+ All Categories
Home > Documents > 5-Aminoimidazole-4-carboxamide-1- D-ribofuranoside ... · pathway and increased expression of cell...

5-Aminoimidazole-4-carboxamide-1- D-ribofuranoside ... · pathway and increased expression of cell...

Date post: 24-Oct-2019
Category:
Upload: others
View: 1 times
Download: 0 times
Share this document with a friend
13
5-Aminoimidazole-4-carboxamide-1--D-ribofuranoside Inhibits Cancer Cell Proliferation in Vitro and in Vivo via AMP-activated Protein Kinase * Received for publication, July 8, 2005, and in revised form, September 21, 2005 Published, JBC Papers in Press, September 21, 2005, DOI 10.1074/jbc.M507443200 Ramandeep Rattan , Shailendra Giri , Avtar K. Singh § , and Inderjit Singh ‡1 From the Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina 29425 and the § Department of Pathology and Laboratory Medicine, Ralph Johnson Veterans Affairs Medical Center, Charleston, South Carolina 29425 5-Aminoimidazole-4-carboxamide-1--4-ribofuranoside (AICAR) is widely used as an AMP-kinase activator, which regulates energy homeostasis and response to metabolic stress. Here, we investigated the effect of AICAR, an AMPK activator, on proliferation of various cancer cells and observed that proliferation of all the examined cell lines was significantly inhibited by AICAR treatment due to arrest in S-phase accompanied with increased expression of p21, p27, and p53 proteins and inhibition of PI3K-Akt pathway. Inhibition in in vitro growth of cancer cells was mirrored in vivo with increased expression of p21, p27, and p53 and attenuation of Akt phosphoryl- ation. Anti-proliferative effect of AICAR is mediated through acti- vated AMP-activated protein kinase (AMPK) as iodotubericidin and dominant-negative AMPK expression vector reversed the AICAR-mediated growth arrest. Moreover, constitutive active AMPK arrested the cells in S-phase by inducing the expression of p21, p27, and p53 proteins and inhibiting Akt phosphorylation, sug- gesting the involvement of AMPK. AICAR inhibited proliferation in both LKB and LKB knock-out mouse embryo fibroblasts to similar extent and arrested cells at S-phase when transfected with domi- nant negative expression vector of LKB. Altogether, these results indicate that AICAR can be utilized as a therapeutic drug to inhibit cancer, and AMPK can be a potential target for treatment of various cancers independent of the functional tumor suppressor gene, LKB. AMP-activated protein kinase (AMPK) 2 is a highly conserved serine/ threonine protein kinase. It is a heterotrimer containing a catalytic () and two regulatory subunits ( and ), each of which have at least two isoforms (1). AMPK is called the “fuel gauge” of the biological system, because it is activated under conditions that deplete cellular ATP and elevate AMP levels, such as glucose deprivation, heat shock, hypoxia, and ischemia (2, 3), and also by hormones like leptin (4), adiponectin (5), catecholamine (6), and interleukin-6 (7). Upon activation, AMPK phos- phorylates and inactivates a number of metabolic enzymes involved in ATP-consuming pathways like fatty acid, cholesterol synthesis, and protein synthesis that include enzymes like acetyl-Co enzyme A carbox- ylase (ACC), fatty acid synthase, 3-hydroxy-3-methylglutaryl-CoA reductase, and mammalian target of rapamycin (mTOR) and activates ATP-generating process like fatty acid oxidation and glucose uptake (8). The mechanisms of activating AMPK include direct allosteric binding of AMP to the subunits and phosphorylation, catalyzed by an upstream AMP kinase (AMPKK), recently identified to be LKB1 (STK11) (9 –11). Recent studies have demonstrated that AMPK can also be activated by other stimuli that do not cause a detectable change in the AMP/ATP ratio, like hyperosmotic stress and pharmacological agents like thiazolidinediones, metformin, and 5-aminoimidazole-4-carbox- amide-1--D-ribofuranoside (AICAR) (11–14). Activation of AMPK has been related with protection from injury and apoptosis caused by myocardial ischemia (15, 16) and apoptosis due to metabolic stress (17–19). In these scenarios, AMPK has been proposed as an anti-apoptotic molecule. However, recent reports have indicated anti-proliferative and pro-apoptotic action of activated AMPK using pharmacological agents or AMPK overexpression. AMPK activation has been shown to induce apoptosis in human gastric cancer cells (20), lung cancer cells (21), prostate cancer (22), pancreatic cells (23), and hepatic carcinoma cells (24) and enhance oxidative stress induced apo- ptosis in mouse neuroblastoma cells (25), by various mechanisms that includes inhibition of fatty acid synthase pathway and induction of stress kinases and caspase 3. AMPK is an anti-growth molecule because of its relationship with two tumor suppressor genes: LKB and TSC2 (tuberous sclerosis com- plex 2). LKB functions as an upstream kinase (AMPKK) that activates AMPK (26). LKB mutations result in Peutz-Jeghers syndrome, which results in predisposition to cancers of the colon, pancreas, breast, and other sites (27–29). Mutations of LKB1 typically occur in the catalytic domain, leading to loss of its kinase activity and presumably a failure to phosphorylate and activate AMPK (30). TSC2 forms a complex with TSC1 and inhibits mTOR, leading to inhibition in protein synthesis and negative regulation of cell size and growth (31). Mutations of TSC1TSC2 causes tuberous sclerosis, which is associated with hamar- tomatous polyps in multiple tissues and an increased risk of cancers (32). In the present study we have investigated the effect of AICAR on cell proliferation in vivo and in vitro in various cancer cell lines. AICAR is converted to its triphosphorylated form ZMP, inside the cell, by an adenosine kinase (14), which acts as an AMP analogue and activates AMPK and its upstream kinase LKB without affecting the ATP:AMP ratio in the cell (14). AICAR-mediated AMPK activation was found to be a proficient inhibitor of cell proliferation and the mechanism of its anti-proliferative effect may be mediated via inhibition of PI3K-Akt * This work was supported by National Institutes of Health Grants NS-22576, NS-34741, NS-40810, NS-37766, and NS-40144. The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked “advertisement” in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. 1 To whom correspondence should be addressed: Children Research Institute, Medical University of South Carolina, 173 Ashley Ave., 5th Floor, Charleston, SC 29425. Tel.: 843-792-7542; Fax: 843-792-7130; E-mail: [email protected]. 2 The abbreviations used are: AMPK, AMP-activated protein kinase; ACC, acetyl-CoA car- boxylase; mTOR, mammalian target of rapamycin; AMPKK, AMPK kinase; AICAR, 5-aminoimidazole-4-carboxamide-1--D-ribofuranoside; TSC2, tuberous sclerosis complex 2; DMEM, Dulbecco’s modified Eagle’s medium; FBS, fetal bovine serum; TdR, thymidine ribotide; PBS, phosphate-buffered saline; PCNA, proliferating nuclear antigen; GFP, green fluorescent protein; eGFP, enhanced GFP; AS, antisense; MS, mis- sense; PI3K, phosphatidylinositol 3-kinase; DN, dominant negarive; CA, constitutively active; MEF, mouse embryo fibroblasts; JNK, c-Jun NH 2 -terminal kinase; MTT, 3-(4,5- dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide; ZMP, AICA riboside mono- phosphate metabolite. THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 280, NO. 47, pp. 39582–39593, November 25, 2005 Printed in the U.S.A. 39582 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 280 • NUMBER 47 • NOVEMBER 25, 2005 by guest on January 10, 2020 http://www.jbc.org/ Downloaded from
Transcript
Page 1: 5-Aminoimidazole-4-carboxamide-1- D-ribofuranoside ... · pathway and increased expression of cell cycle inhibitory proteins p21, p27, and p53, thereby exhibiting potential as an

5-Aminoimidazole-4-carboxamide-1-�-D-ribofuranosideInhibits Cancer Cell Proliferation in Vitro and in Vivo viaAMP-activated Protein Kinase*

Received for publication, July 8, 2005, and in revised form, September 21, 2005 Published, JBC Papers in Press, September 21, 2005, DOI 10.1074/jbc.M507443200

