines etc. (Souderset al., 2009). Many studies (Bugyei-Twum et al., 2014; Shamhartet al., 2014; Tang et al., 2007) have shown that high glucose (HG)stimulates cardiac remodeling by inducing CF proliferation andactivation; however, the molecular mechanisms underlying CF-mediated cardiac remodeling are not clear.

    Myocyte enhancer factor 2 (MEF2) is a widely distributed DNA-binding transcription factor that activates various muscle-specicand growth factor- and stress-induced genes (McKinsey et al.,2002). MEF2 DNA-binding activity depends on four separate gene

    rg/10.1016/j.biocel.2015.10.0122015 Elsevier Ltd. All rights reserved.f Shandong University, Jinan, Shandong, Chinacial Key Engineering Laboratory of Antibody Drugs, School of Medicine, Henan Universign Languages and Literature, Shandong University, Chinaf Medicinal Chemistry, School of Pharmacy, Shandong University, Chinauced extracellular/Smad activation in

    gang Yand,a,

    d Chinese Ministry of Public Health,

    ifeng, Henan, China

  • X. Chen et al. / The International Journal of Biochemistry & Cell Biology 69 (2015) 5261 53

    products in mammals, referred to as MEF2A-D (Black and Olson,1998; Molkentin and Olson, 1996). MEF2A and MEF2D are the pri-mary MEF2s expressed in adult heart. Multiple lines of evidencesuggest that MEF2s play prominent roles in the regulation of cardiachypertroph2009; Xu etin responsediomyocyteor MEF2C ieration andthese studieocytes; the cardiac rem

    Numero2003, 2013pathways, induced carsignaling paet al., 2001;cardiac rem

    We hypohyperglycemliferation anprotective athese hypoexperiment

    2. Materia

    2.1. Lentivi

    A lentivireporter geused to expexpression MEF2A) wacontrol sequ

    2.2. Animal

    Eight-weused for exzotocin (STZet al., 2014were dividelentiviral vement, 1 1same volumple sites. Afconformed published bethical com

    2.3. Primar

    CFs werneonatal mcell morphoperformed cells with n

    Supplemonline versi

    The CFs and shRNA24 h. Then,

    glucose, NG), 33 mM d-glucose (high glucose, HG), or 5 mM d-glucose + 27.5 mM mannose (osmotic control; OC) for 48 h. The cellswere observed under a uorescence microscope to conrm thatmore 90% of the cells were GFP-positive.

    mun

    wereizedboviere i, Cam

    -SMAn LaidinagesLSM

    ester

    teinsrylamyvinyd w

    andainst

    meinhibAPK,llulaor

    adish temscen

    anti

    priTGGA CATCreened fa reafold cd.

    easu

    liferaotimnufarything s, Fons.

    ll mi

    l migion a, scraing 2 h, ell. Tcialy (Kim et al., 2008b; Konno et al., 2010; Pereira et al., al., 2006). MEF2 DNA-binding activities are enhanced

    to biomechanical and neurohormonal stimuli in car-s (Nadruz et al., 2003,2005). Overexpression of MEF2An cultured cardiomyocytes induces sarcomere degen-

    cardiomyocyte elongation (Xu et al., 2006). However,s mainly focused on the effects of MEF2A on cardiomy-function of MEF2A in CFs in diabetes mellitus-inducedodeling remains unknown.us signaling pathways (Kim et al., 2008a; Li et al.,; She et al., 2012), such as the MAPK and TGF-/Smadare implicated in the mediation of hyperglycemia-diac remodeling. MEF2A is intimately linked with thesethways (Liu et al., 2004; Ornatsky et al., 1999; Quinn

    Wales et al., 2014). Therefore, MEF2A may participate inodeling through these signaling pathways in diabetes.thesized that an increased level of MEF2A may facilitateia-induced cardiac remodeling via regulating CF pro-d activation. Knockdown of cardiac MEF2A may have affection in diabetic cardiomyopathy. In order to provetheses, we performed a series of in vitro and in vivos.

    ls and methods

    ral vectors for RNA interference (RNAi)

    ral vector containing a green uorescent protein (GFP)ne and a U6 promoter upstream of the cloning site wasress short-hairpin RNAs (shRNAs) to suppress MEF2Avia RNAi. The target sequence for MEF2A (ShRNA-s 5-GCTTGCCACCTCAGAACTTCT-3 and the negativeence (shRNA-NC) was 5-TTCTCCGAACGTGTCACGT-3.

    model and RNAi

    ek-old C57BL/6J wild-type (WT) mice (2530 g) wereperiments. Diabetes was induced by injecting strepto-) (Sigma, St. Louis, MO) as described previously (Wang). After induction of diabetes (12 weeks), the miced into 3 groups for treatments: shRNA-MEF2A (n = 10),hicle (n = 10) and shRNA-NC control (n = 10). For treat-07 UT/30 l of lentivector withMEF2A-shRNA or thee of vehicle was injected into the left ventricle at multi-ter 16 weeks, the mice were sacriced. All experimentsto the Guide for the Care and Use of Laboratory Animalsy the US National Institutes of Health and approved bymittee of Shandong University.

    y mouse CF culture and treatments

    e isolated from ventricular tissues of 1- to 3-day-oldice. CFs were identied by immunouorescence andlogical analysis (Supplementary Fig. S1). CF culture wasas described previously (Dubey et al., 1997). We usedo more than 4 passages.entary Fig. S1 related to this article can be found, in theon, at http://dx.doi.org/10.1016/j.biocel.2015.10.012.were infected with lentiviruses carrying ShRNA-MEF2A-NC at a multiplicity of infection (MOI) of 20 for

    the cells were exposed to 5 mM d-glucose (normal

    2.4. Im

    CFsmeabilin 5% cells w(Abcamanti-(Jackso6-diamand imscope (

    2.5. W

    Propolyacto polblockefor 2 hies agmatrixtissue p38 Mextracep-Akt, horserat roomlumine

    2.6. Qu

    TheGGAGCTAGGCGTGCCSYBR Gwas ususing mean metho

    2.7. M

    Pro8; Beythe maPhycoecontainsciencedirecti

    2.8. Ce

    CelmigratBrieycontainAfter 7cells/wan artiouorescence

    xed in 4% paraformaldehyde for 15 min and then per- in 0.03% Triton X-100 for 20 min. After being blockedne serum albumin at room temperature for 1 h, thencubated with primary antibodies mouse anti-MEF2Abridge, MA), rabbit anti-p-MEF2A (Abcam), and mouse

