j. biol. chem.-2007-pandey-14291-9

9
Reduc ti on of Low Molecular Weight Prot ei n- tyrosi ne Phosphatase Expression Improves Hyp erg lyc emia and Insulin Sensitivity in Obese Mice * Receiv ed forpubl ica tio n,October12, 2006, an d in rev ise d for m, Ma rch9, 200 7  Pu blish ed, JBC Pape rs in Pres s, Mar ch 12, 2007 , DOI 10.1 074/ jbc. M609626200 San jay K. Pan dey ‡1 , Xi ng Xia n Yu , Lynnet ta M. Watts , M. Dods on Mi chael § , Kyle W. Sl oop § , Amber R. Ri vard , ThomasA. Leedom , Var a PrasadManchem , Laura Samadzadeh , Robert A. McKay , Bret t P.Monia , and Sanja y Bha not From the Met abo lic Dise ase Progra m, Ant isen se Drug Disc ove ry, IsisPharmaceuticals, Car lsba d, CA 92008, and § Endoc rine Discov ery, Lilly Resear ch Labor atories, India napol is, Indi ana 46285 To investigate the role of low molecular weight protein- tyros ine phosphatase (LMW-PTP) in glucose metabolism and insulin act ion,a spe cif ic antisense oli gonucle otide (ASO)  was used to reduce its express ion both  in vitro  a nd  in vivo. Reduct ion of LMW-PTPexpres sion wit h the ASOin cul tured mouse hepatocyt es and in liver and fat tissues of die t-induced obese (DIO) mice and  ob/ob mice led to increased phospho- rylation and activi ty of key insulin signali ng intermediates, including insulin receptor-  subunit, phosphatidylinositol 3-kinase, and Akt in response to insulin stimulation. The ASO-treated DIO and  ob/ob animals showed improved insu- lin sensitivity, which was reflected by a lowering of both plasma insulin and glucose levels and improved glucose and insulin toler ance in DI O mi ce. The treatment did not decrease body weight or increase metabolic rate. These data demonstrate that LMW-PTP is a key negative regulator of insuli n act ion and a potential novel targetfor thetreatmentof  insulin resistance and type 2 diabetes. The incidence of diabetes has been steadily increasing and hasbecome a maj or publichealt h concern. Ove r 85% of dia bet ic patients have type 2 diabetes. Obesity, which can result from a sed ent ary lif e sty le andhigh cal oriediet,is a maj or ris k fac torfor the development of this disorder (1). A hallmark of type 2 dia- betes is insulin resistance, characterized by a decreased insulin response in a va ri et y of ti ssues (2), incl uding li ve r, fa t, and mus - cle. Therefore, increasing insulin sensitivity is a practical strat- egy for the treatment of type 2 diabetes. Insuli n initiates its physi ologic al respon se by bindin g to its membrane-bound receptor (IR, a   2 2 -heterotetramer pro- tein), 2 which causes autophosphorylation of the -subunit and receptor acti vatio n, result ing in subse quent phosp horyla tion of its two major downstream substrates, IRS-1 and IRS-2 (3–5). Phosphorylated IRS-1 and -2 interact with and activate other SH2 domain -conta ining adapter molecules such as NCK2, Grb2, Shc, Syp (4– 8), and the regulatorysubuni t (p85) of phos- phatidylinositol 3-kinase (PI3-K) (9, 10). Activated PI3-K stim- ula tes Akt (or protein kinase B) tha t in turn pho sphory lat es and inactivates glycogen synthase kinase-3 (11), resulting in activa- tion of glyco gen synthase (12), there by increasing the utiliz a- tion of glucose for glycogen synthesis. In fat and muscle, acti-  vation of this pathway also causes transfer of GLUT4 from the cytoplasm to the cell membrane, resulting in increased glucose uptake (13). Therefore, IR-IRS-1/2-PI3-K-Akt signaling cas- cade is a key pathway in mediating the effects of insulin action on blood glucose levels. A number of studies have established a role for intracellular phosphatases in the negative regulation of insulin signaling (14–16) such as protein-tyrosine phosphatase (PTP) 1B that negatively regulates insulin action through dephosphorylating tyrosi ne-pho sphor ylated IR (15, 16). Insulin sensitivit y was enhanced in PTP1B knock-out mice, in which increased tyro- sine phosphorylation of IR was found in liver and muscle but not in adipose tissue. Insulin sensitivity was also increased in ob/ob  and  db/db  mice treated with antisense inhibitors that reduced PTP1B expression in liver and fat (17, 18). Further- more, in these mice insulin signaling was enhanced in liver but not in fat tissue. Therefore, PTP1B negatively regulates insulin signaling with tissue specificity (15, 16). In addition, PTEN and SHIP2 also negatively regulate insulin signaling  in vivo  via dephosphorylating phosphatidylinositol 3,4,5-trisphosphate, which results in decreased phosphorylation and inactivation of Akt. Enhanced insulin sensitivity was observed in  ob/ob  and db/db  mice aft er antise nse reduct ion of PTE N expres sion which was accompanied by increased levels of phosphorylated Akt at serine 473 (Akt Ser473 ) in liver (14). Increased phospho- rylate d Akt Ser473 leve ls in both li ver and muscle we re al so found in SHI P2 knoc k-out mic e (19) . Mor eov er, protec tio n from die t- induced insulin resistance was observed in these knock-out mice whe n feda hig hfatdiet(19).Therefore,intracellularphos- phatases pla y a key role in insulin si gnal ing acti vi ty and in mod- ulating insulin sensitivity. * The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked “ advertise- ment in ac cor da ncewith18 U.S.C.Sect ion1734solely to ind ica te thi s fac t. 1  To whom corresponden ce should be addressed: Metabolic Disease Pro- gram, Antisense Drug Discovery, Isis Pharmaceuticals, 1896 Rutherford Rd., Carlsbad, CA 92008. Tel.: 760-603-2361; Fax: 760-603-3862; E-mail: [email protected]. 2  The abbreviations used are: IR, insulin receptor; IRS, insulin receptor sub - strate; LMW, low molecular weight; PTP, protein-tyrosine phosphatase; PI3-K, phosphatidylinositol 3-kinase; ASO, antisense oligonucleotide; DIO, diet-indu ced obese; GTT,glucose toler ancetest; ITT,insulin toler ancetest; DGAT, acyl-CoA:diacylglyc erol acyltransferase; PTEN, phosphatase and tensin homolog deleted on chromosome ten; SHP2/SHPTP2, Src homol- ogyregion 2-do mainphosphata se-2 ; Akt Ser473 , Aktat ser ine473;BW,body weight; SHIP-2, SH2-containing inositol phosphatase 2.  THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 282, NO. 19, pp. 14 291–14299, May 11, 2007 © 2007 by The American Society for Biochemistry and Molecular Biol ogy, Inc. Printed in the U .S.A. MAY 11, 2007• VOLUME 282 • NUMB ER 19  JOURNAL OF BIOLOGICAL CHEMISTRY  14291   b  y  g  u  e  s  t   ,  o n A  u  g  u  s  t  2 1  , 2  0 1 1 w w  j   b  c .  o r  g D  o l   o  a  d  e  d f  r  o  