Ramandeep Rattan‡, Shailendra Giri‡, Avtar K. Singh§, and Inderjit Singh‡1

From the ‡Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina 29425 and the §Departmentof Pathology and Laboratory Medicine, Ralph Johnson Veterans Affairs Medical Center, Charleston, South Carolina 29425

5-Aminoimidazole-4-carboxamide-1-�-4-ribofuranoside (AICAR)is widely used as an AMP-kinase activator, which regulates energyhomeostasis and response tometabolic stress.Here,we investigatedthe effect of AICAR, an AMPK activator, on proliferation of variouscancer cells and observed that proliferation of all the examined celllines was significantly inhibited by AICAR treatment due to arrestin S-phase accompanied with increased expression of p21, p27, andp53 proteins and inhibition of PI3K-Akt pathway. Inhibition in invitro growth of cancer cells was mirrored in vivo with increasedexpression of p21, p27, and p53 and attenuation of Akt phosphoryl-ation. Anti-proliferative effect of AICAR is mediated through acti-vated AMP-activated protein kinase (AMPK) as iodotubericidinand dominant-negative AMPK expression vector reversed theAICAR-mediated growth arrest. Moreover, constitutive activeAMPK arrested the cells in S-phase by inducing the expression ofp21, p27, andp53proteins and inhibitingAkt phosphorylation, sug-gesting the involvement ofAMPK.AICAR inhibitedproliferation inboth LKB and LKB knock-out mouse embryo fibroblasts to similarextent and arrested cells at S-phase when transfected with domi-nant negative expression vector of LKB. Altogether, these resultsindicate that AICAR can be utilized as a therapeutic drug to inhibitcancer, andAMPKcan be a potential target for treatment of variouscancers independent of the functional tumor suppressor gene, LKB.

AMP-activated protein kinase (AMPK)2 is a highly conserved serine/threonine protein kinase. It is a heterotrimer containing a catalytic (�)and two regulatory subunits (� and �), each of which have at least twoisoforms (1). AMPK is called the “fuel gauge” of the biological system,because it is activated under conditions that deplete cellular ATP andelevate AMP levels, such as glucose deprivation, heat shock, hypoxia,and ischemia (2, 3), and also by hormones like leptin (4), adiponectin (5),catecholamine (6), and interleukin-6 (7). Upon activation, AMPK phos-

phorylates and inactivates a number of metabolic enzymes involved inATP-consuming pathways like fatty acid, cholesterol synthesis, andprotein synthesis that include enzymes like acetyl-Co enzymeA carbox-ylase (ACC), fatty acid synthase, 3-hydroxy-3-methylglutaryl-CoAreductase, and mammalian target of rapamycin (mTOR) and activatesATP-generating process like fatty acid oxidation and glucose uptake (8).The mechanisms of activating AMPK include direct allosteric bindingof AMP to the � subunits and phosphorylation, catalyzed by anupstream AMP kinase (AMPKK), recently identified to be LKB1(STK11) (9–11). Recent studies have demonstrated that AMPK can alsobe activated by other stimuli that do not cause a detectable change in theAMP/ATP ratio, like hyperosmotic stress and pharmacological agentslike thiazolidinediones, metformin, and 5-aminoimidazole-4-carbox-amide-1-�-D-ribofuranoside (AICAR) (11–14).Activation ofAMPKhas been relatedwith protection from injury and

apoptosis caused by myocardial ischemia (15, 16) and apoptosis due tometabolic stress (17–19). In these scenarios, AMPK has been proposedas an anti-apoptotic molecule. However, recent reports have indicatedanti-proliferative and pro-apoptotic action of activated AMPK usingpharmacological agents or AMPK overexpression. AMPK activationhas been shown to induce apoptosis in human gastric cancer cells (20),lung cancer cells (21), prostate cancer (22), pancreatic cells (23), andhepatic carcinoma cells (24) and enhance oxidative stress induced apo-ptosis in mouse neuroblastoma cells (25), by various mechanisms thatincludes inhibition of fatty acid synthase pathway and induction ofstress kinases and caspase 3.AMPK is an anti-growth molecule because of its relationship with

two tumor suppressor genes: LKB and TSC2 (tuberous sclerosis com-plex 2). LKB functions as an upstream kinase (AMPKK) that activatesAMPK (26). LKB mutations result in Peutz-Jeghers syndrome, whichresults in predisposition to cancers of the colon, pancreas, breast, andother sites (27–29). Mutations of LKB1 typically occur in the catalyticdomain, leading to loss of its kinase activity and presumably a failure tophosphorylate and activate AMPK (30). TSC2 forms a complex withTSC1 and inhibits mTOR, leading to inhibition in protein synthesis andnegative regulation of cell size and growth (31). Mutations ofTSC1�TSC2 causes tuberous sclerosis, which is associated with hamar-tomatous polyps in multiple tissues and an increased risk of cancers(32).In the present study we have investigated the effect of AICAR on cell

proliferation in vivo and in vitro in various cancer cell lines. AICAR isconverted to its triphosphorylated form ZMP, inside the cell, by anadenosine kinase (14), which acts as an AMP analogue and activatesAMPK and its upstream kinase LKB without affecting the ATP:AMPratio in the cell (14). AICAR-mediated AMPK activation was found tobe a proficient inhibitor of cell proliferation and the mechanism of itsanti-proliferative effect may be mediated via inhibition of PI3K-Akt

* This work was supported by National Institutes of Health Grants NS-22576, NS-34741,NS-40810, NS-37766, and NS-40144. The costs of publication of this article weredefrayed in part by the payment of page charges. This article must therefore behereby marked “advertisement” in accordance with 18 U.S.C. Section 1734 solely toindicate this fact.

1 To whom correspondence should be addressed: Children Research Institute, MedicalUniversity of South Carolina, 173 Ashley Ave., 5th Floor, Charleston, SC 29425. Tel.:843-792-7542; Fax: 843-792-7130; E-mail: [email protected].

2 The abbreviations used are: AMPK, AMP-activated protein kinase; ACC, acetyl-CoA car-boxylase; mTOR, mammalian target of rapamycin; AMPKK, AMPK kinase; AICAR,5-aminoimidazole-4-carboxamide-1-�-D-ribofuranoside; TSC2, tuberous sclerosiscomplex 2; DMEM, Dulbecco’s modified Eagle’s medium; FBS, fetal bovine serum;TdR, thymidine ribotide; PBS, phosphate-buffered saline; PCNA, proliferating nuclearantigen; GFP, green fluorescent protein; eGFP, enhanced GFP; AS, antisense; MS, mis-sense; PI3K, phosphatidylinositol 3-kinase; DN, dominant negarive; CA, constitutivelyactive; MEF, mouse embryo fibroblasts; JNK, c-Jun NH2-terminal kinase; MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide; ZMP, AICA riboside mono-phosphate metabolite.

THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 280, NO. 47, pp. 39582–39593, November 25, 2005Printed in the U.S.A.

39582 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 280 • NUMBER 47 • NOVEMBER 25, 2005

by guest on January 10, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 2: 5-Aminoimidazole-4-carboxamide-1- D-ribofuranoside ... · pathway and increased expression of cell cycle inhibitory proteins p21, p27, and p53, thereby exhibiting potential as an

pathway and increased expression of cell cycle inhibitory proteins p21,p27, and p53, thereby exhibiting potential as an anti-cancer drug.

MATERIALS AND METHODS

Reagents and Cell Culture—DMEM/F-12, DMEM/4.5 g of glucosemedium, fetal bovine serum (FBS), and Hanks’ balanced salt solutionwere obtained from Invitrogen as was RPMI 1640. AICAR was pur-chased from Toronto Research Chemicals (Ontario, Canada). Iodotu-bericidin was obtained from Calbiochem. [3H]Thymidine ribotide([3H]TdR) was purchased from PerkinElmer Life Sciences. Propiumiodide, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide(MTT), and transfection reagent, FuGENE,were purchased fromRocheApplied Science. The enhanced chemiluminescence (ECL) detectingreagent was from Amersham Biosciences, and the luciferase assay sys-tem was from Promega (Madison, WI). C6 glioma, T98G astrocytoma,U87MG astrocytoma, MCF-7 breast cancer, and PC-3 prostrate carci-noma cell lines were obtained fromATCC (Rockville, MD), hematolog-ical cancer cell lines (CEMT-lymphoblast cells, K-562 chronic myelog-enous leukemia cells) were a kind gift from Dr. J. Barredo (MedicalUniversity of South Carolina). Primary antibodies, p21, p27, and p53were purchased fromSantaCruzBiotechnology (SantaCruz, CA).Anti-bodies against phosphospecific as well as pan-Akt, mTOR, and AMPKwere from Cell Signaling (Beverly, MA). AMPK�2-dominant negative

expression vector (D157A)was a kind gift fromDr. DavidCarling (MRCClinical Sciences Centre, London, UK), and AMPK�1 and �2 constitu-tive expression vectors were kind gifts fromDr. Jin-Zhong Zhang (CaseWestern Reserve University, Cleveland, OH) and Dr. Benoit Viollet(Rene Descartes University, Paris, France), respectively. LKB wild type,LKB dominant negative (kinase dead), STRAD� and MO25� expres-sion vectors were kind gifts from Dr. Dario R. Alessi (Wellcome TrustBiocenter, University of Dundee, Dundee, UK).