    (SigmaAldrich). Subsequently, secondary antibodiesboratories) were added. Nuclei were visualized with 4-o-2-phenylindole (5 mg/ml; Beyotime, Haimen, China)

    were acquired using a laser scanning confocal micro-710; Carl Zeiss, Germany).

    n blot analysis

    were separated by 10% sodium dodecyl sulfate-ide gel electrophoresis (SDS-PAGE) and transferredlidene diuoride membranes. The membranes wereith 5% non-fat milk in TBST at room temperature

    incubated overnight at 4 C with primary antibod- MEF2A, p-MEF2A, -SMA, collagen I, collagen III,talloproteinase 2 (MMP2), MMP9 (all from Abcam),itor of metalloproteinase 1 (TIMP1), TIMP2, TGF-1,

    p-p38 MAPK, Jun NH2-terminal kinase (JNK), p-JNK,r-regulated protein kinase 1/2 (ERK1/2), p-ERK1/2,Akt,-actin (all from Cell Signaling Technology). Then,

    peroxidase-conjugated secondary antibody was addedperature for 2 h. Membranes were visualized by chemi-ce reagent.

    tative RT-PCR

    mers for MEF2A were as follows: forward, 5-AAATAGTCCTGTGG-3, reverse, 5-GGGAGCTTTG-TGACT-3; and for -actin: forward, 5-CACT-TACGA-3, reverse, 5-GTAGTCTGTCAGGTCCCG-3.

    RT-PCR master mix (Life Technologies, Carlsbad, CA)or reactions and quantitative assays were performedl-time PCR detection system (Bio-Rad). The relativehange in expression was calculated using the 2CT

    rement of CF proliferation and apoptosis

    tion was analyzed by using Cell Counting Kit-8 (CCK-e) and Cell-LightTMEdU assay (RiboBio) according tocturers directions. Apoptosis was detected by using arin (PE)-Annexin V Apoptosis Detection Kit (BD559763,7-amino-actinomycin D (7-AAD) marker; BD Bio-ranklin Lanes, NJ) according to the manufacturers

    gration assays

    ration was examined by scratch assay and Transwellssay as described previously (Ibrahim et al., 2015).tch assay was performed as follows: lentiviral vectorsshRNA-NC or shRNA-MEF2A were transfected into CFs.the cells were seeded into a 6-well plate at 1 105he next day, the cells were treated with HG or OC. Then,

    gap was generated by scratching with apipette tip. The

  • 54 X. Chen et al. / The International Journal of Biochemistry & Cell Biology 69 (2015) 5261

    Fig. 1. High glucose (HG) induces MEF2A expression increase and nuclear translo-cation in cardiac broblasts (CFs). (A) CFs were treated with HG for various periods,and MEF2A expression was analyzed by western blotting. (B) Immunocytouores-cence of MEF2A translocation. Scale bar: 25 m. MEF2A stained red; nuclei werecounterstained with DAPI (blue). (C) Immunocytouorescence of p-MEF2A. Scalebar: 25 m. P-MEF2A stained red; nuclei were counterstained with DAPI (blue).(D) Western blot analysis of MEF2A and p-MEF2A protein expression in cyto-plasm (D1) and nucleus (D2). NC: 5 mM glucose, HG: 30 mM glucose, OC: 5 mMd-glucose + 27.5 mM mannose. Data are the mean SD of 3 independent experi-ments; *P < 0.05 vs. NC. (For interpretation of the references to color in this gurelegend, the reader is referred to the web version of this article.)

    Fig. 2. MEF2A inhibition reduces the proliferation of cardiac broblasts (CFs). (A)Cell Counting Kit-8 analysis of the cell viability of CFs treated with glucose forvarious periods. (B) Laser confocal microscopy of Edu staining. Nuclei were coun-terstained with DAPI (blue), red staining indicates cells undergoing proliferation.Scale bar: 25 m. (C) The Edu-positive index was expressed as a percentage (posi-tive/total cell number). (D) Quantitative analysis of the phycoerythrin (PE)-positiverate. (E) Flow cytometry using PE and 7-amino-actinomycin D (7-AAD) staining todetermine cell apoptosis. NC: 5 mM glucose, HG: 30 mM glucose, shRNA-NC+HG:cells transfected with MEF2A negative shRNA control before glucose incubation, sh-MEF2A+HG: cells transfected with MEF2A-shRNA before glucose incubation. OC:5 mM d-glucose + 27.5 mM mannose. Data are mean SD of 3 independent exper-iments; *P < 0.05 vs. NC; #P < 0.05 vs. HG. (For interpretation of the references tocolor in this gure legend, the reader is referred to the web version of this article.)

  • X. Chen et al. / The International Journal of Biochemistry & Cell Biology 69 (2015) 5261 55

    Fig. 3. MEF2Ashowing that Scale bar: 100at different tithat MEF2A kncultured with nal surface ofunder a microthe Transwell sample from 3shRNA-NC+HGcose incubatioincubation. OCindependent ethe referencesof this article.)

    number of 12-h interv

    Transweswell units from Corninchamber, w inhibition reduces cardiac broblast (CF) migration. (A) Scratch assayMEF2A knockdown inhibited CF migration at different time points.

    m. (B) Quantication of the number of cells migrating in the gapme points after scratching. (C) Transwell migration assay showingockdown inhibited CF migration. MEF2A-shRNA-transfected CFs wereHG in Transwell plates (0.8-m pore size) for 8 h. CFs on the exter-

    the Transwell were stained with crystal violet and photographedscope. (D) Quantication of the number of migrated CFs per eld ofplate. Data represent the mean SD from 5 separate elds in each

    independent experiments. NC: 5 mM glucose, HG: 30 mM glucose,: cells transfected with MEF2A-negative shRNA control before glu-n, sh-MEF2A+HG: cells transfected with MEF2A-shRNA before glucose: 5 mM d-glucose + 27.5 mM mannose. Data are the mean SD of 3xperiments; *P < 0.05 vs. NC; #P < 0.05 vs. HG. (For interpretation of

    to color in this gure legend, the reader is referred to the web version

    cells in the gap was monitored for the next 2 days atals.ll migration assay was performed using 24-well Tran-with 8-m porous polycarbonate membranes obtainedg. Serum-starved CFs (105) were added to the upperhereas the bottom chamber was lled with DMEM

    Fig. 4. Inhibitinto myobrotive analysis (of 3 independimages of immstained with DshRNA-NC+HGcose incubatioincubation. OCerences to colthis article.)