Upload: james

Post on 02-Jun-2018

216 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: J. Biol. Chem.-2007-Pandey-14291-9

8/10/2019 J. Biol. Chem.-2007-Pandey-14291-9

http://slidepdf.com/reader/full/j-biol-chem-2007-pandey-14291-9 1/9

Reduction of LowMolecular Weight Protein-tyrosinePhosphatase Expression Improves Hyperglycemiaand Insulin Sensitivity inObeseMice*Received forpublication,October12, 2006, and in revised form, March9, 2007   Published, JBC Papers in Press,March 12, 2007, DOI 10.1074/jbc.M609626200

SanjayK. Pandey‡1, XingXian Yu‡, LynnettaM.Watts‡, M. DodsonMichael§, KyleW.Sloop§, Amber R. Rivard‡,ThomasA. Leedom‡, Vara PrasadManchem‡, LauraSamadzadeh‡, RobertA.McKay‡, Brett P.Monia‡,andSanjay Bhanot‡

From the‡Metabolic Disease Program, Antisense Drug Discovery, Isis Pharmaceuticals, Carlsbad, CA 92008,and §Endocrine Discovery, Lilly Research Laboratories, Indianapolis, Indiana 46285

To investigate the role of low molecular weight protein-

tyrosine phosphatase (LMW-PTP) in glucose metabolism

and insulin action, a specific antisense oligonucleotide (ASO)

 was used to reduce its expression both   in vitro  and   in vivo.

Reduction of LMW-PTPexpression with the ASO in cultured

mouse hepatocytes and in liver and fat tissues of diet-induced

obese (DIO) mice and  ob/ob mice led to increased phospho-

rylation and activity of key insulin signaling intermediates,

including insulin receptor-   subunit, phosphatidylinositol

3-kinase, and Akt in response to insulin stimulation. The

ASO-treated DIO and ob/ob animals showed improved insu-

lin sensitivity, which was reflected by a lowering of both

plasma insulin and glucose levels and improved glucose and

insulin tolerance in DIO mice. The treatment did not

decrease body weight or increase metabolic rate. These data

demonstrate that LMW-PTP is a key negative regulator of 

insulin action and a potential novel targetfor thetreatmentof 

insulin resistance and type 2 diabetes.

The incidence of diabetes has been steadily increasing and

hasbecome a major publichealth concern. Over 85% of diabeticpatients have type 2 diabetes. Obesity, which can result from asedentary life style andhigh caloriediet,is a major risk factorforthe development of this disorder (1). A hallmark of type 2 dia-

betes is insulin resistance, characterized by a decreased insulinresponse in a variety of tissues (2), including liver, fat, and mus-cle. Therefore, increasing insulin sensitivity is a practical strat-egy for the treatment of type 2 diabetes.

Insulin initiates its physiological response by binding to its

membrane-bound receptor (IR, a   22-heterotetramer pro-

tein),2 which causes autophosphorylation of the -subunit and

receptor activation, resulting in subsequent phosphorylation of its two major downstream substrates, IRS-1 and IRS-2 (3–5).Phosphorylated IRS-1 and -2 interact with and activate otherSH2 domain-containing adapter molecules such as NCK2,

Grb2, Shc, Syp (4– 8), and the regulatory subunit (p85) of phos-

phatidylinositol 3-kinase (PI3-K) (9, 10). Activated PI3-K stim-ulates Akt (or protein kinase B) that in turn phosphorylates andinactivates glycogen synthase kinase-3 (11), resulting in activa-

tion of glycogen synthase (12), thereby increasing the utiliza-tion of glucose for glycogen synthesis. In fat and muscle, acti- vation of this pathway also causes transfer of GLUT4 from thecytoplasm to the cell membrane, resulting in increased glucoseuptake (13). Therefore, IR-IRS-1/2-PI3-K-Akt signaling cas-

cade is a key pathway in mediating the effects of insulin actionon blood glucose levels.

A number of studies have established a role for intracellularphosphatases in the negative regulation of insulin signaling(14–16) such as protein-tyrosine phosphatase (PTP) 1B that

negatively regulates insulin action through dephosphorylatingtyrosine-phosphorylated IR (15, 16). Insulin sensitivity wasenhanced in PTP1B knock-out mice, in which increased tyro-sine phosphorylation of IR was found in liver and muscle butnot in adipose tissue. Insulin sensitivity was also increased in

ob/ob  and  db/db  mice treated with antisense inhibitors thatreduced PTP1B expression in liver and fat (17, 18). Further-more, in these mice insulin signaling was enhanced in liver butnot in fat tissue. Therefore, PTP1B negatively regulates insulin

signaling with tissue specificity (15, 16). In addition, PTEN andSHIP2 also negatively regulate insulin signaling   in vivo   viadephosphorylating phosphatidylinositol 3,4,5-trisphosphate,which results in decreased phosphorylation and inactivation of 

Akt. Enhanced insulin sensitivity was observed in  ob/ob  anddb/db   mice after antisense reduction of PTEN expressionwhich was accompanied by increased levels of phosphorylatedAkt at serine 473 (AktSer473) in liver (14). Increased phospho-rylated AktSer473 levels in both liver and muscle were also found

in SHIP2 knock-out mice (19). Moreover, protection from diet-induced insulin resistance was observed in these knock-outmice when feda high fatdiet (19). Therefore,intracellularphos-phatases play a key role in insulin signaling activity and in mod-ulating insulin sensitivity.