Cell Culture—C6 glioma cells, T98G, U87MG, and PC-3 were main-tained in DMEM/F-12 medium supplemented with 10% FBS and anti-biotics. MCF-7 cells were maintained in DMEM/4.5 g of glucose with10% FBS. CEM and K-562 were maintained in RPMI 1640 supple-mented with 10% FBS. LKB knock-out and wild-type mouse embryofibroblasts (MEFs)were a kind gift fromDr. Tomi P.Makela (Institute ofBiomedicine and Helsinki University Central Hospital, BiomedicumHelsinki, University of Helsinki, Finland) and were maintained inDMEM/4.5 g of glucose with 10% FBS, essential amino acids, and anti-biotics. All treatments were done in the presence of serum.

Thymidine Incorporation—Proliferation of cells was determined by[3H]thymidine ribotide ([3H]TdR) incorporation into DNA. 1 � 106

cells per well of adherent cell lines (C6,MCF-7, and PC3) and 0.25� 106

cells/well of suspension cell lines (NALM-6, CEM, CEMP, and K562)were plated in respectivemedias. Cells were incubated for 18–24 hwith

FIGURE 1. AICAR inhibits proliferation of cancercells. Specified numbers of cells (C6, MCF-7, PC3,CEM, and K562) were plated, treated with AICAR(0.25–1 mM), followed by exposure to [3H]thymi-dine for 6 h and subsequent counts. The data isrepresentation of three separate experimentsdone in triplicates. ***, p � 0.001 compared withcontrol; **, p � 0.01 compared with control, NS,non-significant compared with control.

AMPK as a Potential Target for Treatment of Cancer

NOVEMBER 25, 2005 • VOLUME 280 • NUMBER 47 JOURNAL OF BIOLOGICAL CHEMISTRY 39583

by guest on January 10, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 3: 5-Aminoimidazole-4-carboxamide-1- D-ribofuranoside ... · pathway and increased expression of cell cycle inhibitory proteins p21, p27, and p53, thereby exhibiting potential as an

orwithout the presence of AICAR at the indicated concentrations. Eachgroup was exposed to 37 kBq/ml [methyl-3H]thymidine in the samemedium for 6 h. The adherent cells were fixed by 5% trichloroacetic acidand lysed in SDS/NaOH lysis buffer overnight. Radioactivity was meas-ured by Beckman LS3801 liquid scintillation counter (Canada). Suspen-sion cell culture was harvested by cell harvester (Packard instrumentCo., Meriden, CT), and radioactivity was measured by 1450 microbetaliquid scintillation counter (PerkinElmer Life Sciences).

Clonogenic Assay—Cells were treated with AICAR for 18–24 h,trypsinized, counted, and 300 cells/100-mmplate were plated. The cellswere allowed to form colonies, and media was changed every third dayfor 2–3 weeks. The colonies were stained with MTT and enumerated(33).

FlowCytometry Assessment of Cell Cycle—Cellular DNA content wasassessed by flow cytometry. Cells were cultured in 6-well plates andtreated with AICAR or transfections were performed. Cells attached tothe platewere collectedwith trypsin, washed, and resuspended in 100�lof PBS, and 5 ml of 70% ethanol was added slowly while continuousvortexing of cells and were fixed overnight. Next day, cells were spun,washed, and suspended in 400 �l of PBS with addition of 10 mg/literRNase A and 75 �M propidium iodide. Cells were acquired by flowcytometry (BD Biosciences FACSCalibur flow cytometer) using ModfitLT software.

Immunoblot—After a stipulated time of incubation in the presence orabsence of AICAR, cells were scraped, washed with Hanks’ buffer, andsonicated in 50 mM Tris-HCl (pH 7.4) containing protease inhibitors (1mMphenylmethylsulfonyl fluoride, 5�g/ml aprotinin, 5�g/ml antipain,5�g/ml pepstatinA, and 5�g/ml leupeptin). Proteins (50�g/lane) wereresolved by SDS-PAGE and transferred onto nitrocellulosemembranes.Themembranes were blocked for 1 h in 5% nonfat drymilk in TTBS (20mM Tris, 500 mM NaCl, and 0.1% Tween 20, pH 7.5) and incubatedovernight in primary antibody (p21, p27, p53, Akt, �-actin, mTOR,PCNA at 1:2000 dilution) containing 5% nonfat dry milk for non-phos-pho antibodies and containing 5% albumin for phospho-antibodies(Akt-p, mTOR-p at 1:1000 dilution). The blots were washed four timeswith TTBS (5min/wash) and incubated for 45min at room temperaturewith respective horseradish peroxidase-conjugated secondary antibody(1:5000). The blots were washed three times in TTBS and once in 0.1 M

PBS (pH 7.4) at room temperature; protein expression was detectedwith ECL.

Animals—Adult male Wistar rats weighing 200–250 g were pur-chased fromCharles-River Laboratories. Animals weremaintained, andall protocols were approved by the animal use committees of the Med-ical University of South Carolina in compliance with the Guide for theCare and Use of Laboratory Animals (National Institutes of Health).

Tumor Implantation—C6 glioma cells were prepared fresh from cul-ture to ensure optimal viability of cells during tumor inoculation. Thecells were trypsinized, and 106 tumor cells prepared in 100 �l of PBSwere injected subcutaneously in the lateral side of the right hind leg ofthe rats, after shave and sterile preparation. On the 5th day of implan-tation 100mg/kg body weight/day of AICARwas given intra-peritonealuntil the 14th day, when the animals were sacrificed and the tumor wasexcised, weighed, and fixed in formalin (34).

Immunohistochemistry—Tumor sections were processed as previ-ously described (35). In brief, deparaffinized and rehydrated sectionswere microwaved for 10 min in antigen unmasking fluid (Vector Labo-ratories, Burlingame, CA), treated with 3% hydrogen peroxide in meth-anol to eliminate endogenous peroxidase activity and blocked to reducenonspecific staining. Sections were incubated overnight with primary

antibody (1:100) followed by tyramide signal enhancement technique(Renaissance TSA for Immunocytochemistry, PerkinElmer Life Sci-ences) per manufacturer’s instructions. After washing, slides were air-dried andmounted with aqueousmountingmedia (Vectashield, VectorLaboratories). The sectionswere examined under a fluorescencemicro-scope (Olympus BX-60) with an Olympus digital camera (Optronics,Goleta, CA) using a dual band pass filter. Images were captured andprocessed using Adobe Photoshop 7.0.

Transfection Studies—Plasmids were purified using the endotoxin-free plasmid midi prep kit (Qiagen). For transient transfections, C6 gli-oma cells were seeded in 6-well plates and grown to 60–80% confluencein DMEM/F-12 plus 5% FBS without antibiotics and transfected usingFuGENE reagent. 1–3 �g of AMPK DN or AMPK�1CA or AMPK�2CA expression vector along with 1 �g of eGFP expression vector orinsertless expression vector (pcDNA3.1) were used for transfecting.Cells were treated with AICAR for 24 h and processed for GFP-gatedDNA analysis by flow cytometry. Similarly, LKB1 wild type (1 �g) anddominant negative (1�g) alongwith STRAD� (0.5�g) andMO25� (0.5�g) expression vectors were used for transfection studies.