    containing with 4% foon the upprandomly scells were c

    2.9. Gelatin

    Cell cultacrylamidewashing buton X-100)(50 mM Tri37 C. Then250 solutioacid) and sLytic bandsMMP9 (83-

    2.10. Non-ifunction

    Blood pBP-98Acomues were dion of MEF2A attenuates differentiation of cardiac broblasts (CFs)blasts. (A) Representative western blot (upper panel) and quantita-lower panel) of -SMA expression in CFs. Data are the mean SDent experiments, *P < 0.05 vs. NC; #P < 0.05 vs. HG. (B) Representativeunouorescence staining for -SMA (red) in CFs. Nuclei were counter-API (blue). Scale bar = 25 m. NC: 5 mM glucose, HG: 30 mM glucose,: cells transfected with MEF2A-negative shRNA control before glu-n, sh-MEF2A+HG: cells transfected with MEF2A-shRNA before glucose: 5mMd-glucose + 27.5 mM mannose. (For interpretation of the ref-or in this gure legend, the reader is referred to the web version of

    HG or OC. After 12 h of incubation, the cells were xedrmaldehyde and stained with crystal violet. The cellser surface were removed with a cotton swab. Fiveelected elds were photographed and the migratedounted.

    zymography

    ure supernatants were electrophoresed in a 10% poly- gel containing1 mg/ml gelatin. The gel was washed inffer (50 mM TrisHCl [pH 7.5], 100 mM NaCl, 2.5% Tri-

    for 1 h and incubated overnight with enzyme buffers [pH 7.5], 150 mM NaCl, 5 mM CaCl2, 0.02% Brij-35) at, the gel was stained with Coomassie brilliant blue R-n (0.5% Coomassie Blue G-250, 30% methanol, 10% aceticubsequently destained in methanol/acetic acid/H2O.

    of gelatin digestion represented MMP2 (68-kDa) andkDa) activity.

    nvasive analysis of blood pressure and cardiac

    ressure was measured in conscious mice using theputerized tail-cuff system (Softron, Tokyo). Mean val-etermined from at least 3 measurements per mouse.

  • 56 X. Chen et al. / The International Journal of Biochemistry & Cell Biology 69 (2015) 5261

    Fig. 5. MEF2A plays an important role in HG-induced matrix metalloproteinase(MMP)tissue inhibitor of metalloproteinase (TIMP) interactions and collagenproduction in cardiac broblasts (CFs). (AC) Western blot and quantitative analysisof MMP2 and MMP9 protein expression. (DF) Western blot and quantitativeanalysis of TIMP1 and TIMP2 protein expression. (G) Activity of MMP2 and MMP9was studied by zymography by separating culture supernatants from FCs. (H and I)

    Left ventricular dimension and cardiac function were assessed byechocardiography (Vevo770 imaging system) before the mice werekilled. The mice were anesthetized with isourane. M-mode imagesat the level of the papillary muscles were obtained to measurethe interventricular septum (IVS) thickness, left ventricular end-diastolic dimension (LVEDD), LV end-systolic dimension (LVESD),LV posterior wall (LVPW), ejection fraction (EF), and fractionalshortening (FS). Pulsed-wave Doppler echocardiography was usedto measure the ratio of early to late mitral inow velocity (E/A).

    2.11. Histology and immunohistochemistry

    Massonas describechemistry, with primasecondary lowing the Data were a

    2.12. Statis

    The resudata are expANOVA waby Tukeys used for an

    3. Results

    3.1. HG inc

    CFs w(5.525 mMhigh as 2of MEF2A(P < 0.05), apressure (Fafter 6 h of(2.83-fold irescence anof CFs cultuMEF2A shotion of the expression These resul

    3.2. MEF2Aand is not a

    CF prolmyocardialon CF prolCCK-8 assaover 24 h dependent (Fig. 2A).

    Quantitative aquantitative aHG: 30 mM glcontrol beforeshRNA beforeare mean SDs trichrome and picrosirius red staining were performedd previously (Bauman et al., 2014). For immunohisto-heart tissue sections were incubated overnight at 4 Cry antibodies (collagen I and III, Abcam). Addition of theantibody and color development were conducted fol-manufacturers instructions (Jingmei, Shenzhen, China).nalyzed using Image-ProPlus6.0 (Media Cybernetics).

    tical analysis

    lts were from at least 3 independent experiments. Theressed as the mean standard deviation (SD). One-ways used to compare differences among groups, followedtest (2-tailed). SPSS v16.0 (SPSS Inc., Chicago, IL) wasalysis. P < 0.05 was considered statistically signicant.

    reases MEF2A expression and translocation in CFs

    ere exposed to various concentrations) of glucose for 72 h. A concentration as5 mM glucose signicantly increased the level

    as demonstrated by western blot analysisnd these effects were not due to changes in osmoticig. 1A). MEF2A protein expression in the CFs increased

    HG treatment (1.87-fold increase) and peaked at 48 hncrease) as compared with NC and OC. Immunouo-alysis revealed that MEF2A is expressed in the nucleired under NG (5.5 mM glucose) (Fig. 1B). Under HG,

    wed diffuse cytoplasmic staining, indicating transloca-protein. Further, HG increased only nuclear p-MEF2Aand did not affect p-MEF2A translocation (Fig. 1C).ts were conrmed by western blot analysis (Fig. 1D).

    inhibition decreases HG-induced proliferation of CFsssociated with apoptosis

    iferation and migration play an important role in brosis. To examine the effect of MEF2A silencingiferation, we investigated the proliferation of CFs byy. Under HG, the CFs showed a fast growth rateand proliferation signicantly increased in a time-manner when compared with NC and OC (P < 0.05)Inhibition of MEF2A by RNAi signicantly reduced

    nalysis of MMP2 and MMP9 activity. (J and K) Western blot andnalysis of collagen I and III protein expression. NC: 5 mM glucose,ucose, shRNA-NC+HG: cells transfected with MEF2A-negative shRNA

    glucose incubation, sh-MEF2A+HG: cells transfected with MEF2A- glucose incubation. OC: 5 mM d-glucose + 27.5 mM mannose. Data

    of 3 independent experiments; *P < 0.05 vs. NC; #P < 0.05 vs. HG.

  • X. Chen et al. / The International Journal of Biochemistry & Cell Biology 69 (2015) 5261 57

    HG-induced CF proliferation at all tested time points (efciency ofRNAi-mediated knockdown of MEF2A in the CFs is shown in Sup-plementary Fig. S2A2C). The proliferative effect of MEF2A wasconrmed by EdU staining. HG induced an increase in the pro-portion of the HG-ind(Fig. 2B andMEF2A inhE).