* The costs of publication of this article were defrayed in part by the paymentof page charges. This article must therefore be hereby marked “advertise-ment ” in accordancewith18 U.S.C.Section1734solely to indicate this fact.

1 To whom correspondence should be addressed: Metabolic Disease Pro-gram, Antisense Drug Discovery, Isis Pharmaceuticals, 1896 RutherfordRd., Carlsbad, CA 92008. Tel.: 760-603-2361; Fax: 760-603-3862; E-mail:[email protected].

2 The abbreviations used are: IR, insulin receptor; IRS, insulin receptor sub-strate; LMW, low molecular weight; PTP, protein-tyrosine phosphatase;PI3-K, phosphatidylinositol 3-kinase; ASO, antisense oligonucleotide; DIO,diet-induced obese; GTT,glucose tolerancetest; ITT,insulin tolerancetest;DGAT, acyl-CoA:diacylglycerol acyltransferase; PTEN, phosphatase andtensin homolog deleted on chromosome ten; SHP2/SHPTP2, Src homol-

ogyregion 2-domainphosphatase-2; AktSer473, Aktat serine473;BW,bodyweight; SHIP-2, SH2-containing inositol phosphatase 2.

 THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 282, NO. 19, pp. 14 291–14299, May 11, 2007© 2007 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in the U.S.A.

MAY 11, 2007• VOLUME 282 • NUMBER 19   JOURNAL OF BIOLOGICAL CHEMISTRY    14291

Page 2: J. Biol. Chem.-2007-Pandey-14291-9

8/10/2019 J. Biol. Chem.-2007-Pandey-14291-9

http://slidepdf.com/reader/full/j-biol-chem-2007-pandey-14291-9 2/9

A distinct PTP called low molecular weight PTP (LMW-PTP), also known as acid phosphatase locus 1, is an 18-kDa

cytosolic enzyme with two catalytically active isoforms, namedA and B, derived from alternate splicing of a single pre-mRNA(20, 21). It is widely expressed in various tissues in mammals(20, 21), and gene expression analysis from adult mouse tissuesfound that the level of LMW-PTP mRNA is highest in liver and

brain and lowest in skeletal muscle (22). Biochemical studieshave shown that LMW-PTP can dephosphorylate tyrosine-phosphorylated proteins (23, 24). Limited in vitro studies havesuggested a role for LMW-PTP as a negative regulator of insu-

lin-mediated mitogenic and metabolic signaling (23, 25). Inaddition, epidemiological studies have suggested that LMW-PTP levels are associated with dyslipidemia and hyperglycemiain human subjects (26, 27). However, the precise role of LMW-PTP in regulating insulin action  in vivo remains unknown.

Here, we have used a specific antisense oligonucleotide(ASO) to suppress the expression of LMW-PTP in culturedhepatocytes and in liver and fat of diet-induced obese and lep-tin-deficient obese mice to investigate the mechanism of the

insulin-sensitizing effect of LMW-PTP. Previous pharmacoki-netic studies have found that the specific ASO chemistry employed in this study leads to good distribution (and conse-quent activity) in a variety of tissues  in vivo, including liver andadipose tissue (28, 29). Reduction in the levels of both LMW-PTP isoforms in vitro  and  in vivo  increased tyrosine phosphor-

 ylation of IR and AktSer473 and increased IRS-1- and IRS-2-associated PI3-K activities in both liver and fat. The enhancedinsulin signaling activity was accompanied by a reduction inblood glucose and insulin levels and improved insulin and glu-

cose tolerance in obese mice. These results demonstrate for thefirst time a novel role for LMW-PTP as a key negative regulatorof insulin signaling in vivo.

EXPERIMENTAL PROCEDURES

Selection of LMW-PTP ASOs—Rapid throughput screens of about 80 ASOs against LMW-PTP were performed in A549cells, and the reduction of target gene expression was analyzedwith real time quantitative reverse transcription-PCR after

transfection of the cells with different concentrations of theASOs for 24 h. Based on IC50  values, four potent ASOs wereselected,and their in vivo activity wasconfirmed in lean C57BL/6J-Lepob/ mice. The final selection of the ASOs was basedupon the maximal reduction of hepatic LMW-PTP mRNA lev-

els in lean mice without any evidence of overt toxicity. The

selected LMW-PTP ASO (ASO 1), ISIS 288267, was then usedfor the current study. Another LMW-PTP ASO (ASO 2), ISIS288291,was also used in some in vitro experiments for compar-ison. All ASOs have a uniform phosphorothioate backbone

with a 20-base chimeric design, with 2-O-(methoxy)-ethylmodification in the first five and last five bases. The modifi-cation enhances their binding affinity to complimentary sequences and their resistance to the action of nucleases. Anegative control ASO (ISIS 141923), which has the same chem-

ical composition as ISIS 288267 but no complementarities toany known gene sequence, was also included in the study.

 Isolation and Transfection of Primary Hepatocytes—Mouseprimary hepatocytes were prepared according to a method

described earlier (29). Briefly, the mice were anesthetized by Avertin, and the livers were perfused through portal vein with a

buffer containing Ca2/Mg2-free Hanks’ balanced salt solu-tion (Invitrogen), 10 mM Hepes, and 0.5 mM EGTA, pH 7.4, for4 min. Then it was perfused with digestion buffer (William’s Emedium, 10 mM Hepes, 2 mM glutamic acid, 0.63 mg/ml colla-genase B, and 0.01 mg/ml gentamycin) for 6–8 min. Perfused

livers were detached from the animal, and the liver cells weregently dissociated and dispersed in a wash buffer (the digestionbuffer without collagenase but with 10% fetal bovine serum).The cell suspension was then centrifuged at 450–500 rpm in a

CR412 centrifuge (Jouan, Winchester, VA) to separate paren-chymal cells from nonparenchymal cells, and the former werewashed twice with cold phosphate-buffered saline. The paren-chymal cells were then plated and maintained in William’s Emedium supplemented with 10% fetal bovine serum and 1 nM

insulin at 37 °C under 95% O2,5% CO2. After overnight culturethe cells were washed with phosphate-buffered saline and incu-bated with 1–2 ml of transfection mixture containing 150 nM

ASO (control or LMW-PTP ASO), 4.5   g of Lipofectin

(Invitrogen) in William’s E medium for 4–6 h. Then the cellswere switched to a complete medium containing William’s Eand 10% fetal bovine serum for an addition 16–18 h for mRNAanalysis or 48 –55 h for protein analysis. For insulin signaling-related protein analysis, thehepatocyteswere incubatedin Wil-

liam’s E medium containing only 0.1% bovine serum albuminovernight prior to challenge with 10 nM insulin.