Antisense Experiments—To decrease the levels of endogenousAMPK, C6 glioma cells were transfected for 48 h with 25 �M phospho-thiorated antisense (AS) oligonucleotide (5�-CGCCCGTCGTCGT-GCTTCTGC-3�) directly against both the �1- and �2-subunits ofAMPK (36, 37) and a missense (MS) oligonucleotide (5�-CTCCCG-GCTTGCTGCCGT-3�) along with eGFP expression vector (36). Oli-gonucleotides were transfected with FuGENE reagent per themanufac-turer’s instructions. The cells were then treated with AICAR for 24 hand analyzed for cell cycle analysis by flow cytometry.

PI3KActivity—After 2 h of AICAR (1mM) treatment, cells were lysedwith ice-cold lysis buffer containing 1% v/vNonidet P-40, 100mMNaCl,20 mM Tris (pH 7.4), 10 mM iodoacetamide, 10 mM NaF, 1 mM sodiumorthovanadate, and protease inhibitors (Sigma-Aldrich). Lysates wereincubated at 4 °C for 15min, followed by centrifugation at 13,000� g for15 min. The supernatant was precleared with protein A/G-Sepharosebeads (Amersham Biosciences) for 1 h at 4 °C, followed by the addition

FIGURE 2. AICAR inhibits clonogenic potential of transformed cells. C6 glioma andPC3 prostate cells were treated with AICAR for 18 h, trypsinized and 300 cells/100-mmplate were plated to form colonies. The data are a representation of three separateexperiments done in triplicates. ***, p � 0.001 compared with control; **, p � 0.01compared with control.

AMPK as a Potential Target for Treatment of Cancer

39584 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 280 • NUMBER 47 • NOVEMBER 25, 2005

by guest on January 10, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 4: 5-Aminoimidazole-4-carboxamide-1- D-ribofuranoside ... · pathway and increased expression of cell cycle inhibitory proteins p21, p27, and p53, thereby exhibiting potential as an

FIGURE 3. AICAR causes cell cycle arrest in S-phase. C6 glioma and U87MG astrocytoma cells were treated with AICAR at indicated concentrations. After overnight fixation cells weresuspended in PBS with RNase A and propidium iodide and acquired for DNA content by flow cytometry using Modfit LT software. The first peak represents the cells in G0/G1 phase,the second peak with slashed bars represents the cells accumulated in S-phase, and the third peak represents cells in the M-phase. The data are also graphically represented aspercentage of cells in S-phase and M-phase. The data are a representative of three separate experiments. ***, p � 0.001 compared with control; **, p � 0.01 compared with control.

AMPK as a Potential Target for Treatment of Cancer

NOVEMBER 25, 2005 • VOLUME 280 • NUMBER 47 JOURNAL OF BIOLOGICAL CHEMISTRY 39585

by guest on January 10, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 5: 5-Aminoimidazole-4-carboxamide-1- D-ribofuranoside ... · pathway and increased expression of cell cycle inhibitory proteins p21, p27, and p53, thereby exhibiting potential as an

of 1 �g/ml p85 mAb. After 2-h incubation at 4 °C, protein G-Sepharosebeads were added, and the resulting mixture was further incubated for1 h at 4 °C. The immunoprecipitateswerewashed twicewith lysis buffer,once with PBS, once with 0.5 M LiCl and 100 mM Tris (pH 7.6), once inwater, and once in kinase buffer (20 mM HEPES, pH 7.4, 5 mM MgCl2,and 0.25mMEDTA). PI3K activity was determined using a lipidmixtureof 100 �l of 0.1 mg/ml phosphatidylinositol and 0.1 mg/ml phosphati-dylserine dispersed by sonication in 20 mM HEPES (pH 7.0) and 1 mM

EDTA. The reaction was initiated by the addition of 20 �Ci of[�-32P]ATP (3000 Ci/mmol, PerkinElmer Life Sciences) and 100 �M

ATP, and terminated after 15min by the addition of 80�l of 1 NHCl and200 �l of chloroform:methanol (1:1). Phospholipids were separated by

TLC and visualized by exposure to iodine vapor and autoradiography(38).

Statistical Analysis—The datawere statistically analyzed by perform-ing the Student-Newman-Keuls Test.

RESULTS

AICAR Inhibits Proliferation of Cancer Cells—To investigate theeffect of AICAR on the growth of various cancer cell lines, namely PC-3(human prostate cancer cell), MCF-7 (human breast cancer cell line),C6 glioma (rat transformed brain glial cells), U87MG (human astrocy-toma cell line), K-562 (human chronicmyelogenous leukemia cells), andCEM (human T-lymphoblast cells), cells were plated in their respective

FIGURE 4. AICAR inhibits cell proliferation in vivo. A, weight of excised tumor implanted in lateral side of the right flank of rats (n � 8) from vehicle (saline) and AICAR (100 mg/kgbody weight/day) treated. AICAR significantly reduced the tumor mass as compared with vehicle treated animals. *, p � 0.05 compared with vehicle (Bi). Immunofluorescentmicroscopy images showing decreased PCNA expression in AICAR-treated tumor sections as compared with vehicle-treated rats, stained as described under “Materials andMethods.” The number of enumerated cells is depicted graphically (Bii). Data are mean � S.D. of 10 fields from three different experiments. ***, p � 0.001 compared with control. C,Western blot depicting decreased expression of PCNA in the AICAR-treated tumor tissue from 2 different sets of animals as compared with vehicle-treated data. Each set had n � 6.

AMPK as a Potential Target for Treatment of Cancer

39586 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 280 • NUMBER 47 • NOVEMBER 25, 2005

by guest on January 10, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 6: 5-Aminoimidazole-4-carboxamide-1- D-ribofuranoside ... · pathway and increased expression of cell cycle inhibitory proteins p21, p27, and p53, thereby exhibiting potential as an

medium for growth and treatedwith different concentrations of AICAR(0.25–1 mM) for 24 h, and cell proliferation was examined by [3H]thy-midine uptake. AICAR inhibited the proliferation of all cell lines testedsignificantly in a dose-dependent manner (Fig. 1). All tested cell linesunderwent significant proliferation inhibition, indicating that this phe-nomenon is widespread and not limited to a specific cell type/line. Tofurther confirm this observation, a clonogenic assay was performed,where cells were treated with AICAR for 24 h, trypsinized and plated ata density of 300 cells/100-mm plate without AICAR. After 3 weeks,formed colonies were counted by staining the live cells with MTT.AICAR treatment significantly reduced the number of colonies beingformed as compared with the untreated cells (Fig. 2), suggesting that asingle treatment of AICAR treatment is sufficient to cause a sustainedinhibition of proliferation in different cancer cell lines.

AICAR Causes Cell Cycle Arrest in S-phase—Inhibition in prolifera-tion would indicate an anomaly in the cell cycle. To examine this, cellswere treated with AICAR (0.5–1 mM), and phases of cell cycle wereanalyzed by flow cytometry. Treatment of cells with AICAR resulted inaccumulation of cells in S-phase (peak with slashed bars), with almostno cells detected in M-phase (third peak) suggesting that inhibitionin proliferation by AICAR is due to the arrest of cell cycle at S-phase(Fig. 3).

AICAR Inhibits Proliferation in Vivo—To investigate whether theanti-proliferative effects of AICAR extends to the in vivo system, weutilized the rat flank tumor model (34). Wherein, C6 glioma cells (1 �106) were implanted aseptically in the right flank of the rat, and after 5days of tumor formation, animals were treated with 100 mg/kg bodyweight of AICAR intraperitoneally. On day 14, animals were sacrificed,and the tumors were excised, weighed, and fixed. Weight of the tumorswas taken as an index of tumor development and progression. AICARtreatment was able to reduce the growth of tumors in animals signifi-cantly (�50%) when compared with untreated animals (Fig. 4A). Toexamine the status of proliferating cells in vivo, immunohistochemistrywas performed on the sections of excised tumor tissues for PCNA (pro-

liferating cell nuclear antigen), a marker for proliferating cells. AICARsignificantly reduced the expression and number of cells exhibitingPCNA expression, indicating that the number of proliferating cells isreduced by AICAR treatment in vivo as demonstrated by immunohis-tochemistry and its expression by Western blot (Fig. 4, B and C). Thus,the anti-proliferative effect of AICAR is effective in vivo as well and canbe exploited for applications in attenuating cancer cell growth.

