lkb1 deficiency in tie2-cre-expressing cells impairs ischemia

9
LKB1 Deficiency in Tie2-Cre-expressing Cells Impairs Ischemia-induced Angiogenesis * S Received for publication, March 15, 2010, and in revised form, May 18, 2010 Published, JBC Papers in Press, May 19, 2010, DOI 10.1074/jbc.M110.123794 Koji Ohashi ‡1 , Noriyuki Ouchi ‡2 , Akiko Higuchi , Reuben J. Shaw § , and Kenneth Walsh ‡3 From the Molecular Cardiology/Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts 02118 and the § Dulbecco Center for Cancer Research, Molecular and Cell Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037 LKB1 is a tumor suppressor protein whose loss leads to HIF1-mediated activation of a proangiogenic program in intestinal polyps. LKB1 is also protein kinase regulator of AMP- activated protein kinase (AMPK) signaling, which is essential for endothelial cell responses to tissue ischemia. To discern whether LKB1 signaling is either pro- or antiangiogenic, we investigated ischemia-induced revascularization in mice that were deficient for LKB1 in Tie2-Cre-expressing cells. Whereas homozygous deletion of LKB1 led to embryonic lethality, het- erozygous LKB1-knock-out (KO) (Lkb1 flox/ ;Tie2 Tg/ ) mice were viable. Unchallenged heterozygous LKB1-KO mice dis- played normal capillary density, but the revascularization of hind limb following ischemic surgery was significantly impaired as evaluated by laser Doppler flow and capillary density mea- surements. Reduction of LKB1 in cultured endothelial cells, using either small interfering RNA or an adenovirus expressing nonfunctional kinase-dead LKB1 protein, attenuated endothe- lial proliferation, migration, and differentiation into network structures on Matrigel that was accompanied by diminished AMPK phosphorylation at Thr-172. Conversely, adenovirus- mediated LKB1 overexpression (Ad-LKB1) augmented network structure formation, and this was associated with elevated AMPK phosphorylation. The augmented differentiation of endothelial cells into network structures induced by Ad-LKB1 was abrogated by the co-transduction of a dominant negative mutant of AMPK. These observations suggest that the LKB1- AMPK signaling axis in endothelial cells is a positive regulator of the revascularization response to tissue ischemia. LKB1, a serine-threonine protein kinase that is ubiquitously expressed in many organs, including liver, heart, and skeletal muscle (1, 2). Clinically, LKB1 deficiency leads to Peutz-Jeghers syndrome, a disease associated with gastrointestinal polyps and an increased risk of cancers (1, 3). Heterozygous ablation of LKB1 in mice leads to increased gastrointestinal polyps consis- tent with the human syndrome (4, 5). These LKB1-deficient pol- yps up-regulate the HIF1 transcription factor and its down- stream targets (6). Homozygous ablation of LKB1 results in lethality at embryonic day 11, which is associated with disorga- nized capillary network development and an up-regulation of vas- cular endothelial growth factor signaling (7). Collectively, these data indicate that LKB1 has an antiangiogenic function. Consis- tent with this hypothesis, Xie et al. (8) recently reported that over- expression of LKB1 in endothelial cells inhibits network formation on a Matrigel surface in vitro, a surrogate angiogenesis assay. On the other hand, it is conceivable that LKB1 has a proan- giogenic activity within endothelial cells based upon its ability to phosphorylate and activate AMP-activated protein kinase (AMPK). 4 In this regard, AMPK signaling promotes endothelial cell function and angiogenesis in response to hypoxic stress (9 –11), and tissue ischemia is a powerful activating stimulus for AMPK signaling (12). AMPK phosphorylates eNOS at residue Ser-1177 leading to the production of nitric oxide, a key deter- minant of endothelial cell function (9, 13–15). Thus, it is plausi- ble that LKB1 has a proangiogenic function via its ability to stim- ulate AMPK/eNOS signaling in endothelial cells under ischemic conditions. However, one cannot assume a priori that LKB1 is functionally similar to AMPK because it is also likely to have AMPK-independent functions. For example, LKB1 also targets downstream regulators that are distinct from AMPK, including 12 protein kinases (BRSK1, BRSK2, NUAK1, NUAK2, QIK, QSK, SIK, MARK1, MARK2, MARK3, MARK4, and SNRK) (16). Fur- thermore, in addition to LKB1, AMPK is regulated by other upstream protein kinases, including Ca 2 /calmodulin-dependent protein kinase kinase- (CaMKK) and transforming growth fac- tor--activated kinase 1 (TAK1) (17). In particular, studies in cul- tured endothelial cells have provided evidence for a functionally significant CaMKK-AMPK signaling interaction (18). Thus, the quantitative significance of the LKB1-AMPK signaling axis in endothelial cells is not clear, nor is it known whether LKB1 has a pro- or antiangiogenic function in this cell type. Recently, it has been reported that homozygous ablation of LKB1 in cells expressing Cre from the Tie1 promoter also * This work was supported, in whole or in part, by National Institutes of Health Grants AG15052, HL91949, and HL81587 (to K. W.). S The on-line version of this article (available at http://www.jbc.org) contains supplemental Tables 1 and 2 and Figs. 1–3. 1 Supported by the Manpei Suzuki Diabetes Foundation. 2 Supported by an American Heart Association Scientist Development grant, Northeast Affiliate. 3 To whom correspondence should be addressed: Dept. of Molecular Cardi- ology, Whitaker Cardiovascular Institute, Boston University School of Med- icine, 715 Albany St., W611, Boston, MA 02118. Tel.: 617-414-2390; Fax: 617-414-2391; E-mail: [email protected]. 4 The abbreviations used are: AMPK, AMP-activated protein kinase; eNOS, endothelial nitric-oxide synthase; KO, knock-out; Ad, adenovirus; kd, kinase-dead; dn, dominant negative; LDBF, laser Doppler blood flow; siRNA, small interfering RNA; HUVEC, human umbilical vein endothelial cell; TUNEL, terminal deoxynucleotidyl transferase-mediated dUTP nick- end labeling; ca, constitutively active; -gal, -galactosidase; m.o.i., multi- plicity of infection; ACC, acetyl-CoA carboxylase; HIF-1, hypoxia-inducible factor-1; MTS, 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphe- nyl)-2-(4-sulfophenyl)-2H-tetrazolium. THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 285, NO. 29, pp. 22291–22298, July 16, 2010 © 2010 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in the U.S.A. JULY 16, 2010 • VOLUME 285 • NUMBER 29 JOURNAL OF BIOLOGICAL CHEMISTRY 22291 by guest on March 18, 2018 http://www.jbc.org/ Downloaded from

Upload: hoangduong

Post on 11-Jan-2017

216 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: LKB1 Deficiency in Tie2-Cre-expressing Cells Impairs Ischemia

LKB1 Deficiency in Tie2-Cre-expressing Cells ImpairsIschemia-induced Angiogenesis*□S