    Supplemonline versi

    3.3. MEF2A

    To evaluscratch andtime-depen(Fig. 3A andtion to thetranswell massay (Fig. 3

    3.4. MEF2Amyobrobla

    To clarifmyobroblExpression icantly incr(Fig. 4A). M-SMA expimmunou

    3.5. MEF2Acollagen syn

    To gain brosis, weCFs. We obsprotein expwas signicExpression between thshowed thaMMP2 and OC, and shRMMP2 andaltered MMsignicantlyment inhibi

    3.6. MEF2ATGF-1/Sma

    Cellular were enhanNC or OC tincreased preduced HG(Fig. 6BD)ERK1/2 andS3).

    Supplemonline versi

    ignal transduction mechanisms involved in MEF2A and high-glucose (HG)nt in cardiac broblasts (CFs). (AE) Western blot analysis of p-p38, p-Akt,

    p-Smad2, and p-Smad3 in CFs. HG activated the p38, Akt, and TGF-1/Smads in CFs. Treatment with MEF2A-shRNA further enhanced the activation ofand reversed the activation of the Akt (B) and TGF-1/Smad (CE) path-uced by HG. NC: 5 mM glucose, HG: 30 mM glucose, shRNA-NC+HG: cells

    ted with MEF2A-negative shRNA-control before glucose incubation, sh-HG: cells transfected with MEF2A-shRNA before glucose incubation. OC:-glucose + 27.5 mM mannose. Data are the mean SD of 3 independentents; *P < 0.05 vs. NC; #P < 0.05 vs. HG.EdU-positive CFs as compared with NC or OC, whileuced increase was inhibited after silencing of MEF2A

    C). Further, ow cytometric analysis indicated thatibition had no effect on CF apoptosis (Fig. 2D and

    entary Fig. S2 related to this article can be found, in theon, at http://dx.doi.org/10.1016/j.biocel.2015.10.012.

    inhibition limits HG-induced CF migration

    ate the inuence of MEF2A on CF migration, we used Transwell migration assays. HG induced prominentdent chemotaxis of CFs as compared with NC or OC

    B). MEF2A inhibition signicantly suppressed CF migra- level of the controls at all time points tested. Theigration assay results conrmed those of the scratchC and D).

    inhibition attenuates differentiation of CFs intosts

    y the role of MEF2A in the differentiation of CFs intoasts in vitro, we stimulated CFs with HG for 48 h.of -SMA (a myobroblast marker) in CFs was signif-eased by HG stimulation as compared with NC or OCEF2A inhibition signicantly suppressed the increase inression induced by HG. This effect was also observed byorescence analysis (Fig. 4B).

    mediates HG-induced imbalance of MMP-TIMP andthesis in CFs

    further insights into the potential roles of MEF2A in measured MMP expression and collagen synthesis inerved that HG induced an increase in MMP2 and MMP9ression in CFs as compared with NC or OC, whichantly attenuated by inhibition of MEF2A (Fig. 5AC).of TIMP1 and TIMP2 showed no signicant differencese groups (Fig. 5DF). Similarly, gelatin zymographyt HG treatment signicantly increased the activity ofMMP9 in culture supernatants as compared with NC orNA-MEF2A treatment markedly abrogated HG-induced

    MMP9 activity (P < 0.05) (Fig. 5GI). In line with theP activity, collagen I and III protein expression was

    increased in the HG group, while shRNA-MEF2A treat-ted the effect (P < 0.05) (Fig. 5J and K).

    inhibition alleviates HG-induced Akt andd signaling pathway activation in CFs

    levels of activated MAPKs, Akt, TGF-1, and Smad2/3ced at 24 h after HG stimulation as compared withreatment (P < 0.05) (Fig. 6). MEF2A inhibition furtherhosphorylation of p38 in CFs (P < 0.05) (Fig. 6A), while it-induced activation of Akt and TGF-1/Smad (P < 0.05). MEF2A silencing had no effect on the activation of

    JNK under HG treatment (P > 0.05) (Supplementary Fig.

    entary Fig. S3 related to this article can be found, in theon, at http://dx.doi.org/10.1016/j.biocel.2015.10.012.

    Fig. 6. StreatmeTGF-1,pathwayp38 (A) ways indtransfecMEF2A+5 mM dexperim

  • 58 X. Chen et al. / The International Journal of Biochemistry & Cell Biology 69 (2015) 5261

    Fig. 7. MEF2A inhibition prevents cardiac collagen deposition in diabetic mice. (Aand B) Western blot and mRNA analysis of MEF2A in normal and diabetic mousehearts. (C) Typical Masson trichrome (scale bars = 25 m) and picrosiriusredstaining of the heart in diabetic mice (scale bars = 25 m). (D) Quantitativeanalysis of the collagenous area in hearts of diabetic mice. (E) Typical examplesof collagen I and collagen III immunostaining of hearts of diabetic mice. (F and G)

    3.7. MEF2A inhibition attenuates diabetes-induced myocardialbrosis and cardiac dysfunction

    To ascertain the role of MEF2A in myocardial brosis and dys-function associated with diabetes, we investigated these disorders adiabetic mouse model after shRNA-mediated MEF2A in vivo knock-down. STZ induced rapid hyperglycemia in mice as compared withcitrate treatment beginning at 1 week after injection (data notshown). Blood glucose, systolic blood pressure, mean blood pres-sure, diastolic blood pressure, and heart rate were comparableamong the 4 groups at the end of the study. We observed an increasein blood gluin the diabmice did nogroup (P > 0ited lower dvehicle conwas found

    Hypergl(1.627 0.10.562 0.05compared downregula1.689 0.23as compareB and Supp

    Massoncollagen devs. 7.462 1gen depositvs. 30.67 chemical anof brotic mwhereas shdecreased ttreatment (

    Cardiac We observeLVEF, FS, anhigher in dIn diabetic dysfunctionnegative co

    4. Discussi

    This stuMEF2A andattenuated ation into mproductionactivation iheart improbetic mice. clarify the eof HG.