AICARMediates Its Anti-proliferative Action viaAMP-activated Pro-teinKinase—AICAR, is the earliest knownAMPKactivator, andmost ofits effects have been shown to be because of AMPK activation, althoughfew reports of its AMPK-independent effects exist (22). To investigate ifAMPK activation is responsible for the anti-proliferative effectsobserved by AICAR treatment, the phosphorylation of AMPK and itsdownstream target, ACC, an enzyme in the fatty acid synthesis pathway,was taken as an indicator of AMPK activation. AICAR induced thephosphorylation of AMPK and ACC in a dose- and time-dependentmanner as demonstrated inC6 glioma and PC3prostate cell lines (Fig. 5,A and B). Similar phosphorylation of ACC and AMPK was observed invivo, in the AICAR-treated excised tumor tissue (Fig. 5C). Iodotuberi-cidin is an inhibitor of adenosine kinase and inhibits the conversion ofAICAR to its activated form ZMP inside the cell and thus inhibits acti-vation of AMPK by AICAR. Cells were pretreated with iodotubericidin30min before the addition of AICAR (0.5–1mM), and proliferation wasmeasured after 16 h by [3H]thymidine uptake. Iodotubericidin treat-ment inhibited the proliferation arrest caused byAICAR thus indicatingthe involvement of AMPK (Fig. 6A). To further confirm the role ofAMPK,C6 glioma cells were transiently transfectedwith dominant neg-ative (DN) and constitutive active (CA) forms of AMPK along witheGFP expression vector. The cells were treated with AICAR for 18 h,and GFP-positive cells were analyzed by flow cytometry for DNA con-tent to determine the cells in S-phase. C6 glioma cells transfected withAMPK dominant negative were not able to undergo S-phase arrestwhen treated with AICAR (Fig. 6B). Inversely, C6 glioma cells trans-fected with expression vector of constitutive active AMPK�1 were

FIGURE 5. AICAR induces AMPK and ACC phos-phorylation in vitro and in vivo. A, C6 glioma andPC3 prostate cells were treated with AICAR at indi-cated concentrations and harvested at specifiedtime points, cell lysates were processed for thedetection of phospho-AMPK (p-Thr-172) andphospho-ACC by immunoblot as discussed under“Materials and Methods.” The blots are represen-tatives of three individual experiments done. B,cell lysates were prepared from the vehicle- andAICAR-treated tumor tissues from two differentset of animals (each set had n � 6) and processedfor the detection of phospho-AMPK (p-Thr-172)and phospho-ACC as above.

AMPK as a Potential Target for Treatment of Cancer

NOVEMBER 25, 2005 • VOLUME 280 • NUMBER 47 JOURNAL OF BIOLOGICAL CHEMISTRY 39587

by guest on January 10, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 7: 5-Aminoimidazole-4-carboxamide-1- D-ribofuranoside ... · pathway and increased expression of cell cycle inhibitory proteins p21, p27, and p53, thereby exhibiting potential as an

FIGURE 6. AICAR mediated its anti-proliferative action via AMPK. A, cells (CEM, K-526, and PC-3) were pretreated with iodotubericidin (0.1 �M) before addition of AICAR andassayed for [3H]thymidine incorporation. Iodotubericidin reversed the AICAR-induced proliferation block. The data are representative of three separate experiments performed intriplicates. ***, p � 0.001 compared with control; ###, p � 0.001 compared with AICAR (B and C) C6 glioma cells were transiently co-transfected with 2 �g of AMPK dominant negative(DN) (B) or AMPK constitutive active (CA) (C) and 1 �g of eGFP expression vector. The DNA content was normalized by pcDNA3. AICAR was added where indicated, and after 18 h cellswere fixed overnight and analyzed for arrest in S-phase as detailed under “Materials and Methods.” AICAR was not able to arrest the cells in the presence of AMPK DN (B), whereasAMPK CA expression was sufficient to arrest the cells in S-phase and showed additive effect with AICAR (C). The data are representative of three separate experiments. ***, p � 0.001compared with control; ###, p � 0.001 compared with AICAR; ##, p � 0.01 compared with AICAR; #, p � 0.05 compared with AICAR; NS, non-significant compared with control. D, cellswere transfected with AMPK antisense (AS) and missense (MS) oligonucleotides along with eGFP expression vector and treated with AICAR. The level of AMPK protein was reducedby the transfection of AS, whereas MS had no effect (inset) at 72 h. The cells were fixed and processed for GFP-gated S-phase arrest. Antisense of AMPK abolished the AICAR-mediatedS-phase arrest, whereas MS oligonucleotide had no effect. These data are representative of 3 separate experiments. ***, p � 0.001 compared with control; ###, p � 0.001 comparedwith AICAR; NS, non-significant compared with AICAR.

AMPK as a Potential Target for Treatment of Cancer

39588 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 280 • NUMBER 47 • NOVEMBER 25, 2005

by guest on January 10, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 8: 5-Aminoimidazole-4-carboxamide-1- D-ribofuranoside ... · pathway and increased expression of cell cycle inhibitory proteins p21, p27, and p53, thereby exhibiting potential as an

FIGURE 7. AICAR inhibits PI3K-Akt Pathway. A,C6 cells were treated with AICAR (1 mM) for 2 h andprocessed for PI3K activity as described under“Materials and Methods.” AICAR significantlyreduced the PI3K activity as assessed by inositol1,4,5-bisphosphate levels. The inositol 1,4,5-bisphosphate levels were measured by densitom-etry analysis. The blot is representative of threeseparate experiments. **, p � 0.01 compared withcontrol (B) C6 cells were treated with AICAR forincreasing time points as indicated, cells lysateswere prepared and analyzed for Akt (Ser-473) andmTOR (Ser-1448) phosphorylation by Westernblot as detailed under “Materials and Methods.” C,cell lysates were prepared from the treated tumortissue from two different set of animals and pro-cessed for the detection of phospho-Akt, whichwas reduced by AICAR treatment.

FIGURE 8. AICAR regulates the expression of cdkinhibitors via AMPK. Protein expression of cellcycle inhibitors p21, p27, and p53 was increasedby AICAR treatment as analyzed by immunoblot inC6 cells treated with AICAR (A) and tumor tissue(B). C, immunofluorescent microscopy images oftumor sections from vehicle and treated rats,stained with p21, p27, and p53 antibodies asdescribed under “Materials and Methods.” D, C6glioma cells were transiently transfected with 2 �gof AMPK dominant negative (DN) or AMPK consti-tutive active (CA) with DNA normalization donewith pcDNA and treated with AICAR where indi-cated. Cell lysates were prepared and assessed byimmunoblot for p21, p27, and phospho-Aktexpressions. AICAR induced p21 and p27 expres-sion and down-regulated phospho-Akt as before(lane 2), which was reversed by AMPK DN expres-sion (lane 3) and had no effect with AICAR treat-ment (lane 4). AMPK CA �1 and �2 overexpressionwas able to induce the p21 and p27 expressionand attenuate phospho-Akt by itself (lanes 5 and6). The blots are representatives of three individualexperiments done.

AMPK as a Potential Target for Treatment of Cancer

NOVEMBER 25, 2005 • VOLUME 280 • NUMBER 47 JOURNAL OF BIOLOGICAL CHEMISTRY 39589

by guest on January 10, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 9: 5-Aminoimidazole-4-carboxamide-1- D-ribofuranoside ... · pathway and increased expression of cell cycle inhibitory proteins p21, p27, and p53, thereby exhibiting potential as an

found to be arrested at S-phase similar to AICAR-treated cells (Fig. 6C).Further, we used the antisense (AS) approach to knock out the expres-sion of AMPK. C6 glioma cells were transfected with AMPK antisense(AS) and missense (MS) oligonucleotides, and levels of AMPK wereobserved 48 h post transfection. The level of AMPK protein was atten-uated by AS, whereasMS had no effect (Fig. 6D, inset).Moreover, trans-fectionwith antisense of AMPK alongwithGFP expression vector in C6glioma cells significantly reduced the AICAR-mediated S-phase arrest;however, MS oligonucleotide did not affect the potential of AICAR toarrest cells in S-phase (Fig. 6D). Taken together, these evidences pointstrongly toward a role for AMPK as an effective anti-proliferativesystem.