Received for publication, March 15, 2010, and in revised form, May 18, 2010 Published, JBC Papers in Press, May 19, 2010, DOI 10.1074/jbc.M110.123794

Koji Ohashi‡1, Noriyuki Ouchi‡2, Akiko Higuchi‡, Reuben J. Shaw§, and Kenneth Walsh‡3

From the ‡Molecular Cardiology/Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts02118 and the §Dulbecco Center for Cancer Research, Molecular and Cell Biology Laboratory, The Salk Institute for BiologicalStudies, La Jolla, California 92037

LKB1 is a tumor suppressor protein whose loss leads toHIF1�-mediated activation of a proangiogenic program inintestinal polyps. LKB1 is also protein kinase regulator of AMP-activated protein kinase (AMPK) signaling, which is essentialfor endothelial cell responses to tissue ischemia. To discernwhether LKB1 signaling is either pro- or antiangiogenic, weinvestigated ischemia-induced revascularization in mice thatwere deficient for LKB1 in Tie2-Cre-expressing cells. Whereashomozygous deletion of LKB1 led to embryonic lethality, het-erozygous LKB1-knock-out (KO) (Lkb1flox/�;Tie2Tg/�) micewere viable. Unchallenged heterozygous LKB1-KO mice dis-played normal capillary density, but the revascularization ofhind limb following ischemic surgery was significantly impairedas evaluated by laser Doppler flow and capillary density mea-surements. Reduction of LKB1 in cultured endothelial cells,using either small interfering RNA or an adenovirus expressingnonfunctional kinase-dead LKB1 protein, attenuated endothe-lial proliferation, migration, and differentiation into networkstructures on Matrigel that was accompanied by diminishedAMPK phosphorylation at Thr-172. Conversely, adenovirus-mediated LKB1 overexpression (Ad-LKB1) augmented networkstructure formation, and this was associated with elevatedAMPK phosphorylation. The augmented differentiation ofendothelial cells into network structures induced by Ad-LKB1was abrogated by the co-transduction of a dominant negativemutant of AMPK. These observations suggest that the LKB1-AMPK signaling axis in endothelial cells is a positive regulatorof the revascularization response to tissue ischemia.

LKB1, a serine-threonine protein kinase that is ubiquitouslyexpressed in many organs, including liver, heart, and skeletalmuscle (1, 2). Clinically, LKB1 deficiency leads to Peutz-Jegherssyndrome, a disease associated with gastrointestinal polyps andan increased risk of cancers (1, 3). Heterozygous ablation ofLKB1 in mice leads to increased gastrointestinal polyps consis-

tent with the human syndrome (4, 5). These LKB1-deficient pol-yps up-regulate the HIF1� transcription factor and its down-stream targets (6). Homozygous ablation of LKB1 results inlethality at embryonic day 11, which is associated with disorga-nized capillary network development and an up-regulation of vas-cular endothelial growth factor signaling (7). Collectively, thesedata indicate that LKB1 has an antiangiogenic function. Consis-tent with this hypothesis, Xie et al. (8) recently reported that over-expressionofLKB1 inendothelial cells inhibitsnetwork formationon aMatrigel surface in vitro, a surrogate angiogenesis assay.On the other hand, it is conceivable that LKB1 has a proan-

giogenic activity within endothelial cells based upon its abilityto phosphorylate and activate AMP-activated protein kinase(AMPK).4 In this regard,AMPKsignaling promotes endothelialcell function and angiogenesis in response to hypoxic stress(9–11), and tissue ischemia is a powerful activating stimulus forAMPK signaling (12). AMPK phosphorylates eNOS at residueSer-1177 leading to the production of nitric oxide, a key deter-minant of endothelial cell function (9, 13–15). Thus, it is plausi-ble that LKB1 has a proangiogenic function via its ability to stim-ulate AMPK/eNOS signaling in endothelial cells under ischemicconditions. However, one cannot assume a priori that LKB1 isfunctionally similar to AMPK because it is also likely to haveAMPK-independent functions. For example, LKB1 also targetsdownstreamregulators that are distinct fromAMPK, including 12protein kinases (BRSK1, BRSK2, NUAK1, NUAK2, QIK, QSK,SIK, MARK1, MARK2, MARK3, MARK4, and SNRK) (16). Fur-thermore, in addition to LKB1, AMPK is regulated by otherupstreamprotein kinases, includingCa2�/calmodulin-dependentprotein kinase kinase-� (CaMKK�) and transforming growth fac-tor-�-activated kinase 1 (TAK1) (17). In particular, studies in cul-tured endothelial cells have provided evidence for a functionallysignificant CaMKK�-AMPK signaling interaction (18). Thus, thequantitative significance of the LKB1-AMPK signaling axis inendothelial cells is not clear, nor is it known whether LKB1 has apro- or antiangiogenic function in this cell type.Recently, it has been reported that homozygous ablation of

LKB1 in cells expressing Cre from the Tie1 promoter also* This work was supported, in whole or in part, by National Institutes of Health

Grants AG15052, HL91949, and HL81587 (to K. W.).□S The on-line version of this article (available at http://www.jbc.org) contains

supplemental Tables 1 and 2 and Figs. 1–3.1 Supported by the Manpei Suzuki Diabetes Foundation.2 Supported by an American Heart Association Scientist Development grant,

Northeast Affiliate.3 To whom correspondence should be addressed: Dept. of Molecular Cardi-

ology, Whitaker Cardiovascular Institute, Boston University School of Med-icine, 715 Albany St., W611, Boston, MA 02118. Tel.: 617-414-2390; Fax:617-414-2391; E-mail: [email protected].

4 The abbreviations used are: AMPK, AMP-activated protein kinase; eNOS,endothelial nitric-oxide synthase; KO, knock-out; Ad, adenovirus; kd,kinase-dead; dn, dominant negative; LDBF, laser Doppler blood flow;siRNA, small interfering RNA; HUVEC, human umbilical vein endothelialcell; TUNEL, terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling; ca, constitutively active; �-gal, �-galactosidase; m.o.i., multi-plicity of infection; ACC, acetyl-CoA carboxylase; HIF-1�, hypoxia-induciblefactor-1�; MTS, 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphe-nyl)-2-(4-sulfophenyl)-2H-tetrazolium.

THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 285, NO. 29, pp. 22291–22298, July 16, 2010© 2010 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in the U.S.A.

JULY 16, 2010 • VOLUME 285 • NUMBER 29 JOURNAL OF BIOLOGICAL CHEMISTRY 22291

by guest on March 18, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 2: LKB1 Deficiency in Tie2-Cre-expressing Cells Impairs Ischemia

results in embryonic lethality (19). Analysis of Lkb1flox/flox;Tie1Tg/� embryos revealed bloodless yolk sacs and pericardialswelling, and blood vessels within these embryos appear dilatedand congested with blood. Although these phenotypes showthat LKB1 is an important regulator of proper vascular devel-opment, nothing is known about the function of LKB1 in theadult vascular cells nor whether LKB1 has a pro- or antiangio-genic function in this context. To address these issues, we gen-erated a mouse model of partial LKB1 reduction by matingLkb1flox/flox mice with Tie2-Cre transgenic mice to produceheterozygous LKB1-KOmice. Analyses of thesemice revealed apartial reduction in LKB1 expression in vascular endothelialcells, and this was associated with impaired ischemia-inducedangiogenic responses. A series of in vitro studies further suggestthat endothelial cell LKB1 exerts proangiogenic activities via anAMPK-dependent pathway.