 Mice Care and Treatments—Animal care and other proce-dures were the same as described previously (29). In brief, maleC57BL/6J mice and C57BL/6J-Lepob/ob (ob/ob) mice were pur-

chased from the Jackson Laboratory at 6–7 weeks of age andmaintained with 12-h light/dark cycle with free access to foodand water. C57BL/6J mice were fed a diet containing 60 kcal %fat (research diet D12492; Research Diets, New Brunswick, NJ)for 12 weeks to induce obesity and insulin resistance. The ani-

mals were then divided into different groups based on BW,blood glucose, and insulin concentrations and treated (subcu-taneous injection) with LMW-PTP ASO or control ASO (dis-solved in saline) at a dose of 25 mg/kg BW or with a similar volume of saline, twice a week for 6 weeks. A group of lean

C57BL/6J mice fed normal rodent chow and injected withsaline served as normal controls. At the end of the study, themice were sacrificed, blood was collected through cardiacpuncture, and different tissues were dissected and quickly fro-

zen in liquid N2 until further analysis.

For the ob/ob mice study, after acclimation to the animal carefacility for 4–5 days, the mice were grouped based on BW andblood glucose levels and injected with LMW-PTP ASO at 25mg/kg BW or a similar volume of saline twice a week for 4

weeks. Because the negative control ASO treatment did notexhibit specific metabolic effects as compared with saline treat-ment in the DIO mice study, it was not included in the  ob/obmice study.

For theinsulin challenge studies, DIO mice were treated with

LMW-PTP ASO or control ASO at a dose of 50 mg/kg BW orwith a similar volume of saline, twice a week for 2 weeks, andob/ob mice were treated with LMW-PTP ASO at 25 mg/kg BW or saline twice a week for 4 weeks. The animals were then fasted

Reductionof LMW-PTP Improves InsulinSensitivity 

14292   JOURNAL OF BIOLOGICAL CHEMISTRY    VOLUME 282• NUMBER 19• MAY 11, 2007

Page 3: J. Biol. Chem.-2007-Pandey-14291-9

8/10/2019 J. Biol. Chem.-2007-Pandey-14291-9

http://slidepdf.com/reader/full/j-biol-chem-2007-pandey-14291-9 3/9

overnight and given a bolus intraperitoneal injection of insulinat 2 units/kg of BW or vehicle. Ten minutes later, the animals

were sacrificed, and liver and epididymal fat were collected andquickly frozen in liquid N2 for further analysis. All of the aboveexperiments were performed in accordance with InstitutionalAnimal Care and Use Committee guidelines.

 Metabolic Rate Measurement —After 3.5 weeks of treatment,

metabolic rate in ob/ob mice was measured using indirect cal-orimetry (Oxymax System; Columbus Instruments, Columbus,OH). The animals were acclimated to metabolic chambers for24 h before initiation of the measurement. For each treatment

group, five animals were measured for a 24-h period. Biochemical Analysis—Plasma insulin levels were measured

with a commercial enzyme-linked immunosorbent assay kit(ALPCO diagnostics, manufactured by Mercodia) according tothe manufacturer’s instructions. Plasma glucose, triglycerides,

cholesterol, aspartate transaminase, and alanine transaminaselevels were measured with an Olympus Analyzer AU400(Melville, NY).

Gene Expression Analysis—For gene expression analysis,

total RNA from hepatocytes was isolated by using a QiagenRNAeasy kit, andtotal RNAfrom animal tissues wasisolatedby homogenizing tissues in RLT buffer (Qiagen) followed by cen-trifugation with cesium chloride gradient. Real time quantita-tive reverse transcription-PCR was performed with custom-made reverse transcription-PCR enzymes and reagents kit

(Invitrogen), primer and probe sets designed with PrimerExpress Software (PE Applied Bioscience, Foster City, CA), andan ABIprism 7700sequence detector (PE Applied Biosciences).For the analysis, 100ng of total RNA was used for each reaction.

Each sample was run in duplicate or triplicate, and the mean values were used to calculate the mRNA levels and gene expres-sion. The expression was normalized with the amount of totalRNA loaded that was determined with a RiboGreen assay.

 Insulin and Glucose Tolerance Tests (ITT and GTT)—ITT

andGTTinDIOmicewereconductedafter4.5and5.5weeksof treatment, respectively. The mice were fasted for 4 h prior toITT and overnight prior to GTT. Blood glucose levels weremeasured before insulin or glucose injection (0 min, base-line values), and the animals were then administered (intraperito-

neally) insulin (R-insulin; Lilly Research Laboratories, Indian-apolis, IN) at a dose of 0.5 unit/kg BW for ITT or glucose at 1.0g/kg of BW. Blood glucose was then measured at 30,60, 90, and120 min after insulin or glucose injection using a Glucometer(Abbott Laboratories, Bedford, MA).

 Histological Analysis—Liver samples were fixed in 10% buff-ered formalin and embedded in paraffin wax for staining withhematoxylin and eosin or directly embedded in an optimal tem-perature embedding medium followed by immediate snap-

freezing in liquid N2   for oil-red O staining. Multiple adjacent6-m sections were cut and mounted on glass slides and thenstained. Images of the histological sections were analyzed.

Western Blot Analysis—The cells were lysed, or tissues werehomogenized in a lysis buffer (150 mM NaCl, 50 mM Tris, pH

7.5, 1% Triton X-100, 0.5% Nonidet P-40, 0.25% sodium deoxy-cholate, 1 mM EDTA, 1 mM EGTA, 1 mM NaOV, 1 mM NaF)containing protease inhibitor mixture I (Calbiochem). Thelysates were cleared by centrifugation for 15 min at 12,000  g .

Equal amounts of total protein for different samples were sep-arated on 10% or 16% SDS-PAGE gels under reduced condi-

tionsand then transferred onto polyvinylidene difluoride mem-branes. The blots were then incubated with antibody againstIR- subunit (C-19; Santa Cruz, CA), tyrosine-phosphorylatedproteins (4G10; Upstate Biotechnology Inc., Lake Placid, NY)or Ser473-phosphorylated Akt (Cell Signaling, Danvers, MA).