AICAR/AMPK Inhibits the PI3K-Akt Pathway—Because AICARinhibits cell proliferation and PI3K-Akt is one of the most importantpathways regulating proliferation, we examined the effect of AICAR onthe PI3K-Akt pathway. C6 glioma cells were treatedwithAICAR (1mM)for 2 h, and PI3K activity was assessed using phosphoinositol as a sub-strate, and we observed that AICAR treatment significantly reduced thePI3K activity (Fig. 7A). One of the downstream effectors of PI3K, Akt isthe main mediator regulating proliferation (39). AICAR also reducedthe phosphorylation of Akt in vitro and in vivo (Fig. 7, Bi and C). It alsoinhibited the phosphorylation (Ser-1448) of mTOR (Fig. 7Bii), which isa downstream target of Akt and regulates protein synthesis and cellgrowth (40). Thus, attenuation of the PI3K-Akt pathway may be one ofthe mechanisms by which AMPK negatively regulates growth.

AICAR Regulates the Expression of Cyclin-dependent Kinase Inhibi-tors via AMPK—Because AICAR inhibits the cell proliferation byarresting cells at S-phase in vitro as well as in vivo, we examined theexpression of cyclin-dependent kinase (cdk) inhibitors, which bind tocyclin-cdk complexes and inhibit the progression of cell cycle. AICARinduced the expression of p21 and p27, the cip/kip protein cdk inhibi-tors in a time-dependent manner (Fig. 8A). It also induced the expres-sion of p53, which is known to regulate the cell cycle as well as p21expression (41). The expression of p21, p27, and p53 proteins, were alsoincreased in vivo, as assessed by immunohistochemistry of tissue sec-tions and by Western blot analysis of protein isolated from excisedtumor tissue (Fig. 8, B andC). The effect of AICAR on the expression ofgrowth regulators is mediated via activation of AMPK, because trans-fected AMPK DN abolished the AICAR-mediated induction of p21,whereas the CA form of AMPK�1 and �2 induced the expression byitself (Fig. 8D). In case of p27, AMPK DN reduced the AICAR-inducedexpression, butAMPK�1 and�2CA formswere only able to induce p27protein marginally compared with AICAR. In case of Akt, DNAMPK�2-transfected cells did not respond to the AICAR-mediatedinhibition in Akt phosphorylation, whereas, in CA-transfected cells,there was significant inhibition (Fig. 8E), indicating that AMPK activa-tion is responsible for increase in cdk inhibitor protein expressions andinhibition of Akt phosphorylation.

LKB (AMPKK) Status Does Not Affect AICAR-mediated GrowthArrest—LKB is a recently discovered upstream target of AMPK (AMPKkinase, AMPKK), which phosphorylates AMPK at Thr-172 for its fullactivation. LKB itself is a tumor suppressor gene and inactivation of LKBresults in predisposition to various cancers (27–29). It is being hypoth-esized that the anti-tumor effects of LKB are due to AMPK activity. Toexamine the possible involvement of LKB in AICAR/AMPK-inducedgrowth arrest, we utilized LKB knock-out (LKB�/�) and LKB WT(LKB�/�) MEF cell lines. AICAR was able to induce the phosphoryla-tion of AMPK and ACC to a similar extent in both MEFs (Fig. 9A).AICARwas able to inhibit proliferation in both knock-out andwild-typeMEFs to a similar extent (Fig. 9B). This was further supported by similar

S-phase arrest observed when C6 cells were transiently transfected withLKB dominant negative and wild-type expression vector along withexpression vectors of its cofactors, STRAD� and MO25�, and treatedwith AICAR (Fig. 9C). These data indicate that AMPK activation byAICAR is sufficient to cause growth arrest and does not require activa-tion by LKB.

DISCUSSION

In this study we have demonstrated that AMPK activation by AICARresults in growth arrest at S-phase due to inhibition of PI3K-Akt path-way and up-regulation of cdk inhibitors, independent of its upstreamkinase LKB. This inference is based on the following observations: 1)Treatment of various cancer cell lines by AICAR attenuated the prolif-

FIGURE 9. LKB (AMPKK) status does not affect AICAR mediated growth arrest. A, LKBknock-out (LKB�/�) and wild-type (LKB�/�) mouse embryo fibroblast were treated withAICAR and analyzed by immunoblot for AMPK and ACC phosphorylation. The blots arerepresentatives of three individual experiments done. B, LKB knock-out (LKB�/�) andwild-type (LKB�/�) mouse embryo fibroblast were treated with varying concentrationsof AICAR and assayed for [3H]thymidine incorporation. The data are representative ofthree separate experiments. ***, p � 0.001 compared with control. C, C6 glioma cellswere transiently transfected with 1 �g of LKB dominant negative (DN) or LKB wild type(WT) with STRAD� (0.5 �g) and MO25� (0.5 �g) expression vectors, eGFP, and pcDNAand treated with AICAR where indicated. After 18 h cells were fixed overnight and ana-lyzed for arrest in S-phase as detailed under “Materials and Methods.”

AMPK as a Potential Target for Treatment of Cancer

39590 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 280 • NUMBER 47 • NOVEMBER 25, 2005

by guest on January 10, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 10: 5-Aminoimidazole-4-carboxamide-1- D-ribofuranoside ... · pathway and increased expression of cell cycle inhibitory proteins p21, p27, and p53, thereby exhibiting potential as an

eration both in vitro and in vivo studies. 2) The attenuation of cellproliferation was due to the activation of AMPK as evident from aden-osine kinase inhibitor studies (iodotubericidin), the use of expressionvectors (dominant negative and constitutive active) and AMPK anti-sense experiments. 3) The growth arrest is mediated by inhibition ofPI3K activity and Akt phosphorylation and up-regulation of cell-cycleinhibitor proteins p21, p27, and p53. 4) Activation of AMPK in theabsence of LKB also results in growth arrest. We show here the directrelation betweenAMPK activation and growth inhibition in vitro and invivo. In addition, these observations strongly indicateAICAR, anAMPKactivator to be an efficient anti-proliferative agent in vitro and in vivo.Being a pharmacological activator of AMPK, AICAR has been used

extensively to study its role in physiology (13, 14). It has recently beenshown to have anti-inflammatory properties that were reported to bemediated by AMPK activation (36, 42), although its AMPK-independ-ent effects have also been reported (22). In our study, AICAR mediatesits effect via activation of AMPK, which is supported by both pharma-cological (iodotubericidin) as well as molecular approaches (DN, CA,and AS AMPK transfections). Treatment of iodotubericidin, inhibitorof conversion of AICAR to ZMP, abrogated the S-phase arrest of cells(Fig. 6A). Moreover, inhibition of AMPK either by its DN expressionvector or AS oligonucleotide also resulted in the reversal of AICAR-mediated growth arrest, whereas AMPK CA expression was able toaccumulate cells in S-phase (Fig. 6, B–D). AMPK DN also blunted theelevated expression of p21 and p27, whereasAMPKCA itself was able toinduce their expression (Fig. 8). The induction of p27 by AMPK CA �1and �2 seems to be marginal as compared with p21. The observationrepresented here is consistent and reproducible. Right now we do nothave an explanation for this disparity. One of the possible explanationscould be that p27 regulation is related to the differential localization ofthe �1 (cytosol) and �2 (nuclear) isoforms of the catalytic subunits ofAMPK, which is yet to be established in terms of p27 regulation. AICARseems to affect the PI3K-Akt proliferation pathway, becauseAICARandAMPK CA inhibited PI3K activity and Akt phosphorylation (Fig. 7).