EXPERIMENTAL PROCEDURES

Materials—Mouse CD31 antibody was purchased from BDPharmingen. Antibodies LKB1 and eNOSwere purchased fromSanta Cruz Biotechnology. Antibodies phospho-AMPK (Thr-172) and phospho-eNOS (Ser-1177) were purchased from CellSignaling Technology (Beverly, MA). Phospho-ACC (Ser-79)antibody was purchased from Upstate Biotechnology. Tubu-lin antibody was purchased from Oncogene (Cambridge, MA).Antibody CD45 was purchased from R&D. Adenovirus vectorscontaining the gene for �-galactosidase (Ad-�-gal), the � sub-unit of LKB1 (Ad-LKB1), kinase-dead LKB1, which expressesthe Asp to Alamutation at position 194 in the kinase domain ofhuman LKB1 (Ad-kd-LKB1), constitutive active AMPK anddominant negative AMPK�2 (Ad-dn-AMPK) were describedpreviously (9, 20–22).Mouse Model of Angiogenesis—Lkb1flox/�;Tie2Tg/� mice and

littermate control mice (Lkb1flox/�;Tie2�/�) in the FVB/Nbackground were used for this study. The floxed LKB1 strainwas described previously (23). Mice expressing Cre recombi-nase under control of the Tie2 gene promoter (Tie2-Cre) werepurchased from Jackson Laboratory. Lkb1flox/�;Tie2Tg/� micewere interbred with Lkb1flox/flox;Tie2�/� littermates to gener-ate endothelial cell-specific LKB1 diminished mice (Lkb1flox/�;Tie2Tg/�) and littermate control mice (Lkb1flox/�;Tie2�/�).Study protocols were approved by the Institutional AnimalCare andUseCommittee at BostonUniversity.Mice at the agesof 8–11weeks were subjected to unilateral hind limb surgery toremove the left femoral artery and vein under anesthesia (10,24, 25). Prior to surgery, body weight and systolic blood pres-sure were determined using a tail cuff pressure analysis systemon conscious mice.Laser Doppler Blood Flow Analysis—Hind limb blood flow

was measured on postoperative days 0, 3, 7, 14, and 28 using alaser Doppler blood flow (LDBF) analyzer (Moor LDI; MoorInstruments). LDBF was assessed quantitatively as changes inthe laser image using different color pixels. To avoid mouse tomouse and experiment to experiment variations, hind limbblood flow was expressed as the ratio of left (ischemic) to right(nonischemic) LDBF signal.Capillary Density Analysis—Capillary density within the

thigh adductor muscle was quantified in histological sections.

Muscle samples were imbedded in OCT compound (Miles),snap frozen in liquid nitrogen, and cut into 5-�m slices. Capil-lary endothelial cells were identified by immunohistostainingfor CD31 (PECAM-1; BD Biosciences). Fifteen randomly cho-sen microscopic fields from three different sections in each tis-sue block were examined for the presence of capillary endothe-lial cells for each mouse specimen. Capillary density wasexpressed as the number of CD31-positive cells/muscle fiber.Mouse Lung Endothelial Cell Isolation—Mouse lung endo-

thelial cells were isolated from 8-week-old Lkb1flox/�;Tie2Tg/�heterozygous LKB1-KO and Lkb1flox/�;Tie2�/� control miceas described previously (25). Mice were killed by CO2 inhala-tion. The lungs were excised, minced, and digested with 0.1%collagenase in phosphate-buffered saline for 45min. The digestwas homogenized by multiple passages through a 14-gaugeneedle and then filtered through a 100-�m tissue sieve. The cellsuspension was isolated by immunoselection with CD31-con-jugated (BD Pharmingen) magnetic beads (Invitrogen). Whenplated cells reached confluence, a second immunoisolation wasperformed by ICAM-2-conjugated (BD Pharmingen) magneticbeads (Invitrogen).RNA Interference—The siRNAs targeting LKB1 were pur-

chased from Dharmacon (ON-TARGET plus SMART PoolHuman STK11, L-005035-00). Control cultures were trans-fected with unrelated siRNAs (Dharmacon, ON-TARGET plusControl Pool Nontargeting pool). HUVECs were transfectedfor 48 h with siRNAs by using Lipofectamine 2000 reagent(Invitrogen) according to the manufacturer’s instructions.Assessment of Endothelial Cell Responses—Migration activity

was measured under basal conditions in the absence of angio-genic-stimulatory growth factors using a modified Boydenchamber assay as described previously (15). Serum-deprivedHUVECs were resuspended in endothelial cell growth medi-um-2 with 0.5% fetal bovine serum. Cell suspension (250 �l,2.0 � 104 cells/well) were placed to the Transwell fibronectin-coated insert (6.4-mm diameter, 3.0-�m pore size, BD Bio-sciences). Migrated cells on the lower surface of the membranewere fixed and stained with Giemsa stain solution, and eightrandom microscopic fields/well were quantified. The forma-tion of network structures by HUVECs on growth factor-re-duced Matrigel (BD Biosciences) was performed as describedpreviously (26). Twenty four-well culture plates were coatedwith Matrigel according to the manufacturer’s instructions.Serum-deprived HUVECs were seeded on coated plates at 5 �104 cells/well in endothelial cell basementmedium-2 with 0.5%fetal bovine serum and incubated at 37 °C for 18 h. The degreeof cell network formation was quantified bymeasuring the net-work area in three randomly chosen fields from each well usingImageJ software. Each experiment was repeated three times.Cell Proliferation—Cell proliferation was assessed by direct

cell counting using a hemocytometer and the CellTiter 96AQueous kit (Promega) according to themanufacturer’s instruc-tions with MTS reagent. Briefly, in direct cell counting,HUVECs were plated at 10,000 cells/cm2 on 24-well plates andincubated in endothelial cell basal medium-2 with 2% fetalbovine serum. HUVECs were transfected with siRNAs accord-ing to the manufacturer’s instructions. After 48 h, cell numberwas assessed by a hemocytometer. In the assay using the Cell-

Role of Endothelial Cell LKB1 for Revascularization

22292 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 285 • NUMBER 29 • JULY 16, 2010

by guest on March 18, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 3: LKB1 Deficiency in Tie2-Cre-expressing Cells Impairs Ischemia