The blots were also incubated with antibody against PTP1B,SHP2/SHPTP2 (from Upstate Biotechnology Inc.), PTEN (CellSignaling, Danvers, MA and Upstate Biotechnology Inc.), orLMW-PTP antibody (kindly provided by Dr. Thomas Mustelin

of Burnham Institute, La Jolla, CA). The signals were detectedby using horseradish peroxidase-conjugated goat anti-rabbitIgG antibody and ECL detection reagents.

 Immunoprecipitation—The clarified lysates were first incu-bated with protein agarose A/G beads (1:1 ratio) for 3–4 h at

4 °C followed by incubation with anti-phosphotyrosine anti-body for another 3–4 h at 4 °C. The immunocomplex waswashed with lysis buffer three times and then boiled inLaemmli’s sample buffer and Western blotted with anti-IR-

subunit antibody (Santa Cruz Biotechnology, Santa Cruz, CA)as described earlier.

 PI 3-Kinase Activity Assay—The PI3-K activity wasmeasuredas described previously (30). Briefly, theclarifiedlysates (500gof protein) were subjected to immunoprecipitation with 1g of 

IRS-1 or IRS-2 antibody for 2 h at 4 °C, followed by incubationwith protein A/G-Sepharose for an additional 2 h. The immu-nocomplexes were then washed and used for PI3-K activity assay using   L--phosphatidylinositol and [ -32P]ATP as sub-strates. The reaction was stopped by the addition of a mix of 

CHCl3:CH3OH:HCl (100:200:2). The produced phosphoryla-ted lipid was extracted with CHCl3  and separated on a TLCplate. The plate was then exposed to a Kodak film. The radio-activity associated with PIP3   spots was quantified by ImageQuant, or the PIP3   spots were scratched off the plates and

counted in scintillation counter.Statistical Analysis—The values are expressed as the

means S.E. of at least three to five  in vitro  or five  in vivoindependent measurements per treatment. Statistical differ-ence across treatment groups was determined using Stu-

dent’s t  test or one-way analysis of variance with Tukey HSDmultiple comparisons. Differences were considered signifi-cant at p 0.05.

RESULTS

 Reduction of LMW-PTP Expression Enhances Insulin Signal-ing in Mouse Hepatocytes—To investigate the role of LMW-PTP in insulin signaling, mouse primary hepatocytes weretransfected with LMW-PTP ASO or vehicle. LMW-PTP ASO

treatment reduced LMW-PTP mRNA levels by 90% (Fig. 1 A),which was associated with dramatic reduction in the proteinlevels for both A and B isoforms (Fig. 1 B). Reprobing the mem-brane with anti-PTP1B antibody revealed no compensatory increase in PTP1B protein levels in the ASO-treated cells (Fig.

1 B). We next examined the effect of LMW-PTP reduction onthe insulin activation of IR and Akt, two critical members of theinsulin signaling cascade. LMW-PTP ASO or control ASOtransfected mouse primary hepatocytes were incubated with

Reductionof LMW-PTP Improves InsulinSensitivity 

MAY 11, 2007• VOLUME 282 • NUMBER 19   JOURNAL OF BIOLOGICAL CHEMISTRY    14293

Page 4: J. Biol. Chem.-2007-Pandey-14291-9

8/10/2019 J. Biol. Chem.-2007-Pandey-14291-9

http://slidepdf.com/reader/full/j-biol-chem-2007-pandey-14291-9 4/9

insulin (10 nM) for 10 min, and cell lysates were prepared forWestern blot analysis. Tyrosine phosphorylation levels of IRsubunit (pY-IR; Fig. 1C ) and Ser473 phosphorylation levels of Akt (pAktSer473; Fig. 1 D) were higher in LMW-PTP ASO-

treated cells than in control ASO-treated groups after insu-lin stimulation. Under these conditions there was no changein the total protein levels of either IR subunit (Fig. 1C ) orAkt (Fig. 1 D).

To further confirm that these insulin enhancing effects

were secondary to a specific antisense reduction of LMW-PTP, we evaluated the effect of second ASO (ASO 2) that wastargeted to a different region of the LMW-PTP mRNA. ASO2 reduced LMW-PTP expression to a similar extent as com-

pared to ASO 1 and also enhanced thephosphorylation levelsof AktSer473 to a similar extent upon insulin stimulation (Fig. 1 D,middle panel ). These data suggest that reduction of LMW-PTPexpression sensitized the response of the hepatocytes to insulinstimulation without any compensation by PTP1B.

Suppression of LMW-PTP Improves Insulin Sensitivityin DIO Mice with No Change in Body Weight —To ascertain the role of LMW-PTP in insulin action in vivo, we suppressed its expres-sion with the ASO in high fat diet-induced obese (DIO) mice.

After being fed a 60 kcal % fat dietfor 12 weeks, the mice were treated

with saline or a control ASO orLMW-PTP ASO (dissolved insaline) at a dose of 25 mg/kg of BW twice a week for 6 weeks. LMW-PTP ASO treatment reduced

LMW-PTP mRNA levels by about90% in liver and 75% in fat as com-pared with controls (Fig. 2 A), whichresulted in a significant reduction in

both tissues of LMW-PTP proteinlevels (Fig. 2 B). As was observed inhepatocytes, ASO-mediated reduc-tion of LMW-PTP did not cause acompensatory increase in PTP1B

protein levels in either tissue (Fig.2 B). Furthermore, we examined theexpression of SHP2/SHPTP2 andPTEN; two well known phosphata-

ses that play an important role inglucose homeostasis (14, 19). West-ern blot analysis revealed that therewas no significant difference in theprotein levels of both these phos-phatases between LMW-PTP ASO

treatment group and saline or con-trol ASO treatment group in liveror fat tissues of DIO mice (Fig. 2 B,top  and  middle panels). Note that

we observed two strong bands forSHP2/SHPTP2 in the liver tissuesfrom both DIO and   ob/ob   mice(Figs. 2 B   and 3 B,   bottom panels,respectively) but one band for

SHP2/SHPTP2 from the fat tissuesof either animal model (Figs. 2 B   and 3 B,   bottom panels,respectively).