These evidences point strongly toward AMPK activation being themajor cause of growth arrest. Thus, AMPK can be considered as a neg-ative regulator of proliferation and can modulate protein expression tothis effect, classifying it as a tumor suppressor system that can beexploited for attenuation of cancers.Activation of AMPK by AICAR, metformin, or thiazolidinediones or

expression of constitutively active mutants has been shown to causedeath or attenuate the growth of cancer cells. AICAR- and rosiglita-zone-mediated AMPK activation caused proliferation block and celldeath by inhibiting fatty acid and protein synthesis pathways andincreasing p21 expression in prostate cells (22). Adenosine-inducedAMPK was shown to cause apoptosis in gastric cancer cells (20), andactivation of AMPK by AICAR and its CA form was shown to causeapoptosis in pancreatic cells by inducing JNK pathway (23). Similarly,AMPK induced JNK and caspase 3 activity resulting in apoptosis in livercells (24). AMPK activation was also demonstrated to enhance H2O2-mediated apoptosis in neuroblastoma cells by inducingNF-� and p38-JNKpathways (25). These studies, alongwith the present study, suggestsAMPK as an efficient growth inhibitor and apoptosis inducer. On theother hand, it also has been shown to have a protective effect on stress-injured cells in heart ischemia and reperfusion injury model (15, 18).AMPK activation protects primary astrocytes from fatty acid-induceddeath by inhibiting de novo ceramide synthesis (17) and protects humanumbilical vein endothelial cells from hyperglycemia by inhibition ofcaspase 3 and Akt activation (18) and by similar mechanism in thymo-cytes (19). In pancreatic cancer cells, AMPK was shown to bestow tol-erance toward nutrient deprivation (43). These studies presentedAMPK as a protective agent. The reason for these apparently opposingeffects of AMPK activation in cell survival and cell death is not known,but it can be speculated that in actively dividing cancer cells, the inhibi-tion of ATP-consuming processes by AMPK may be less compatiblewith their survival, whereas in non-dividing cells, where the protectiveeffects of AMPK have been observed under acute stress, the shutdownof ATP-consuming pathways may not alter the balance for survival.

FIGURE 10. A schematic representation of effectof AICAR on proliferation of cancer cells. AICAR,upon entering the cell, is converted to ZMP, whichactivates AMPK. Upon activation AMPK increasesthe expression of p21, p27, and p53 proteins,which may be responsible for the S-phase arrestbeing observed. On the other hand it inhibits thePI3K activity and Akt phosphorylation, whichresults in inhibition of mTOR and speculative reg-ulation of other targets like MDM2, Bad, andcaspase 9, resulting in a proliferation and cellgrowth block. The overall signaling taking placeresults in a sustained proliferation arrest, whichcan ultimately lead to loss of viability due to onsetof senescence or apoptotic pathways.

AMPK as a Potential Target for Treatment of Cancer

NOVEMBER 25, 2005 • VOLUME 280 • NUMBER 47 JOURNAL OF BIOLOGICAL CHEMISTRY 39591

by guest on January 10, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 11: 5-Aminoimidazole-4-carboxamide-1- D-ribofuranoside ... · pathway and increased expression of cell cycle inhibitory proteins p21, p27, and p53, thereby exhibiting potential as an

AMPKas a tumor suppressor systemhas gainedmore scientific inter-est because of its link with two tumor suppressors. One of these is thetuberous sclerosis complex 2 (TSC2), mutations of which cause tuber-ous sclerosis, an autosomal dominant disorder, which in humans isassociated with hamartomatous polyps in multiple tissues and anincreased risk of cancers. TCS2 forms an inhibitory complex with TSC1and inhibits mTOR, leading to negative regulation of cell size andgrowth (31, 32). TSC2 is phosphorylated and inhibited by Akt. Theobservation thatAMPKphosphorylates and activatesTSC2 suggests yetanother mechanism for the inhibitory effect of AMPK on growth (44,45). In this regard our observation that AMPK inhibits PI3K activity andAkt phosphorylation (Fig. 7), points toward another route for regulatingnot only the TSC2-mTOR pathway but also other downstream playersof Akt that control proliferation. The tumor suppressor, LKB1 (Stk11)functions as an upstream kinase, which phosphorylates and activatesAMPK in physiological settings. Mutations of LKB1 have been found inPeutz-Jeghers syndrome, an autosomal dominant disorder character-ized by multiple hamartomatous polyps (benign overgrowth of differ-entiated tissues) of the colon and a predisposition to cancers of thecolon, pancreas, and other locations in the gastrointestinal tract (27, 29).Mutations of LKB1 typically occur in the catalytic domain, leading toloss of its kinase activity and presumably a failure to phosphorylateAMPK. According to our observation in LKB-null MEF cells, AICARwas able to activate AMPK and inhibit proliferation in both cells to asimilar extent (Fig. 9), indicating that AMPK can act as a tumor sup-pressor even in absence of functional LKB. Thus, activation of AMPKbyany other means or agents can also be exploited for the therapeutics forPeutz-Jeghers syndrome, where LKB is mutated.Taken together, we conclude that activation of AMPK, by AICAR in

this case results in activation of p21, p27, and p53, which results in cellcycle arrest. The elevation in the expression of p21may be a direct effectof AMPK or may be occurring via p53, which remains to be explored.On the other hand it not only inhibits TSC2 complex as well docu-mented by others (44, 45) but also attenuates the PI3K and Akt activity,which would further add to the negative regulation of TSC2-mTORpathway (Fig. 10). Inhibition of Akt would also result in negative regu-lation of MDM2 that would help in up-regulating p53 activity, andforkhead, BAD, and caspase 9 (46), which would all assist inmanifestingthe anti-proliferative effect of AMPK activation. The exact mechanismsneed to be worked out, because the regulation of proliferation and cellgrowth by AMPK appears to be quite complex. Sustained inhibition ofcell cycle and/or proliferation can be hypothesized to lead to cell deathby senescence or apoptosis. Overall, AMPK activation by AICAR or anyother pharmacological agent is an attractive target for cancer therapy.

Acknowledgments—We thank Dr. J. Barredo (Medical University of SouthCarolina, SC) for his kind gift of CEM T-lymphoblast and K-562 chronicmyelogenous leukemia cell lines. We thank Dr. David Carling (MRC ClinicalSciences Centre, London, UK) for his kind gift of AMPK�2-dominant negativeexpression vector (D157A). We thank Dr. Jin-Zhong Zhang (Case WesternReserve University, Cleveland, OH) and Dr. Benoit Viollet (Rene DescartesUniversity, Paris, France) for their kind gift of AMPK�1 and �2 constitutiveactive constructs, respectively. We thank Dr. Dario R. Alessi (Wellcome TrustBiocentre, University of Dundee, andDundee, UK) for the kind gift of LKBwildtype, LKBdominant negative (kinase-dead), STRAD�, andMO25� expressionvectors. We thank Dr. Tomi P. Makela (Institute of Biomedicine and HelsinkiUniversity Central Hospital, Biomedicum Helsinki, University of Helsinki,Finland) for his kind gift of LKB knock-out and wild-type mouse embryo fibro-blasts. We thank Joyce Bryan, Carrie Barnes, and Hope Terry for laboratoryassistance.

REFERENCES1. Hardie, D. G., and Carling, D. (1997) Eur. J. Biochem. 246, 259–2732. Davies, S. P., Carling, D., and Hardie, D. G. (1989) Eur. J. Biochem. 186, 123–1283. Kemp, B. E., Stapleton, D., Campbell, D. J., Chen, Z. P.,Murthy, S.,Walter,M., Gupta,

A., Adams, J. J., Katsis, F., van Denderen, B., Jennings, I. G., Iseli, T., Michell, B. J., andWitters, L. A. (2003) Biochem. Soc. Trans. 31, 162–168

4. Minokoshi, Y., Kim, Y. B., Peroni, O. D., Fryer, L. G.,Muller, C., Carling, D., andKahn,B. B. (2002) Nature 415, 339–343

5. Yamauchi, T., Kamon, J., Minokoshi, Y., Ito, Y., Waki, H., Uchida, S., Yamashita, S.,Noda, M., Kita, S., Ueki, K., Eto, K., Akanuma, Y., Froguel, P., Foufelle, F., Ferre, P.,Carling, D., Kimura, S., Nagai, R., Kahn, B. B., and Kadowaki, T. (2002) Nat. Med. 8,1288–1295

6. Ruderman, N. B., Park, H., Kaushik, V. K., Dean, D., Constant, S., Prentki, M., andSaha, A. K. (2003) Acta Physiol. Scand. 178, 435–442

7. Kelly, M., Keller, C., Avilucea, P. R., Keller, P., Luo, Z., Xiang, X., Giralt, M., Hidalgo,J., Saha, A. K., Pedersen, B. K., and Ruderman, N. B. (2004) Biochem. Biophys. Res.Commun. 320, 449–454

8. Luo, Z., Saha, A. K., Xiang, X., and Ruderman, N. B. (2005)Trends Pharmacol. Sci. 26,69–76

9. Hong, S. P., Leiper, F. C., Woods, A., Carling, D., and Carlson, M. (2003) Proc. Natl.Acad. Sci. U. S. A. 100, 8839–8843

10. Woods, A., Johnstone, S. R., Dickerson, K., Leiper, F. C., Fryer, L. G., Neumann, D.,Schlattner, U., Wallimann, T., Carlson, M., and Carling, D. (2003) Curr. Biol. 13,2004–2008

11. Hawley, S. A., Boudeau, J., Reid, J. L., Mustard, K. J., Udd, L., Makela, T. P., Alessi,D. R., and Hardie, D. G. (2003) J. Biol. 2, 28

12. Fryer, L. G., Parbu-Patel, A., and Carling, D. (2002) J. Biol. Chem. 277, 25226–2523213. Sullivan, J. E., Brocklehurst, K. J., Marley, A. E., Carey, F., Carling, D., and Beri, R. K.