Titer 96 AQueous kit, HUVECs were plated at a density of 0.5 �104 cells/well in a 96-well plate and incubated in endothelial cellbasal medium-2 with 2% fetal bovine serum for 48 h. The490-nm absorbance was measured after 1-h incubations withMTS.Assessment of Endothelial Cell Apoptosis—Apoptosis was

assayed by a terminal deoxynucleotidyl transferase-mediateddUTPnick-end labeling (TUNEL) stainingmethodwith a com-mercial kit (Roche) (27). The mean number of apoptotic(TUNEL-positive) cells from three random fields (�100) ineach well was calculated.Western Blot Analysis—Tissue samples were homogenized

in lysis buffer containing 20mMTris-HCl (pH 8.0), 1%NonidetP-40, 150 mM NaCl, 0.5% deoxycholic acid, 1 mM sodiumorthovanadate, and a protease inhibitor mixture (Sigma).Immunoblot analysis was performed with antibodies at a1:1000 dilution, followed by incubation with a secondary anti-body conjugated with horseradish peroxidase at a 1:5000 dilu-tion. An ECL Plus Western blotting detection kit (AmershamBiosciences) was used.Statistical Methods—Data are presented as mean � S.E. Dif-

ferences between groups were evaluated by the Student’s t testor analysis of variance with Fisher’s protected least significantdifference test. A p value �0.05 denoted the presence of a sta-tistically significant difference. All calculations were performedby using StatView for Windows, version 5.0.

RESULTS

Decreased LKB1Expression in Endothelial Cells Isolated fromLkb1flox/�;Tie2Tg/� Mice—To investigate the role of LKB1 inangiogenesis in vivo, we produced conditional LKB1 knock-outmice using Cre-loxP system. Lkb1flox/flox mice were crossedwith Tie2-Cre transgenic (Tie2Tg/�) mice. An examination of226 F2 offspring revealed that Lkb1flox/flox;Tie2Tg/� (homozy-gous-KO) mice were not generated by this cross (sup-plemental Table 1). This result indicates that homozygous Lkb1ablation with Tie2-Cre leads to embryonic death, and this isconsistent with the findings of Londesborough et al. (19), whoutilized a Tie1-Cre mouse system to ablate this gene.LKB1 expression levels were examined in mouse lung endo-

thelial cells isolated from Lkb1flox/�;Tie2Tg/� (heterozygousLKB1-KO), Lkb1flox/�;Tie2�/� (control), and Lkb1flox/flox;Tie2�/� mice. LKB1 expression was decreased in endothelialcells isolated fromLkb1flox/�;Tie2Tg/�mice by�50%comparedwith those from Lkb1flox/�;Tie2�/� and Lkb1flox/flox;Tie2�/�

mice (Fig. 1). Correspondingly, Cre expression was detectedonly in mouse lung endothelial cells from heterozygousLKB1-KO but not control mice (Fig. 1). As expected from theexpression of Tie2-Cre in hematopoietic cells (28), LKB1expression was also reduced in spleen, but expression waslargely maintained in liver and kidney (supplemental Fig. 1).Impaired Ischemia-induced Angiogenesis in Heterozygous

LKB1-KOMice—To evaluate in vivo effects of LKB1 deficiencyin Tie2-Cre-expressing cells, hind limb ischemia surgery wasperformed on Lkb1flox/�;Tie2�/� (control) and heterozygousLKB1-KO mice. Prior to the operation, there were no signifi-cant differences in body weight, systolic blood pressure, heartrate, and fasting plasma glucose levels between these mice at 8

weeks of age (supplemental Table 2). Fig. 2A shows represen-tative laser Doppler images of hind limb blood flow before sur-gery and at different time points after surgery. Consistent withprior reports (10, 24, 25), ischemic hind limb blood flow perfu-sion increased to 80% of the nonischemic limb by day 28 incontrol mice (Fig. 2B). In contrast, blood flow in heterozygousLKB1-KOmice was significantly less than that in control mice,particularly at 14 and 28 days after surgery.To examine revascularization at the microcirculatory level,

capillary density was measured in nonischemic and ischemicmuscle by immunohistological staining of CD31. Fig. 2C showsrepresentative photos of adductor muscle tissue immuno-stained with CD31. Quantitative analysis revealed that the fre-quency of CD31-positive cells was greater in ischemic musclesthan in nonischemic muscles in control mice at day 28 afterischemic surgery, but the proportion of CD31-positive cells inischemic limbs was significantly less in heterozygous LKB1-KOmice compared with control mice (Fig. 2D). Staining with theleukocytemarker CD45 revealed�100-fold fewer positive cells(supplemental Fig. 2). The capillary density of nonischemicmuscle did not differ statistically between the two groups.These data show that a reduction of LKB1 expression results inimpaired revascularization in response to ischemia.To evaluate the regulation of candidate effectors down-

streamofLKB1intherevascularizationresponse, thephosphor-ylation level of eNOS at Ser-1177 was examined in ischemicskeletal muscles of heterozygous LKB1-KO and control mice.Quantitative Western blot analysis revealed that ischemic sur-gery significantly increased the phosphorylation levels of eNOSin ischemicmuscles of control mice, but this up-regulation was

FIGURE 1. Reduced LKB1 expression in endothelial cells isolated fromLkb1flox/�;Tie2Tg/� (heterozygous LKB1-KO) mice. Upper, Western blotanalysis for LKB1, Cre, and �-tubulin in endothelial cells isolated from lung ofLkb1flox/�;Tie2�/�, Lkb1flox/�;Tie2Tg/�, and Lkb1flox/flox;Tie2�/� mice. Lower,histogram indicating LKB1 expression reproducible in isolated lung endothe-lial cells from Lkb1flox/�;Tie2Tg/� mice (n � 6 in each group).

Role of Endothelial Cell LKB1 for Revascularization

JULY 16, 2010 • VOLUME 285 • NUMBER 29 JOURNAL OF BIOLOGICAL CHEMISTRY 22293

by guest on March 18, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 4: LKB1 Deficiency in Tie2-Cre-expressing Cells Impairs Ischemia

significantly abrogated in the muscle of heterozygousLKB1-KO mice on postoperative day 7 (Fig. 2, E and F).LKB1 Regulates Angiogenic Cell Responses in HUVECs—En-

dothelial cell differentiation into network structures on Matri-gel was employed to examine LKB1 signaling at a mechanistic

level. As shown in Fig. 3A, endo-thelial cells isolated from heterozy-gous LKB1-KO mice exhibitedlower levels of network formationthan those from Lkb1flox/�;Tie2�/�

and Lkb1flox/flox;Tie2�/�. To cor-roborate these observations, LKB1knockdown experiments were per-formed with HUVECs. Transduc-tion of HUVECs with siRNA target-ing LKB1 led to a dose-dependentreduction in LKB1 protein expres-sion (Fig. 3B). Accordingly, increas-ing levels of LKB1-siRNA led to adose-dependent reduction in theability of HUVECs to differentiateinto network structures onMatrigel(Fig. 3B). We also examined theconsequences of LKB1 loss of func-tion using an adenoviral vectorexpressing kinase-dead LKB1 (Ad-kd-LKB1). Treatment of HUVECswith Ad-kd-LKB1 at a m.o.i. of 50also abrogated polyphenol-inducedphosphorylation of AMPK by 80%(data not shown). Under thesetransduction conditions, Ad-kd-LKB1 significantly diminished theability of HUVECs to form cell net-works relative to HUVECs treatedwith a control vector (Fig. 3C). Con-versely, the gain of LKB1 function,via transduction of an adenoviralvector expressing the � subunit ofwild-type LKB1 (Ad-LKB1), aug-mented the morphogenesis ofHUVECs on Matrigel (Fig. 3D).Endothelial cell migration and