    We foundecreased a

    Quantitative aof mice. Conmice intramyoMEF2A: DM mmean SD of cose and blood pressure, and a decrease in heart rateetic mice (P < 0.05). Injection of shRNA-NC in diabetict change these parameters as compared with diabetic.05). However, mice injected with shRNA-MEF2A exhib-iastolic blood pressure and a higher heart rate than thetrol group (P < 0.05), while no effect on blood glucose(Table 1).ycemia signicantly increased MEF2A mRNA15-fold increase) and protein (0.835 0.042 vs.4, P < 0.05) expression in diabetic mice hearts aswith healthy controls. ShRNA-MEF2A treatmentted the myocardial MEF2A mRNA (0.301 0.027 vs.4) and protein (0.323 0.025vs. 1.008 0.093) levelsd with vehicle treatment in diabetic mice (Fig. 7A andlementary Fig. 2DF).s trichrome staining showed that diabetes increasedposition as compared with control mice (26.80 5.53%.16%, P < 0.05). ShRNA-MEF2A treatment reduced colla-ion as compared with vehicle treatment (15.12 3.50%4.20%, P < 0.05) (Fig. 7C and D). Similarly, immunohisto-alysis showed that diabetes enhanced the expressionarkers collagen I and III as compared with the control,RNA-MEF2A transfection of diabetic mice signicantlyhe levels of collagen I and III as compared with vehicleFig. 7EG).function was assessed at 24 weeks after STZ injection.d an increase in LVEDD and LVESD, and a decrease ind E/A in diabetic mice. IVS thickness and LVPWd wereiabetic than in control mice, although no signicantly.mice, shRNA-MEF2Atreatment prevented these cardiacs (LVEDD, LVESD, LVEF, FS and E/A) as compared withntrol shRNA (P < 0.05) (Table 2).

    on

    dy demonstrated that HG increased the expression of induced nuclear translocation in CFs. MEF2A inhibitionHG-induced CF proliferation, migration, and differenti-yobroblasts, and reduced MMP activity and collagen

    by blocking Akt and TGF-1/Smad signaling pathwayn CFs. Further, it was shown that MEF2A knockdown inved cardiac dysfunction and collagen deposition in dia-To the best of our knowledge, this is the rst report toffect of MEF2A on the function of CFs in the condition

    d that HG induced MEF2A expression peaked at 48 h butt 72 h in CFs. We hypothesize that higher expression of

    nalysis of the collagen I- and collagen III-stained areas in heartstrol: normal mice. DM: diabetic mellitus. DM+shRNA-NC: DMcardially injected withMEF2A-negative shRNA-control. DM+shRNA-ice intramyocardially injected with MEF2A-shRNA. Data are the

    3 independent experiments; *P < 0.05 vs. control; # P < 0.05 vs. DM.

  • X. Chen et al. / The International Journal of Biochemistry & Cell Biology 69 (2015) 5261 59

    Table 1Blood glucose, blood pressure, and heart rate measurements.

    Blood glucose (mM) SBP (mmHg) MBP (mmHg) DBP (mmHg) HR (pm)

    Control 7.35 0.68 119 7 86 7 69 8 598 16DM 33.89 0.97* 135 5* 102 7* 94 7* 545 22*DM+shRNA-NC 35.65 1.56* 138 5* 106 9* 96 9* 524 26*DM+shRNA-MEF2A 34.95 1.08 129 3 96 5 74 3# 594 15#

    The parameters were tested at 24 weeks after diabetes induction. SBP: systolic blood pressure; MBP: mean blood pressure; DBP: diastolic blood pressure; and HR:heart rate. Control: normal mice; DM: diabetic cardiomyopathy; DM+shRNA-NC: DM mice intramyocardially injected with shRNA-control. DM+shRNA-MEF2A: DM miceintramyocardially injected with MEF2A shRNA. All results are presented as the mean SEM. n = 10 per group.

    * P < 0.05 vs. control.# P < 0.05 vs. vehicle control.

    MEF2A is mainly because of increased phosphorylated p-MEF2Aexpression. P-MEF2A is located in the nucleus, and HG inducedtranslocation of MEF2A from the nucleus to the cytoplasm; thus, thep-MEF2A in the nucleus decreased at 72 h, resulting in the suddendecrease in MEF2A at 72 h.

    The results that HG induced MEF2A nuclear translocation wereconsistent with previous reports of increased MEF2A expressionin STZ-induaddition, Mshowed thaSTZ-inducefrom the nIn line witMEF2 to thedensity (Wacytoplasm activity of a(Link et al., 2(Xu et al., 2relocalizatiowhich MEF

    HG-indupathophysiremodelingtion reduceto curative function in

    The diffeincreased inby expressidisorganizeincreases swith increa2014; Yuendifferentiatour study. uated diffesuggested t

    remodeling by activating the differentiation of broblasts intomyobroblasts.

    MMPs play important roles in the maintenance and degradationof the ECM, thus contributing to the process of cardiac remodeling(Li et al., 2011). Studies have shown upregulation of MMP9 andMMP2 activity in diabetic heart (Li et al., 2011, 2012). Inhibition ofMMP activity has been suggested to have a cardioprotective role

    etes. activwas

    a lin ECMich fd othn acn. Wand d

    knosreg

    the actiontive rF2A ther,es-in

    actare antiatalsh, bser

    Akt, nd t/Smas a dmanied

    d pat Simi

    Table 2Echocardiogra

    IVS; d (mm)LVEDD (mmLVESD (mm)LVPWd (mmEF (%) FS (%) E/A

    IVS; d, intervediastolic postemice. DM: diawith shRNA-M

    * P < 0.05 vs# P < 0.05 vsced diabetic mouse heart (Feng et al., 2008, 2010). Inora et al. (Mora and Pessin, 2000; Mora et al., 2001)t MEF2A was decreased in heart nuclear extracts fromd diabetic rats, suggesting that MEF2A translocateducleus to the cytoplasm after HG treatment in CFs.h our observations, TGF--induced relocalization of

    cytoplasm was seen in myogenic cells grown to highng et al., 2011). Shuttling between the nucleus and theis a rapid and efcient mechanism for regulating the

    transcription factor, as has been observed for FOXO009; Zanella et al., 2008), NFAT (Wolff et al., 2010), p53008), etc. Therefore, we hypothesize that HG-inducedn of the MEF2A in the CFs might be a mechanism by

    2A regulates CF function under HG.ced CF proliferation and migration are the mainological mechanism underlying diabetes-induced ECM. In the present study, we showed that MEF2A inhibi-d CF proliferation and migration, which may contributetreatment targeting ECM remodeling and cardiac dys-diabetes.rentiation of broblasts into myobroblasts is strongly

    the myocardium of failing hearts and is characterizedon of -smooth muscle actin and increased formation ofd collagen matrix (Cucoranu et al., 2005). HG treatmentpontaneous differentiation of CFs to myobroblastssing passage when compared with LG (Shamhart et al.,

    et al., 2010). In line with these observations, enhancedion of myobroblasts was seen in HG-treated CFs inIn addition, we found that MEF2A inhibition atten-rentiation of CFs into myobroblasts. These resultshat MEF2A might play a role in diabetes-induced ECM

    in diabMMP9which strates

    TheIII, whrial anCollagefunctioin CFs MEF2Aand dyble fordysfunprotecfor ME

    Furdiabetthat HGwhich differeand Wthese oMAPK,we fouTGF-MEF2 iand hu2009; W/Sma2001).

    phic assessment.