(1994) FEBS Lett. 353, 33–3614. Corton, J. M., Gillespie, J. G., Hawley, S. A., and Hardie, D. G. (1995) Eur. J. Biochem.

229, 558–56515. Russell, R. R., 3rd, Li, J., Coven, D. L., Pypaert,M., Zechner, C., Palmeri,M., Giordano,

F. J., Mu, J., Birnbaum, M. J., and Young, L. H. (2004) J. Clin. Invest. 114, 495–50316. Nishino, Y., Miura, T., Miki, T., Sakamoto, J., Nakamura, Y., Ikeda, Y., Kobayashi, H.,

and Shimamoto, K. (2004) Cardiovasc. Res. 61, 610–61917. Blazquez, C., Geelen, M. J., Velasco, G., and Guzman, M. (2001) FEBS Lett. 489,

149–15318. Ido, Y., Carling, D., and Ruderman, N. (2002) Diabetes 51, 159–16719. Stefanelli, C., Stanic, I., Bonavita, F., Flamigni, F., Pignatti, C., Guarnieri, C., and

Caldarera, C. M. (1998) Biochem. Biophys. Res. Commun. 243, 821–82620. Saitoh, M., Nagai, K., Nakagawa, K., Yamamura, T., Yamamoto, S., and Nishizaki, T.

(2004) Biochem. Pharmacol. 67, 2005–201121. Li, J., Jiang, P., Robinson, M., Lawrence, T. S., and Sun, Y. (2003) Carcinogenesis 24,

827–83422. Xiang, X., Saha, A. K.,Wen, R., Ruderman,N. B., and Luo, Z. (2004)Biochem. Biophys.

Res. Commun. 321, 161–16723. Kefas, B. A., Cai, Y., Ling, Z., Heimberg,H., Hue, L., Pipeleers, D., andVan deCasteele,

M. (2003) J. Mol. Endocrinol. 30, 151–16124. Meisse, D., Van de Casteele, M., Beauloye, C., Hainault, I., Kefas, B. A., Rider, M. H.,

Foufelle, F., and Hue, L. (2002) FEBS Lett. 526, 38–4225. Jung, J. E., Lee, J., Ha, J., Kim, S. S., Cho, Y.H., Baik, H.H., andKang, I. (2004)Neurosci.

Lett. 354, 197–20026. Hardie, D. G., Scott, J. W., Pan, D. A., and Hudson, E. R. (2003) FEBS Lett. 546,

113–12027. 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.,Olschwang, S., Olsen, A. S., Stratton,M. R., dela Chapelle, A., and Aaltonen, L. A. (1998) Nature 391, 184–187

28. 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

29. Nakanishi, C., Yamaguchi, T., Iijima, T., Saji, S., Toi, M., Mori, T., and Miyaki, M.(2004) Oncology 67, 476–479

30. Forcet, C., Etienne-Manneville, S., Gaude, H., Fournier, L., Debilly, S., Salmi,M., Baas,A., Olschwang, S., Clevers, H., and Billaud, M. (2005) Hum. Mol. Genet. 14,1283–1292

31. Inoki, K., Zhu, T., and Guan, K. L. (2003) Cell 115, 577–59032. Li, Y., Corradetti, M. N., Inoki, K., and Guan, K. L. (2004) Trends Biochem. Sci. 29,

32–3833. Giocanti, N., Hennequin, C., Rouillard, D., Defrance, R., and Favaudon, V. (2004)Br. J.

Cancer 91, 2026–203334. Parsa, A. T., Chakrabarti, I., Hurley, P. T., Chi, J. H.,Hall, J. S., Kaiser,M.G., andBruce,

J. N. (2000) Neurosurgery 47, 993–999; discussion 999–100035. Nath, N., Giri, S., Prasad, R., Singh, A. K., and Singh, I. (2004) J. Immunol. 172,

1273–1286

AMPK as a Potential Target for Treatment of Cancer

39592 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 280 • NUMBER 47 • NOVEMBER 25, 2005

by guest on January 10, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 12: 5-Aminoimidazole-4-carboxamide-1- D-ribofuranoside ... · pathway and increased expression of cell cycle inhibitory proteins p21, p27, and p53, thereby exhibiting potential as an

36. Giri, S., Nath,N., Smith, B., Viollet, B., Singh, A. K., and Singh, I. (2004) J. Neurosci. 24,479–487

37. Culmsee, C., Monnig, J., Kemp, B. E., and Mattson, M. P. (2001) J. Mol. Neurosci. 17,45–58

38. Giri, S., Rattan, R., Singh, A. K., and Singh, I. (2004) J. Immunol. 173, 5196–520839. Brazil, D. P., Yang, Z. Z., and Hemmings, B. A. (2004) Trends Biochem. Sci. 29,

233–24240. Harrington, L. S., Findlay, G. M., and Lamb, R. F. (2005) Trends Biochem. Sci. 30,

35–4241. el-Deiry,W. S., Tokino, T., Velculescu, V. E., Levy, D. B., Parsons, R., Trent, J. M., Lin,

D., Mercer, W. E., Kinzler, K. W., and Vogelstein, B. (1993) Cell 75, 817–82542. Nath,N., Giri, S., Prasad, R., Salem,M. L., Singh, A. K., and Singh, I. (2005) J. Immunol.

175, 566–57443. Kato, K., Ogura, T., Kishimoto, A., Minegishi, Y., Nakajima, N., Miyazaki, M., and

Esumi, H. (2002) Oncogene 21, 6082–609044. Kimura, N., Tokunaga, C., Dalal, S., Richardson, C., Yoshino, K., Hara, K., Kemp, B. E.,

Witters, L. A., Mimura, O., and Yonezawa, K. (2003) Genes Cells 8, 65–7945. Bolster, D. R., Crozier, S. J., Kimball, S. R., and Jefferson, L. S. (2002) J. Biol. Chem. 277,

23977–2398046. Song, G., Ouyang, G., and Bao, S. (2005) J. Cell Mol. Med. 9, 59–71

AMPK as a Potential Target for Treatment of Cancer

NOVEMBER 25, 2005 • VOLUME 280 • NUMBER 47 JOURNAL OF BIOLOGICAL CHEMISTRY 39593

by guest on January 10, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 13: 5-Aminoimidazole-4-carboxamide-1- D-ribofuranoside ... · pathway and increased expression of cell cycle inhibitory proteins p21, p27, and p53, thereby exhibiting potential as an

Ramandeep Rattan, Shailendra Giri, Avtar K. Singh and Inderjit Singh via AMP-activated Protein Kinasein Vivo and in VitroProliferation -D-ribofuranoside Inhibits Cancer Cellβ5-Aminoimidazole-4-carboxamide-1-

doi: 10.1074/jbc.M507443200 originally published online September 21, 20052005, 280:39582-39593.J. Biol. Chem. 

  10.1074/jbc.M507443200Access 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

  http://www.jbc.org/content/280/47/39582.full.html#ref-list-1

This article cites 46 references, 9 of which can be accessed free at

by guest on January 10, 2020http://w

ww

.jbc.org/D

ownloaded from


Recommended