proliferation contribute to the an-giogenic response. Thus, the effectsof decreased LKB1 signaling onthese parameters were also investi-gated. Using a modified Boydenchamber method, we found thatsiRNA targeting LKB1 led to areduction in endothelial cell migra-tion (Fig. 3E). Similarly, treatmentwith siRNA targeting LKB1 signifi-cantly diminished endothelial cellproliferation compared with unre-lated siRNA treatment using anMTS-based assay and direct cellcounting (Fig. 3, F and G).

To test the prosurvival effect of LKB1, HUVECs weredeprived of serum in the presence of siRNA directed againstLKB1 or unrelated sequence. After 48 h, TUNEL stainingwas performed. Quantitative analysis revealed that treat-ment with LKB1 siRNA significantly increased the fraction

FIGURE 2. Impaired ischemia-induced revascularization in Lkb1flox/�;Tie2Tg/ � (heterozygous LKB1-KO)mice. A, representative LDBF images showing decreased perfusion in ischemic limbs of Lkb1flox/�;Tie2Tg/�

mice. A low perfusion signal (blue) was observed in the ischemic hind limbs of Lkb1flox/�;Tie2Tg/� mice, whereasa high perfusion signal (white to red) was detected in Lkb1flox/�;Tie2�/� on postoperative day 28. B, quantitativeanalysis of the ischemic/nonischemic LDBF ratio of Lkb1flox/�;Tie2�/� (open circles) and Lkb1flox/�;Tie2Tg/� (filledcircles) mice on postoperative days 0, 3, 7, 14, and 28 (n � 6). *, p � 0.05 for Lkb1flox/�;Tie2�/� mice. C, repre-sentative immunostaining of nonischemic and ischemic muscle tissues with anti-CD31 antibody (brown) onpostoperative day 28. D, quantitative analyses of capillary density in nonischemic and ischemic muscles ofLkb1flox/�;Tie2�/� and Lkb1flox/�;Tie2Tg/� mice on postoperative day 28 (n � 6 in each group). Capillary densitywas expressed as the number of capillaries/muscle fiber. E, Western blot analysis of phosphorylated eNOS atSer-1177 (P-eNOS), total eNOS (eNOS), and �-tubulin (Tubulin) in nonischemic and ischemic muscles ofLkb1flox/�;Tie2�/� and Lkb1flox/�;Tie2Tg/� mice on postoperative day 7. F, quantitative analysis of Western blots.*, p � 0.05 versus Lkb1flox/�;Tie2�/� (n � 4 in each group).

Role of Endothelial Cell LKB1 for Revascularization

22294 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 285 • NUMBER 29 • JULY 16, 2010

by guest on March 18, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 5: LKB1 Deficiency in Tie2-Cre-expressing Cells Impairs Ischemia

of TUNEL-positive cells compared with that of unrelatedsiRNA (Fig. 3H).LKB1 Regulates Angiogenic Cell Responses via an AMPK-de-

pendent Pathway—To investigate the signaling pathwaysdownstreamof LKB1 in endothelial cells,Western blot analyseswere performed on cells treated with siRNA targeting LKB1 oran unrelated siRNA. Knockdown of LKB1 by siRNA decreasedphosphorylation levels of AMPK at Thr-172 andACC at Ser-79under basal conditions (Fig. 4A).Whereas the ablation of LKB1by siRNA had little or no effect on eNOS phosphorylation atSer-1179 under basal conditions, the knockdown of LKB1diminished metformin-induced phosphorylation of eNOS inHUVECs (Fig. 4B). To examine the consequences of LKB1 gainof function, HUVECs were transduced with Ad-LKB1 leadingto a 2-fold overexpression of the �-subunit of LKB1 (Fig. 4C).Overexpression of LKB1 led to an increase in the phosphoryla-tion levels of ACC, AMPK, and eNOS (Fig. 4C).To investigate whether LKB1 overexpression promotes an in

vitro angiogenic response through the activation of AMPK,HUVECs were co-transduced with an adenovirus producing

dominant negative AMPK (Ad-dn-AMPK) or a control vectorexpressing �-galactosidase (Ad-�-gal). Treatment of HUVECswith Ad-LKB1 resulted in enhanced LKB1 expression andphosphorylation of ACC, a downstream kinase of AMPK (Fig.5A). Transduction with Ad-dn-AMPK abolished Ad-LKB1-stimulated phosphorylation of ACC (Fig. 5A). The increase innetwork formation induced by Ad-LKB1 was significantlyabrogated by co-transduction with Ad-dn-AMPK (Fig. 5A).Furthermore, the increase in endothelial cell proliferationinduced byAd-LKB1was significantly reduced by co-transduc-tion with Ad-dn-AMPK (supplemental Fig. 3).To explore the LKB1-AMPK signaling axis in endothelial

cells further, we tested whether the impairment in network for-mation in HUVECs treated with siRNA targeting LKB1 couldbe rescued by treatment with an adenovirus expressing consti-tutively activeAMPK (Ad-ca-AMPK). Ad-ca-AMPK treatmentincreased phosphorylation of ACC both in the presence andabsence of LKB1 (Fig. 5B). Whereas LKB1 knockdown dimin-ished network formation onMatrigel, transduction with Ad-ca-AMPK reversed the effect of LKB1 ablation (Fig. 5B).

FIGURE 3. Diminished LKB1 expression impairs endothelial cell functions. A, network formation assay in isolated mice lung endothelial cells from Lkb1flox/�;Tie2�/�, Lkb1flox/�;Tie2Tg/�, and Lkb1flox/flox;Tie2�/� mice is shown. *, p � 0.05 versus Lkb1flox/�;Tie2�/� (n � 6 in each group). B, treatment with siRNA targetingLKB1 dose dependently reduced LKB1 protein levels and abrogated network structures in HUVECs. Forty-eight hours after siRNA transfection, differentiationassays were performed (n � 3 in each group). C, network structures under 24-h treatment with adenovirus kinase-dead LKB1 (Ad-kd-LKB1) or adenovirusproducing �-galactosidase (Ad-�-gal) in HUVECs (50 m.o.i. each, n � 4 in each group). D, transduction with an adenovirus expressing the � subunit of LKB1promotes endothelial cell morphogenesis relative to control HUVEC cultures transduced with Ad-�-gal. E–H, treatment with siRNA targeting LKB1 decreasedmigration (E), proliferation (F and G), and increased apoptosis (H) in HUVECs. Forty-eight hours after siRNA transfection, migration and proliferation assays wereperformed using a modified Boyden chamber method and MTS-based assay, respectively (n � 4 in each group at migration assay, n � 8 in each group atproliferation assay). In direct cell counting, HUVECs were plated at 10,000 cells/cm2 on 24-well plates and incubated in endothelial cell basal medium-2 with 2%fetal bovine serum for 48 h after siRNA transfection. Cell number was assessed by a hemocytometer (n � 4 in each group). Apoptosis was assayed by TUNELstaining method. The mean number of apoptotic (TUNEL-positive) cells from three random fields (�100) in each well was calculated (n � 4 in each group).