    Control DM

    0.77 0.04 0.79 0.06 ) 3.28 0.15 3.83 0.13*

    2.02 0.14 2.88 0.26*

    ) 1.05 0.18 1.10 0.20

    71.99 4.45 55.33 2.58*40.96 3.29 31.83 1.96*1.45 0.12 0.96 0.08*

    ntricular septum thickness (diastole); LVEDD, left ventricular end-diastolic dimension; LVrior wall thickness; EF: ejection fraction, calculated. FS: fractional shortening, calculatebetic cardiomyopathy. DM+shRNA-NC: DM mice intramyocardially injected with shRNEF2A. n = 10, data are presented as the mean SD.. control.. DM. In the present study, we demonstrated that MMP2 andities were increased in response to HG in broblasts,

    reversed by MEF2A inhibition. This nding demon-k between MEF2A and MMPs in diabetes.

    mainly consists of molecules of collagen types I andorm brils and provide most of the connective mate-er structures in the myocardium (Pelouch et al., 1993).cumulation results in the development of heart dys-e found that HG promoted collagen I and III synthesisiabetic mouse hearts, which was partially inhibited byckdown. CF migration, proliferation, and differentiation,ulation of the MMPs were considered to be responsi-bnormal collagen deposition, which resulted in cardiac

    in diabetic mice. However, MEF2A inhibition played aole. Taken together, our ndings suggest a general rolein modulating ECM remodeling in the diabetic heart.

    we studied the molecular mechanism of MEF2A induced ECM remodeling. Previous studies have outlinedivates MAPK, Akt, and TGF-/Smad signaling pathways,ssociated with cell growth, proliferation, migration, andion (Al-Khalili et al., 2004; Sharma et al., 2012; Shiojima2002; Verrecchia and Mauviel, 2002). In agreement withvations, we demonstrated that HG activated the p38and TGF-/Smad signaling pathways in CFs. Meanwhile,hat MEF2A inhibition abated activation of the Akt andd signaling pathways. Previous studies have shown thatownstream effector of the PI3K/Akt pathway in neurons

    umbilical vein endothelial cells (HUVECs) (Sako et al.,mann et al., 2005), and a critical component of the TGF-hway in C2C12 myoblasts (Liu et al., 2004; Quinn et al.,larly, MEF2A might play a role in CFs via the Akt and

    DM+shRNA-NC DM+shRNA-MEF2A

    0.76 0.07 0.78 0.033.97 0.16* 3.20 0.122.71 0.18* 1.93 0.15

    1.15 0.14 1.08 0.15

    51.62 3.15* 64.73 4.6226.85 3.47* 37.46 2.870.98 0.12* 1.32 0.10#

    ESD: left ventricular end-systolic dimension; LVPW: left ventriculard; E/A: ratio of early to late mitral inow velocity. Control: normalA-control. DM+shRNA-MEF2A: DM mice intramyocardially injected

  • 60 X. Chen et al. / The International Journal of Biochemistry & Cell Biology 69 (2015) 5261

    TGF-/Smad signaling pathways. However, in our study, MEF2Ainhibition increased the phosphorylation of p38 in CFs, which maybe partially explained by the fact that MEF2A is a nuclear targetof the p38 MAPK signaling pathway (Chang et al., 2002; Ornatskyet al., 1999;mechanism

    In conclin CFs. MEFpartially referentiationsignaling pinhibition, betic cardiotypes involvand endoth

    Conict of

    The auth

    Acknowled

    The studgram of ChFoundationState PrograInnovative

    References

    Al-Khalili, L., Kcell differeenhanced Scand. 180

    Ban, C.R., Twigmechanism575596.

    Bauman, T.M.,et al., 2014glandular

    Black, B.L., Olsmyocyte e167196.

    Bugyei-TwumHigh glucoand contri

    Chang, C.I., Xuof MAP kinMEF2A an

    Cucoranu, I., C2005. NADdifferentia

    Dubey, R.K., Gadenosinerole of A2B

    Feng, B., Chen,cardiomyoPhysiol. En

    Feng, B., Chen,cardiomyo

    Ibrahim, R., Lelamentatphosphoryresistance

    Kim, M.S., KewAcute diabmitogen-aDiabetes 5

    Kim, Y., Phan, MEF2D tramice. J. Cli

    Kong, P., Chrisbrosis. Ce

    Konno, T., CheHeterogenpredicts foS. A. 107, 1

    Li, C.J., Lv, L., Li, H., Yu, D.M., 2012. Cardiac brosis and dysfunction in experimentaldiabetic cardiomyopathy are ameliorated by alpha-lipoic acid. Cardiovasc.Diabetol. 11, 73.

    Li, G., Li, Y., Liu, S., Shi, Y., Chi, Y., Liu, G., et al., 2013. Gremlin aggravateshyperglycemia-induced podocyte injury by a TGFbeta/smad dependent

    aling puangtrace010, J., Zhlpain

    1 dia, Oyarical

    ctive i228ang, J2-dep156y, T.All diviin, J.Dc helixU. S. A, Pessicle-spsporte, Yangrms asulin Jr., W.. Foca

    in the 68, 87Jr., W.-induyocyt

    y, O.I.,. Postein. N, V., Diacellu120.A.H., C., 2009rtrop2..A., Ya-mod

    29, 73 Fukuiopoiesphoinr 2. J.rt, P.E., 2014iac b

    S., Liu-142 bng adaang,resselation, I., Wogene, C.A.,

    Circ. R., Zhanotes

    ugh a. Cell. ia, F.,ugh thlation., Hashysis inMAPK., Zhanvastatits sigiol. 30.K., W

    igh-metic cann, M/Akt-d Wales et al., 2014), and might be a regulatory feedback.usion, the present study claried the role of MEF2A2A regulates HG-induced ECM remodeling, likely bygulating FC proliferation, migration, myobroblast dif-, and MMP activity via Akt and TGF-/Smad-dependentathways. Given the cardioprotective effects of MEF2AMEF2A may be a potential therapeutic target for dia-myopathy. However, the effects ofMEF2A in other celled in diabetic cardiomyopathy, such as cardiomyocyteselial cells, need to be further explored.