As expected, high fat feeding resulted in increased plasmainsulin and glucose levels in the mice. Treatment with LMW-

PTP ASO lowered both insulin and glucose levels as comparedwith the controls (Fig. 2C ). In fact, both insulin and glucoselevels in LMW-PTP ASO-treated mice were similar to thoseseen in chow-fed mice after 3.5 and 4.5 weeks of treatment,respectively (Fig. 2C ). These results suggest that reduced

LMW-PTP expression following ASO treatment increasedinsulin sensitivity. This was further confirmed by GTT and ITTconducted in these mice, in which glucose excursions after glu-cose or insulin challenge were significantly lower in LMW-PTP

ASO-treated animals relative to saline or control ASO-treatedmice (Fig. 2 D).

To further address the mechanisms underlying the effects of LMW-PTP inhibition in DIO mice, mRNA levels of variousgenes involved in glucose and lipid metabolism were deter-

mined. The analysis demonstrated that LMW-PTP treatmentcaused a significant reduction in the levels of glucose-6-phos-phatase mRNA, which could in part contribute to the positiveeffects on glucose metabolism (Fig. 2 E ). Taken together, these

FIGURE 1. Suppression of LMW-PTP expression by LMW-PTP ASO leads to increased insulin signalingactivityin mouse primary hepatocytes. Transfection with LMW-PTP ASO suppressed LMW-PTP mRNA levelsby more than 90% in mouse primary hepatocytes ( A), which caused marked reduction in the protein levels of LMW-PTPbut notPTP-1B(B). Challengewith insulin demonstrated thatthe increase in bothtyrosinephospho-rylationof IR subunits (pY-IR; C )andSer473 phosphorylation of Akt(AktSer473; D) wasmuch greater inthe cellstreated with the LMW-PTP ASO  versus   those treated with control ASO. The data and representative blotspresented were from three to five experiments with duplicate or triplicate for each treatment. *, p 0.05;**, p 0.01. IP , immunoprecipitation; WB, Western blot; MW , molecular weight; Cont , control; Ins, insulin.

Reductionof LMW-PTP Improves InsulinSensitivity 

14294   JOURNAL OF BIOLOGICAL CHEMISTRY    VOLUME 282• NUMBER 19• MAY 11, 2007

Page 5: J. Biol. Chem.-2007-Pandey-14291-9

8/10/2019 J. Biol. Chem.-2007-Pandey-14291-9

http://slidepdf.com/reader/full/j-biol-chem-2007-pandey-14291-9 5/9

data indicate that suppression of LMW-PTP with a specificASO improved insulin sensitivity in DIO mice.

In addition, treatment with LMW-PTP ASO resulted in sig-nificant lowering of plasma cholesterol levels (Table 1) and livertriglyceride content (34.5 4.5 mg/g versus 55.6 8.0 mg/g in

control ASO group  versus  66.3 8.4 mg/g of tissue in salinegroup; p 0.01 versus either group). However, ASO treatmentdid not affect BW (Table 1). Gene expression analysis foundthat LMW-PTP ASO treatment did not cause changes in the

expression of hepatic lipogenicgenes, including fatty acid synthase,

stearoyl-CoA desaturase 1, dia-cylgycerol acyltransferase (DGAT)2, or the genes related to lipid home-ostasis in fat, including hormone-sensitive lipase, fatty acid synthase,

DGAT1, and DGAT2 (Fig. 2 E ). Fur-thermore, ASO treatment was welltolerated, and no evidence of livertoxicity as determined by plasma

aspartate transaminase and alaninetransaminase activities (Table 1)was observed.

Suppression of LMW-PTP Also Improves Insulin Sensitivity in ob/ob

 Mice with No Change in BodyWeight —To extend our investiga-tion on the role of LMW-PTP ininsulin action and glucose metabo-

lism, LMW-PTP ASO was used tosuppress its expression in a geneti-cally obese mouse model, and theresultant metabolic effects werestudied. For this purpose, leptin-de-ficient ob/ob mice were treated with

LMW-PTP ASO at a dose of 25mg/kg or a similar volume of salinetwice a week for 4 weeks. ASO treat-ment resulted in a reduction of 

LMW-PTP mRNA levels by  90%in liver and 60% in fat (Fig. 3 A).Western blot analysis of liver sam-ples demonstrated that both iso-forms of LMW-PTP protein were

reduced by more than 75%  versuscontrols (Fig. 3 B). A significantreduction in LMW-PTP proteinlevels of both the isoforms werealso found in the fat samples from

LMW-PTP ASO-treated mice(Fig. 3 B). Again, no significantcompensatory increase in PTP1B,SHP2/SHPTP2 or PTEN (data notshown) protein levels were ob-

served (Fig. 3 B).LMW-PTP ASO treatment im-

proved insulin sensitivity in   ob/obmice. After 4 weeks of treatment

both fedand fastedplasma glucose levelswere markedly low-ered in ASO-treated mice than in the controls (Fig. 3, C  and D). Furthermore, LMW-PTP ASO treatment resulted inapproximately a 50% reduction in plasma insulin levels ver- sus  controls (Fig. 3 E ). These data demonstrate that suppres-

sion of LMW-PTP with ASO also improved insulin sensitiv-ity in ob/ob mice.

LMW-PTP ASO treatment also improved hepatic steatosisin  ob/ob  mice. Histological examination of hematoxylin and

FIGURE 2. Reduction of LMW-PTP expression by LMW-PTPASO improved insulin sensitivity in DIO mice. Treatment with LMW-PTP ASO reduced LMW-PTP mRNA levels by about 90 and 75% in liver and fat, respec-tively ( A), whichresultedin dramaticreduction in LMW-PTPprotein levels (B) butno effecton PTP1B,SHP2, andPTEN protein levels in both tissues as compared with thecontrols(B). TheASO treatment reduced fedplasmainsulin and glucose levels (C ), improved glucose (IPGTT ) and insulin tolerance (ITT ; D), decreased hepatic glu-cose-6-phosphatase gene expression but did not affect the expression of the key lipogenic genes (fatty acid

synthase, stearoyl-CoA desaturase 1, and DGAT2) in liver or the genes (hormone-sensitive lipase ( HSL), fattyacid synthase (FAS), DGAT1, and DGAT2) related to lipid homeostasis in fat ( E ). The data and representativeblots presentedwere from four to six mice foreach treatmentgroup. *, p 0.05; **, p 0.01 versus either DIOcontrol group. Cont , control; WAT , white adipose tissue.