Role of Endothelial Cell LKB1 for Revascularization

JULY 16, 2010 • VOLUME 285 • NUMBER 29 JOURNAL OF BIOLOGICAL CHEMISTRY 22295

by guest on March 18, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 6: LKB1 Deficiency in Tie2-Cre-expressing Cells Impairs Ischemia

Transduction of Ad-ca-AMPK alone also increased networkformation in this assay.

DISCUSSION

LKB1 is a tumor suppressor (29–31), and enhanced expres-sion of LKB1 in cancer cells results in attenuated tumor growth,invasion, and metastasis, at least in part, through its ability toinhibit tumor angiogenesis (32). Consistent with these observa-tions, whole body, homozygous LKB1 ablation results in anembryonic lethal phenotype that is associated with increasedvascular endothelial growth factor expression and abnormalcapillary development (7). Based on these observations, LKB1has been viewed as an antiangiogenic factor. However, AMPK,a downstream effector of LKB1, functions in endothelial cells

to promote angiogenic responsesunder hypoxic conditions (9). Thus,it is conceivable that the angiogen-ic-regulatory properties of LKB1are cell type-specific, resulting indifferent regulatory responses invascular versus parenchymal cells.Because nothing is known about

the function of endothelial cell LKB1in the postnatal revascularizationresponse to tissue ischemia, we ana-lyzed the consequences of acute hindlimb ischemia in adultmice that weredeficient for LKB1 expression inTie2-Cre-expressing cells. In thepresent study, no viable pups wereproduced from matings intended toproduce the homozygous ablation ofthe LKB1 allele with Tie2-Cre. Thesefindings are consistent with therecent report of embryonic lethalitywhen a different transgenic Cre-ex-pressing line (Tie1-Cre) was used toablate the floxed LKB1 allele (19).Consequently, we utilized het-erozygous LKB1-KO mice forthe hind limb ischemia study.Although Lkb1flox/�;Tie2Tg/� miceexhibit approximately half thelevel of LKB1 protein in lungendothelial cells, their vascularnetwork is indistinguishable fromcontrol mice under nonstress con-ditions. However, this reductionin LKB1 expression in Tie2-Cre-expressing cells led to an impairedrevascularization response to tis-sue ischemia that was associatedwith reductions in ischemia-in-duced eNOS phosphorylation inthe tissue. The diminished tis-sue reperfusion in heterozygousLKB1-KO mice was consistentwith anatomic evidence of a

reduction in capillary density in the limbs that were recov-ering from ischemia. Collectively, these results show that apartial reduction of LKB1 signaling in vascular endothelialcells impairs the revascularization response to ischemic con-ditions in adult mice.Although many studies have used the Tie2-Cre model for

gene ablation in vascular endothelial cells, it is documented thatthese Tie2-Cre transgenic mice also express Cre in hematopoi-etic cells (28). Because hematopoietic cells are important for arevascularization response (33, 34), we analyzed LKB1 expres-sion levels in spleen, which contains many hematopoietic cells.LKB1 expression in spleen was reduced in the heterozygousLKB1-KO mice compared with control mice. Therefore, LKB1signaling in hematopoietic derived cells, such as macrophages

FIGURE 4. Transduction with siRNA targeting LKB1 diminished an AMPK signaling in HUVECs. A, LKB1 isrequired for full AMPK signaling. Western blot analysis for LKB1, phosphorylated ACC at Ser-79 (pACC),phosphorylated AMPK at Thr-172 (pAMPK), total AMPK (AMPK), and �-tubulin (Tubulin) is shown. Rightpanel shows quantification of LKB1, pAMPK, and pACC. *, p � 0.05 versus unrelated siRNA (n � 3 in eachgroup). HUVECs were treated with siRNA targeting LKB1 for 48 h. B, LKB1 is required for metformin-stimulated eNOS phosphorylation. Western blot analysis for LKB1, phosphorylated ACC, phosphorylatedeNOS (P-eNOS), and �-tubulin is shown. HUVECs were treated with siRNA targeting LKB1 for 48 h followedby treatment with metformin (2 mM) or vehicle for 3 h. A representative Western immunoblot is shown.C, LKB1 overexpression activates AMPK signaling and leads to an increase in eNOS phosphorylation.Western blot analysis for LKB1, phosphorylated-ACC at Ser-79, phosphorylated-AMPK at Thr-172, totalAMPK, phosphorylated-eNOS at Ser-1177, total eNOS, and �-tubulin is shown. HUVECs were infected withAd-�-gal or Ad-LKB1 for 24 h. Right panels show quantitative Western blot analyses for P-AMPK andP-eNOS. **, p � 0.01 versus Ad-�-gal treatment (n � 3 in each group).

Role of Endothelial Cell LKB1 for Revascularization

22296 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 285 • NUMBER 29 • JULY 16, 2010

by guest on March 18, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 7: LKB1 Deficiency in Tie2-Cre-expressing Cells Impairs Ischemia

or vascular progenitor cells, may also contribute to thisimpaired vascularization phenotype.The in vivo findings in the murine ischemic hind limbmodel

were corroborated by a series of in vitro experiments that exam-ined the behavior of cultured endothelial cells. Reductions inLKB1 expression in HUVECs using siRNA ablation led toreductions in the ability of these cells to proliferate, migrate,and differentiate into network structures on Matrigel. LKB1ablation also exacerbated endothelial cell apoptosis under condi-tions of serum deprivation. Similarly, LKB1 inactivation using anadenoviral vector expressing kinase-dead LKB1 (Ad-kd-LKB1)

diminished the ability of HUVECs toform cell networks, whereas adenovi-rus-mediated overexpression of the�subunit of LKB1 promoted this mor-phogenic response. Endothelial cellsisolated from lungs of heterozygousLKB1-KO mice also displayed im-paired network formation in themorphogenesis.The cell culture experiments per-