    interest

    ors declare no conict of interest.

    gements

    y is supported by the National 973 Basic Research Pro-ina (No. 2015CB553604), the National Natural Science

    of China (No. 91439201, 81170275, 81370412) and them of National Natural Science Foundation of China forResearch Group (No. 81321061).

    ramer, D., Wretenberg, P., Krook, A., 2004. Human skeletal musclentiation is associated with changes in myogenic markers andinsulin-mediated MAPK and PKB phosphorylation. Acta Physiol., 395403.g, S.M., 2008. Fibrosis in diabetes complications: pathogenics and circulating and urinary markers. Vasc. Health Risk Manag. 4,

    Nicholson, T.M., Abler, L.L., Eliceiri, K.W., Huang, W., Vezina, C.M.,. Characterization of brillar collagens and extracellular matrix ofbenign prostatic hyperplasia nodules. PLOS ONE 9, e109102.on, E.N., 1998. Transcriptional control of muscle development bynhancer factor-2 (MEF2) proteins. Annu. Rev. Cell Dev. Biol. 14,

    , A., Advani, A., Advani, S.L., Zhang, Y., Thai, K., Kelly, D.J., et al., 2014.se induces Smad activation via the transcriptional coregulator p300butes to cardiac brosis and hypertrophy. Cardiovas. Diabetol. 13, 89., B.E., Akella, R., Cobb, M.H., Goldsmith, E.J., 2002. Crystal structuresase p38 complexed to the docking sites on its nuclear substrated activator MKK3b. Mol. Cell 9, 12411249.lempus, R., Dikalova, A., Phelan, P.J., Ariyan, S., Dikalov, S., et al.,(P)H oxidase 4 mediates transforming growth factor-beta1-inducedtion of cardiac broblasts into myobroblasts. Circ. Res. 97, 900907.illespie, D.G., Mi, Z., Jackson, E.K., 1997. Exogenous and endogenous

    inhibits fetal calf serum-induced growth of rat cardiac broblasts: receptors. Circulation 96, 26562666.

    S., Chiu, J., George, B., Chakrabarti, S., 2008. Regulation ofcyte hypertrophy in diabetes at the transcriptional level. Am. J.docrinol. Metab. 294, E1119E1126.

    S., George, B., Feng, Q., Chakrabarti, S., 2010. miR133a regulatescyte hypertrophy in diabetes. Diabetes Metab. Res. Rev. 26, 4049.moine, A., Bertoglio, J., Raingeaud, J., 2015. Human enhancer ofion 1-induced colorectal cancer cell migration: role of serinelation and interaction with the breast cancer anti-estrogen

    3 protein. Int. J. Biochem. Cell Biol. 64, 4557.alramani, G., Puthanveetil, P., Lee, V., Kumar, U., An, D., et al., 2008a.etes moderates trafcking of cardiac lipoprotein lipase through p38ctivated protein kinase-dependent actin cytoskeleton organization.7, 6476.D., van Rooij, E., Wang, D.Z., McAnally, J., Qi, X., et al., 2008b. Thenscription factor mediates stress-dependent cardiac remodeling inn. Investig. 118, 124132.tia, P., Frangogiannis, N.G., 2014. The pathogenesis of cardiacll. Mol Life Sci. 71, 549574.n, D., Wang, L., Wakimoto, H., Teekakirikul, P., Nayor, M., et al., 2010.eous myocyte enhancer factor-2 (Mef2) activation in myocytescal scarring in hypertrophic cardiomyopathy. Proc. Natl. Acad. Sci. U.809718102.

    signLi, J.H., H

    in ex63, 2

    Li, Y., Maof catype

    Link, W.Chemsele2839

    Liu, D., KMEF1557

    McKinseof ce

    MolkentbasiSci.

    Mora, S.mustran

    Mora, S.isofoof in

    Nadruz 2005roleRes.

    Nadruz Loadby m

    Ornatsk1999prot

    Pelouchextr101

    Pereira, et alhypee847

    Quinn, Zas coRes.

    Sako, K.,Angphofacto

    Shamhaet alcard

    Sharma,miRduri

    She, T., Wsuppregu

    Shiojimaangi

    Souderscell.

    Tang, MpromthroMol

    Verrecchthroregu

    Wales, Sanalp38

    Wang, YAtorand Phys

    Wang, Wof hdiab

    WiedmaPI3Kathway. J. Cell. Biochem. 114, 21012113., X.R., Zhu, H.J., Johnson, R., Lan, H.Y., 2003. Role of TGF-beta signalingllular matrix production under high glucose conditions. Kidney Int.2019.u, H., Singh, M., Hill, D., Greer, P.A., et al., 2011. Targeted inhibitionreduces myocardial hypertrophy and brosis in mouse models ofbetes. Diabetes 60, 29852994.zabal, J., Serelde, B.G., Albarran, M.I., Rabal, O., Cebria, A., et al., 2009.interrogation of FOXO3a nuclear translocation identies potent andnhibitors of phosphoinositide 3-kinases. J. Biol. Chem. 284,400..S., Derynck, R., 2004. TGF-beta-activated Smad3 repressesendent transcription in myogenic differentiation. EMBO J. 23,6.., Zhang, C.L., Olson, E.N., 2002. MEF2: a calcium-dependent regulatorsion, differentiation and death. Trends Biochem. Sci. 27, 4047.., Olson, E.N., 1996. Combinatorial control of muscle development by-loop-helix and MADS-box transcription factors. Proc. Natl. Acad.. 93, 93669373.n, J.E., 2000. The MEF2A isoform is required for striatedecic expression of the insulin-responsive GLUT4 glucoser. J. Biol. Chem. 275, 1632316328., C., Ryder, J.W., Boeglin, D., Pessin, J.E., 2001. The MEF2A and MEF2Dre differentially regulated in muscle and adipose tissue during statesdeciency. Endocrinology 142, 19992004., Corat, M.A., Marin, T.M., Guimaraes Pereira, G.A., Franchini, K.G.,l adhesion kinase mediates MEF2 and c-Jun activation by stretch:

    activation of the cardiac hypertrophic genetic program. Cardiovasc.97., Kobarg, C.B., Constancio, S.S., Corat, P.D., Franchini, K.G., 2003.ced transcriptional activation of c-jun in rat myocardium: regulatione enhancer factor 2. Circ. Res. 92, 243251.