Reductionof LMW-PTP Improves InsulinSensitivity 

MAY 11, 2007• VOLUME 282 • NUMBER 19   JOURNAL OF BIOLOGICAL CHEMISTRY    14295

Page 6: J. Biol. Chem.-2007-Pandey-14291-9

8/10/2019 J. Biol. Chem.-2007-Pandey-14291-9

http://slidepdf.com/reader/full/j-biol-chem-2007-pandey-14291-9 6/9

eosin-stained liver sections showed that there was a large num-ber of multilobular and unilobular cytoplasmic lipid droplets incontrol animals. In contrast, the lipid droplets were greatly reduced in both size and number as a result of LMW-PTP ASO

treatment (Fig. 3 F ). This finding was further confirmed withoil-red O staining in which the intense oil-red O-stained lipidspots found in saline-treated mice were markedly reduced inLMW-PTP ASO-treated mice (Fig. 3 F ). This improvement inliver steatosis was accompanied by an improvement in liver

function, as demonstrated by a decrease in plasma aspartatetransaminase and alanine transaminase levels in these mice(Table 1). The treatment lowered plasma triglyceride levels butnot cholesterol levels (Table 1). However, the ASO treatment

did not cause changes in either BW or metabolic rate as deter-mined by a lack of effect on both VO2 (Table 1) and respiratory quotient (data no shown).

Suppression of LMW-PTP ASO Improves Insulin Action in

 Both Liver and Fat —To investigate the possible mechanismsunderlying the improved insulin sensitivity after LMW-PTPASO treatment, both DIO and ob/ob mice treated with LMW-PTP ASO were challenged with insulin, and the activities of thekey insulin signaling enzymes were examined in both liver and

fat. Western blot analysis found that treatmentwith LMW-PTPASO caused a greater increase in pY-IR levels versus controlsupon insulin stimulation in liver both in DIO (Fig. 4 A) andob/ob (Fig. 5 A) mice and in fat in  ob/ob mice (data not shown)without affecting the total protein levels of IR subunit. LMW-

PTP ASO treatment significantly reduced the basal levels of pY-IR versus controls (Fig. 4C ). Injection of insulin increasedpY-IR   levels by over 5-fold  versus   the basal levels in ASO-treated mice (Fig. 4C ;  p    0.001), whereas it only increased

pY-IR by about 1.2-fold in control mice (Fig. 4C ; p 0.05). Adecreased basal pY-IR level in ASO-treated ob/ob mice versuscontrols was also observed (Fig. 5 A). The decreased basal pY-IR   levels probably were due to ASO-caused reduction of plasma insulin levels as shown in Figs. 2C   and 3 E . Taken

together, these data demonstrate increased response of the IR

to insulin after LMW-PTP ASO treatment.The activity of PI3-K, a key intermediate component of the

insulin signaling pathway, was also measured in  ob/ob  micetreated with LMW-PTP ASO. In liver, IRS-1-associated PI3-K

activity was found to be increased by  2.5-fold in the ASO-treated mice upon insulin administration (Fig. 5 B), which wasin sharp contrast to saline-treated animals in which insulinstimulation did not cause any increase in activity (Fig. 5 B).IRS-2 associated PI3-K activity was enhanced by over 55% in

the liver of the ASO-treated animals but not in controls uponinsulin stimulation (Fig. 5C ). Increased IRS-1 associated PI3-Kactivity (60%) was also observed in fat from LMW-PTP ASO-treated ob/ob  mice, but not in fat from saline-treated mice

FIGURE 3.   Suppression of LMW-PTP expression by LMW-PTP ASOimproved insulin sensitivity inob/ob mice. Treatment with LMW-PTP ASOreducedLMW-PTPmRNAlevelsbyabout90%inliverand60%infat( A),whichresultedin similar degrees of reductionin the LMW-PTPproteinlevels (bothAand B isoforms) in liver and fat, respectively, without any compensatorychanges in PTP1B, SHP2, and PTEN expression in these tissues (B). ASO treat-ment caused reduced fed (C ) and fasted (D) plasma glucose levels as well asinsulin levels (E ). The ASO treatment improved liver steatosis, as demon-strated by hematoxylin andeosinstainingand oil-red O staining (F ). Thedataandrepresentative blotspresented werefrom at leastfive micefor each treat-ment group. *,  p 0.05; **,  p 0.01  versus  controls.  WAT , white adiposetissue; H & E , hematoxylin and eosin.

TABLE 1

Bodyweight, plasma cholesterol, triglyceride levels, transaminaseactivities, andmetabolic rate inmiceThe data are expressed as the means S.E. Chol, plasma cholesterol; TG, triglyc-eride; AST, aspartatetransaminase; ALT, alanine transaminase;VO

2, O

2consump-

tion rate; BW, body weight.

Saline  Control

ASOLMW-PTP

ASO

DIO mice (n 6)

BW (g), 0 weeks 39.8 1.3 39.4 0.6 39.8 0.7BW (g), 5 weeks 39.9 1.4 39.2 1.0 38.4 0.4Chol (mg/dl), 6 weeks 183.7 6.7 181.4 7.3 135.8 9.2a

AST (IU/L) 40.7 5.6 39.4 4.9 36.3 2.1ALT (IU/L) 62.4 7.3 64.1 4.3 63.9 3.3

ob/ob mice (n 5)BW (g), 0 weeks 36.3 0.4 38.4 0.8BW (g), 5 weeks 54.5 0.8 55.9 0.9Chol (mg/dl), 4 weeks 182.9 61.0 189.7 47.3TG (mg/dl), 4 weeks 168.0 3.1 75.0 5.4b

AST (IU/L) 167.0 9.0 111.0 7.0ALT (IU/L) 264.0 20.0 198.0 16.0VO

2(ml/kg0.75/h)

In dark 921.9 25.5 906.2 25.0In light 875.7 52.0 838.5 22.2

a p 0.05 versus control.b p 0.01 versus control.