formed in this study indicate thatthe proangiogenic activity of LKB1is mediated by its ability to regulateAMPK through the activating phos-phorylation of Thr-172. These find-ings are consistent with other stud-ies in cultured vascular endothelialcells showing that LKB1 functionsto regulate AMPK. For example,AICAR, but not thrombin, activatesAMPK in an LKB1-dependent path-way in HUVECs (18). Other studieshave shown that reactive nitrogenspecies activate AMPK in endo-thelial cells through an LKB1-dependent manner (35). It has alsobeen shown that LKB1 is a regulatorof AMPK signaling in nonvasculartissues. Liver-specific LKB1 defi-ciency causes hyperglycemia associ-ated with reduced hepatic AMPKactivity and an increase in hepaticgluconeogenesis (23). Liver-specificLKB1 deficiency results in impairedresponses to metformin, an antidi-abetic drug that activates AMPK(23). Skeletal muscle-specific LKB1deficiency also leads to reducedAMPK phosphorylation at basaland AICAR- or phenformin-stimu-lated conditions (36, 37). Finally,ablation of LKB1 in the heart leadsto hypertrophy that is associatedwith a reduction in AMPK signaling(38).It is well established that Akt sig-

naling functions downstream ofgrowth factors in endothelial cells to promote angiogenesis(39). Whereas hypoxia will activate AMPK signaling in endo-thelial cells (40), these conditions will lead to a reduction in Aktsignaling (9, 41). Thus, we speculate that hypoxia-inducedAMPK activation serves to promote a vascularization responseunder conditions of stress and nutritional deprivation whenAkt signaling is down-regulated. This proposed revasculariza-tion function of AMPK is consistent with thewidely recognizedcytoprotective role of this kinase in fuel preservation while thetissue recovers from the ischemic injury. The data reportedhere support the notion that the LKB1-AMPK signaling axis

FIGURE 5. LKB1 overexpression enhances endothelial cell morphogenesis in an AMPK-dependent man-ner in HUVECs. Twenty-four hours after infection with adenoviral constructs encoding �-galactosidase (Ad-�-gal), LKB1 (Ad-LKB1) (100 m.o.i.), network formation assays were performed in HUVECs. A, left panels showrepresentative photographs of endothelial cell differentiation into network structures. Right upper panels showrepresentative Western blots for phosphorylated-ACC (Ser-79) and LKB1. Right lower panel shows quantitativeanalysis of network area under treatment with adenovirus producing dominant negative AMPK (Ad-dn-AMPK)(10 m.o.i.) for 24 h. n � 4 in each group. B, left panels show representative photographs of endothelial celldifferentiation into network structures. Right upper panels show representative Western blots for phosphory-lated-ACC (Ser-79) and LKB1. Right lower panel shows quantitative analysis of network area with 24-h stimula-tion by adenovirus producing constitutive active AMPK (Ad-ca-AMPK) (50 m.o.i.) under 48-h treatment withsiRNA targeting LKB1 (1 nM) (n � 4 in each group).

Role of Endothelial Cell LKB1 for Revascularization

JULY 16, 2010 • VOLUME 285 • NUMBER 29 JOURNAL OF BIOLOGICAL CHEMISTRY 22297

by guest on March 18, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 8: LKB1 Deficiency in Tie2-Cre-expressing Cells Impairs Ischemia

promotes proangiogenic responses in cultured endothelialcells, including proliferation, migration, survival, eNOS activa-tion and differentiation into network structures. Furthermore,its expression in Tie2-Cre-expressing cells is essential for arobust revascularization response to ischemia in vivo. In con-trast to these findings, another group recently reported thatadenovirus-mediated overexpression of LKB1 inhibits the for-mation of networks by HUVECs on Matrigel (8). The reasonsfor these disparate findings are not clear, but they indicate theneed for further in vitro and in vivo investigations to appreciatemore fully the role of endothelial cell LKB1 in angiogenesis.In summary, we demonstrated for the first time that the het-

erozygous deficiency of LKB1 expression in Tie2-Cre-express-ing cells leads to an impaired revascularization response to tis-sue ischemia in vivo. These results were corroborated by aseries of in vitro studies showing that reductions in LKB1impair endothelial cell morphogenesis, whereas the overex-pression of LKB1 stimulates this process. These data indicatethat LKB1 plays a role in stress-induced tissue revascularizationthrough its ability to regulate AMPK signaling in postnatal vas-cular endothelial cells.

REFERENCES1. Hemminki, A.,Markie, D., Tomlinson, I., Avizienyte, E., Roth, S., Loukola,

A., Bignell, G.,Warren,W., Aminoff, M., Hoglund, P., Jarvinen, H., Kristo,P., Pelin, K., Ridanpaa, M., Salovaara, R., Toro, T., Bodmer, W., Ol-schwang, S., Olsen, A. S., Stratton, M. R., de la Chapelle, A., and Aaltonen,L. A. (1998) Nature 391, 184–187

2. Jenne, D. E., Reimann, H., Nezu, J., Friedel,W., Loff, S., Jeschke, R.,Muller,O., Back, W., and Zimmer, M. (1998) Nat. Genet. 18, 38–43

3. Hemminki, A., Tomlinson, I., Markie, D., Jarvinen, H., Sistonen, P.,Bjorkqvist, A.M., Knuutila, S., Salovaara, R., Bodmer,W., Shibata, D., de laChapelle, A., and Aaltonen, L. A. (1997) Nat. Genet. 15, 87–90

4. Bardeesy, N., Sinha, M., Hezel, A. F., Signoretti, S., Hathaway, N. A., Shar-pless, N. E., Loda, M., Carrasco, D. R., and DePinho, R. A. (2002) Nature419, 162–167

5. Miyoshi, H., Nakau, M., Ishikawa, T. O., Seldin, M. F., Oshima, M., andTaketo, M. M. (2002) Cancer Res. 62, 2261–2266

6. Shackelford, D. B., Vasquez, D. S., Corbeil, J., Wu, S., Leblanc, M., Wu,C. L., Vera, D. R., and Shaw, R. J. (2009) Proc. Natl. Acad. Sci. U.S.A. 106,11137–11142

7. Ylikorkala, A., Rossi, D. J., Korsisaari, N., Luukko, K., Alitalo, K., Henke-meyer, M., and Makela, T. P. (2001) Science 293, 1323–1326

8. Xie, Z., Dong, Y., Zhang, J., Scholz, R., Neumann,D., andZou,M.H. (2009)Mol. Cell. Biol. 29, 3582–3596

9. Nagata, D., Mogi, M., and Walsh, K. (2003) J. Biol. Chem. 278,31000–31006

10. Shibata, R., Ouchi, N., Kihara, S., Sato, K., Funahashi, T., and Walsh, K.(2004) J. Biol. Chem. 279, 28670–28674

11. Ouchi, N., Shibata, R., and Walsh, K. (2005) Circ. Res. 96, 838–84612. Kudo, N., Barr, A. J., Barr, R. L., Desai, S., and Lopaschuk, G. D. (1995)

J. Biol. Chem. 270, 17513–1752013. Chen, Z. P., Mitchelhill, K. I., Michell, B. J., Stapleton, D., Rodriguez-

Crespo, I., Witters, L. A., Power, D. A., Ortiz de Montellano, P. R., andKemp, B. E. (1999) FEBS Lett. 443, 285–289