    Cox, D.M., Tangirala, P., Andreucci, J.J., Quinn, Z.A., Wrana, J.L., et al.,-translational control of the MEF2A transcriptional regulatoryucleic Acids Res. 27, 26462654.xon, I.M., Golfman, L., Beamish, R.E., Dhalla, N.S., 1993. Role oflar matrix proteins in heart function. Mol. Cell. Biochem. 129,

    lemente, C.F., Cardoso, A.C., Theizen, T.H., Rocco, S.A., Judice, C.C.,. MEF2C silencing attenuates load-induced left ventricularhy by modulating mTOR/S6K pathway in mice. PLoS ONE 4,

    ng, C.C., Wrana, J.L., McDermott, J.C., 2001. Smad proteins functionulators for MEF2 transcriptional regulatory proteins. Nucleic Acids2742.hara, S., Minami, T., Hamakubo, T., Song, H., Kodama, T., et al., 2009.tin-1 induces Kruppel-like factor 2 expression through aositide 3-kinase/AKT-dependent activation of myocyte enhancer

    Biol. Chem. 284, 55925601.., Luther, D.J., Adapala, R.K., Bryant, J.E., Petersen, K.A., Meszaros, J.G.,. Hyperglycemia enhances function and differentiation of adult ratroblasts. Can. J. Physiol. Pharmacol. 92, 598604., J., Wei, J., Yuan, H., Zhang, T., Bishopric, N.H., 2012. Repression ofy p300 and MAPK is required for survival signalling via gp130ptive hypertrophy. EMBO Mol. Med. 4, 617632.

    X., Gan, Y., Kuang, D., Yue, J., Ni, J., et al., 2012. Hyperglycemias cardiac stem cell homing to peri-infarcted myocardium via

    of ERK1/2 and p38 MAPK activities. Int. J. Mol. Med. 30, 13131320.alsh, K., 2002. Role of Akt signaling in vascular homeostasis andsis. Circ. Res. 90, 12431250.Bowers, S.L., Baudino, T.A., 2009. Cardiac broblast: the renaissancees. 105, 11641176.g, W., Lin, H., Jiang, H., Dai, H., Zhang, Y., 2007. High glucosethe production of collagen types I and III by cardiac broblasts

    pathway dependent on extracellular-signal-regulated kinase 1/2.Biochem. 301, 109114.

    Mauviel, A., 2002. Transforming growth factor-beta signalinge Smad pathway: role in extracellular matrix gene expression and. J. Investig. Dermatol. 118, 211215.emi, S., Blais, A., McDermott, J.C., 2014. Global MEF2 target gene

    cardiac and skeletal muscle reveals novel regulation of DUSP6 by-MEF2 signaling. Nucleic Acids Res. 42, 1134911362.g, M.X., Meng, X., Liu, F.Q., Yu, G.S., Zhang, C., et al., 2011.in suppresses LPS-induced rapid upregulation of toll-like receptor 4naling pathway in endothelial cells. Am. J. Physiol. Heart Circ.0, H1743H1752.ang, B., Lu, Q.H., Zhang, W., Qin, W.D., Liu, X.J., et al., 2014. Inhibition

    obility group box 1 improves myocardial brosis and dysfunction inrdiomyopathy. Int. J. Cardiol. 172, 202212.., Wang, X., Tang, X., Han, M., Li, M., Mao, Z., 2005.ependent regulation of the transcription factor myocyte enhancer

  • X. Chen et al. / The International Journal of Biochemistry & Cell Biology 69 (2015) 5261 61

    factor-2 in insulin-like growth factor-1- and membranedepolarization-mediated survival of cerebellar granule neurons. J. Neurosci.Res. 81, 226234.

    Wolff, M., Kredel, S., Haasen, D., Wiedenmann, J., Nienhaus, G.U., Kistler, B., et al.,2010. High content screening of CXCR2-dependent signalling pathways. Comb.Chem. High Throughput Screen. 13, 315.

    Xu, G.W., Mawji, I.A., Macrae, C.J., Koch, C.A., Datti, A., Wrana, J.L., et al., 2008. Ahigh-content chemical screen identies ellipticine as a modulator of p53nuclear localization. Apoptosis: Int. J. Prog. Cell Death 13, 413422.

    Xu, J., Gong, N.L., Bodi, I., Aronow, B.J., Backx, P.H., Molkentin, J.D., 2006. Myocyteenhancer factors 2A and 2C induce dilated cardiomyopathy in transgenic mice.J. Biol. Chem. 281, 91529162.

    Yuen, A., Laschinger, C., Talior, I., Lee, W., Chan, M., Birek, J., et al., 2010.Methylglyoxal-modied collagen promotes myobroblast differentiation.Matrix Biol.: J. Int. Soc. Matrix Biol. 29, 537548.

    Zanella, F., Rosado, A., Garcia, B., Carnero, A., Link, W., 2008. Chemical geneticanalysis of FOXO nuclear-cytoplasmic shuttling by using image-based cellscreening. Chembiochem: Eur. J. Chem. Biol. 9, 22292237.

    Inhibition of MEF2A prevents hyperglycemia-induced extracellular matrix accumulation by blocking Akt and TGF-1/Smad activ...1 Introduction2 Materials and methods2.1 Lentiviral vectors for RNA interference (RNAi)2.2 Animal model and RNAi2.3 Primary mouse CF culture and treatments2.4 Immunofluorescence2.5 Western blot analysis2.6 Quantitative RT-PCR2.7 Measurement of CF proliferation and apoptosis2.8 Cell migration assays2.9 Gelatin zymography2.10 Non-invasive analysis of blood pressure and cardiac function2.11 Histology and immunohistochemistry2.12 Statistical analysis

    3 Results3.1 HG increases MEF2A expression and translocation in CFs3.2 MEF2A inhibition decreases HG-induced proliferation of CFs and is not associated with apoptosis3.3 MEF2A inhibition limits HG-induced CF migration3.4 MEF2A inhibition attenuates differentiation of CFs into myofibroblasts3.5 MEF2A mediates HG-induced imbalance of MMP-TIMP and collagen synthesis in CFs3.6 MEF2A inhibition alleviates HG-induced Akt and TGF-1/Smad signaling pathway activation in CFs3.7 MEF2A inhibition attenuates diabetes-induced myocardial fibrosis and cardiac dysfunction

    4 DiscussionConflict of interestAcknowledgementsReferences