14. Zhang, Y., Lee, T. S., Kolb, E.M., Sun, K., Lu, X., Sladek, F.M., Kassab,G. S.,Garland, T., Jr., and Shyy, J. Y. (2006) Arterioscler. Thromb. Vasc. Biol. 26,

1281–128715. Ouchi, N., Kobayashi, H., Kihara, S., Kumada, M., Sato, K., Inoue, T.,

Funahashi, T., and Walsh, K. (2004) J. Biol. Chem. 279, 1304–130916. Lizcano, J.M., Goransson,O., Toth, R., Deak,M.,Morrice,N.A., Boudeau,

J., Hawley, S. A., Udd, L., Makela, T. P., Hardie, D. G., and Alessi, D. R.(2004) EMBO J. 23, 833–843

17. Witczak, C. A., Sharoff, C. G., and Goodyear, L. J. (2008)Cell Mol. Life Sci.65, 3737–3755

18. Stahmann, N.,Woods, A., Carling, D., andHeller, R. (2006)Mol. Cell. Biol.26, 5933–5945

19. Londesborough, A., Vaahtomeri, K., Tiainen, M., Katajisto, P., Ekman, N.,Vallenius, T., and Makela, T. P. (2008) Development 135, 2331–2338

20. Fujio, Y., and Walsh, K. (1999) J. Biol. Chem. 274, 16349–1635421. Noga, A. A., Soltys, C. L., Barr, A. J., Kovacic, S., Lopaschuk, G. D., and

Dyck, J. R. (2007) Am. J. Physiol. Heart Circ. Physiol. 292, H1460–H146922. Zhou, J., Huang,W., Tao, R., Ibaragi, S., Lan, F., Ido, Y.,Wu, X., Alekseyev,

Y. O., Lenburg, M. E., Hu, G. F., and Luo, Z. (2009) Oncogene 28,1993–2002

23. Shaw, R. J., Lamia, K. A., Vasquez, D., Koo, S. H., Bardeesy, N., Depinho,R. A., Montminy, M., and Cantley, L. C. (2005) Science 310, 1642–1646

24. Galasso, G., Schiekofer, S., Sato, K., Shibata, R., Handy, D. E., Ouchi, N.,Leopold, J. A., Loscalzo, J., and Walsh, K. (2006) Circ. Res. 98, 254–261

25. Ohashi, K., Ouchi, N., Sato, K., Higuchi, A., Ishikawa, T. O., Herschman,H. R., Kihara, S., and Walsh, K. (2009)Mol. Cell. Biol. 29, 3487–3499

26. Skurk, C., Maatz, H., Rocnik, E., Bialik, A., Force, T., andWalsh, K. (2005)Circ. Res. 96, 308–318

27. Shibata, R., Sato, K., Pimentel, D. R., Takemura, Y., Kihara, S., Ohashi, K.,Funahashi, T., Ouchi, N., andWalsh, K. (2005) Nat. Med. 11, 1096–1103

28. Alva, J. A., Zovein, A. C., Monvoisin, A., Murphy, T., Salazar, A., Harvey,N. L., Carmeliet, P., and Iruela-Arispe, M. L. (2006) Dev. Dyn. 235,759–767

29. Tiainen,M., Vaahtomeri, K., Ylikorkala, A., andMakela, T. P. (2002)Hum.Mol. Genet. 11, 1497–1504

30. Tiainen, M., Ylikorkala, A., and Makela, T. P. (1999) Proc. Natl. Acad. Sci.U.S.A. 96, 9248–9251

31. Karuman, P., Gozani, O., Odze, R. D., Zhou, X. C., Zhu,H., Shaw, R., Brien,T. P., Bozzuto, C. D., Ooi, D., Cantley, L. C., and Yuan, J. (2001)Mol. Cell7, 1307–1319

32. Zhuang, Z. G., Di, G. H., Shen, Z. Z., Ding, J., and Shao, Z. M. (2006)Mol.Cancer Res. 4, 843–849

33. Arras, M., Ito, W. D., Scholz, D., Winkler, B., Schaper, J., and Schaper, W.(1998) J. Clin. Invest. 101, 40–50

34. Stabile, E., Kinnaird, T., la Sala, A., Hanson, S. K.,Watkins, C., Campia, U.,Shou, M., Zbinden, S., Fuchs, S., Kornfeld, H., Epstein, S. E., and Burnett,M. S. (2006) Circulation 113, 118–124

35. Xie, Z., Dong, Y., Zhang, M., Cui, M. Z., Cohen, R. A., Riek, U., Neumann,D., Schlattner, U., and Zou, M. H. (2006) J. Biol. Chem. 281, 6366–6375

36. Sakamoto, K., McCarthy, A., Smith, D., Green, K. A., GrahameHardie, D.,Ashworth, A., and Alessi, D. R. (2005) EMBO J. 24, 1810–1820

37. Koh, H. J., Arnolds, D. E., Fujii, N., Tran, T. T., Rogers, M. J., Jessen, N., Li,Y., Liew, C. W., Ho, R. C., Hirshman, M. F., Kulkarni, R. N., Kahn, C. R.,and Goodyear, L. J. (2006)Mol. Cell. Biol. 26, 8217–8227

38. Ikeda, Y., Sato, K., Pimentel, D. R., Sam, F., Shaw, R. J., Dyck, J. R., andWalsh, K. (2009) J. Biol. Chem. 284, 35839–35849

39. Shiojima, I., and Walsh, K. (2002) Circ. Res. 90, 1243–125040. Zou, M. H., Hou, X. Y., Shi, C. M., Kirkpatick, S., Liu, F., Goldman, M. H.,

and Cohen, R. A. (2003) J. Biol. Chem. 278, 34003–3401041. Zou, M. H., Hou, X. Y., Shi, C. M., Nagata, D., Walsh, K., and Cohen, R. A.

(2002) J. Biol. Chem. 277, 32552–32557

Role of Endothelial Cell LKB1 for Revascularization

22298 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 285 • NUMBER 29 • JULY 16, 2010

by guest on March 18, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 9: LKB1 Deficiency in Tie2-Cre-expressing Cells Impairs Ischemia

Koji Ohashi, Noriyuki Ouchi, Akiko Higuchi, Reuben J. Shaw and Kenneth WalshAngiogenesis

LKB1 Deficiency in Tie2-Cre-expressing Cells Impairs Ischemia-induced

doi: 10.1074/jbc.M110.123794 originally published online May 19, 20102010, 285:22291-22298.J. Biol. Chem. 

  10.1074/jbc.M110.123794Access the most updated version of this article at doi:

 Alerts:

  When a correction for this article is posted• 

When this article is cited• 

to choose from all of JBC's e-mail alertsClick here

Supplemental material:

  http://www.jbc.org/content/suppl/2010/05/19/M110.123794.DC1

  http://www.jbc.org/content/285/29/22291.full.html#ref-list-1

This article cites 41 references, 28 of which can be accessed free at

by guest on March 18, 2018

http://ww

w.jbc.org/

Dow

nloaded from