Reductionof LMW-PTP Improves InsulinSensitivity 

14296   JOURNAL OF BIOLOGICAL CHEMISTRY    VOLUME 282• NUMBER 19• MAY 11, 2007

Page 7: J. Biol. Chem.-2007-Pandey-14291-9

8/10/2019 J. Biol. Chem.-2007-Pandey-14291-9

http://slidepdf.com/reader/full/j-biol-chem-2007-pandey-14291-9 7/9

Page 8: J. Biol. Chem.-2007-Pandey-14291-9

8/10/2019 J. Biol. Chem.-2007-Pandey-14291-9

http://slidepdf.com/reader/full/j-biol-chem-2007-pandey-14291-9 8/9

knock-out mice were resistant to BW gain on a high fat diet anddisplayed an increased metabolic rate (15, 16). The PTP1BASO-treated   ob/ob   mice had lowered lipogenesis with

decreased lipogenic gene expression in fat (17). SHIP2 knock-out micealsoshowed a similar phenotype to that seen in PTP1Bknock-out mice in this regard (19). However, in this study bothLMW-PTP ASO-treated DIO and ob/ob mice showed no sig-nificant difference in BW from the controls. Metabolic rate

measurement in  ob/ob  mice did not reveal any difference ineither O2  consumption rate or respiratory quotient betweentreatment groups. Gene expression analysis also failed to iden-tify differences in the expression of key lipogenic genes in liver

or the genes related to lipid homeostasis in fat among the DIO

groups. Therefore, these two intracellular phosphatases play distinct roles in regulating metabolism in the body.

Nonalcoholic steatohepatitis can result from chronic accu-mulation of fat in the liver. Epidemiological studies suggest that

increased LMW-PTP activity is associated with high levels of serum triglycerides in obese and diabetic subjects where inci-dence of fatty liver and nonalcoholic steatohepatitis is a com-mon occurrence (26). In this study, a significant decrease inliver triglyceride levels in DIO mice and an improvement in

liver steatosis in   ob/ob   mice following ASO treatment wasobserved. This observation, coupled with unchanged expres-sion of the key hepatic lipogenic genes, suggests increasedhepatic fatty acid oxidation in these ASO-treated mice. In addi-

tion, decreased plasma cholesterollevels in DIO mice and decreased

plasma triglyceride levels in  ob/obmice were also observed in thisstudy. These data suggest that, inaddition to a role in regulation of insulin action, LMW-PTP may also

be involved in regulation of lipidmetabolism. Further studies areneeded to delineate the mechanismof the lipid-lowering effects of LMW-PTP ASO in these mice.

Several phosphatases, includingPTP1B, LAR, SHIP2, and PTEN,have been reported to exert nega-tive regulation on insulin action

through their dephosphorylationaction on different components of the insulin signaling pathway (14 –16, 19, 31, 32). Here, using an anti-

sense approach, we demonstratethat LMW-PTP also plays a key role in modulating insulin sig-naling and insulin sensitivity   invivo. This effect of LMW-PTP wasdistinct and independent of 

PTP1B, SHP2/SHPTP2, or PTEN.Whether additional PTPs or otherphosphatases exist that areinvolved in regulation of severalcomponents of insulin signaling

activities under normal and dia-betic conditions remains to be determined.

In conclusion, the present study demonstrates for the firsttime that reduction of LMW-PTP protein levels in insulin-re-

sistant obese animals improves insulin action and glucosemetabolism by positively regulating insulin signaling pathway in liver and adipose tissue. Thus, LMW-PTP could be a noveldrug target for the treatment of type 2 diabetes.

 Acknowledgments—We thank Nunzio Bottini and Tom Mustelin for 

the LMW-PTP antibody; Sue Murray, Sheri Booten, and Sara Petok 

 for assistance in the studies; and Gene Hung for help in histological 

analysis.

REFERENCES

1. Bazzano, L. A., Serdula, M., and Liu, S. (2005)   J. Am. Coll. Nutr.   24,

310–319

2. Reaven, G. M. (1988) Diabetes 37, 1595–1607

3. Rosen, O. (1987) Science 237, 1452–1458

4. Myers, M. G., Jr., and White, M. F. (1993) Diabetes 42, 643–650

5. White, M. F., and Kahn, C. R. (1994) J. Biol. Chem. 269, 1– 4

6. Kuhne, M. R., Pawson, T., Lienhard, G. E., and Feng, S. E. (1993) J. Biol.

Chem. 268, 11479 –11481

7. Lee, C. H., Li,W., Nishimura, R.,Zhou, M.,Batzer, A. G.,Myers, M. G., Jr.,

White, M. F., Schlessinger, J., and Skolnik, E. Y. (1993) Proc. Natl. Acad.

Sci. U. S. A. 90, 11713–11717

8. Skolnik, E.Y., Lee, C. H., Batzer, A., Vicentini, L. M., Zhou, M., Daly, R.,

Myers, M. J., Jr., Backer, J. M., Ullrich, A., White, M. F., and Schlessinger,

FIGURE 5. Suppression of LMW-PTP expression with ASO leads to augmented insulin signaling in liverand fat inob/ob mice. After treatment with LMW-PTP ASOor saline in ob/ob micefor 4 weeks, the micewerefasted overnight and then challenged with an intraperitoneal bolus injection of saline or insulin. ASO treat-ment enhanced the levels of tyrosine-phosphorylated IR subunits (pY-IR) but not the total levels of IRsubunits in liver ( A; each lane represents three pooled samples) and fat (data not shown), which resulted inincreased IRS-1- and IRS-2-associated PI3-K activities in liver ( B and C ) and fat (D) and increased Ser473 phos-phorylation levels of Akt(pAktSer473)inliver(E ) andfat (datanot shown). Thedata andrepresentative figures forB–E were from samples ofat leastthreemiceforeachtreatmentwith repeatsof analysis.*, p0.05; **, p0.01.Cont , control; PIP 

3, phosphatidylinositol 1,4,5-trisphosphate.

Reductionof LMW-PTP Improves InsulinSensitivity 

14298   JOURNAL OF BIOLOGICAL CHEMISTRY    VOLUME 282• NUMBER 19• MAY 11, 2007

Page 9: J. Biol. Chem.-2007-Pandey-14291-9

8/10/2019 J. Biol. Chem.-2007-Pandey-14291-9

http://slidepdf.com/reader/full/j-biol-chem-2007-pandey-14291-9 9/9

J. (1993) EMBO J. 12, 1929–1936

9. Lavan, B. E., Kuhne, M. R., Garner, C. W., Anderson, D., Reedijk, M.,

Pawson, T., and Lienhard, G. E. (1992) J. Biol. Chem. 267, 11631–11636

10. Myers, M. G., Jr., Backer, M. G., Sun, X. J., Shoelson, S., Hu, P., Schless-

inger,J., Yoakim, M.,Schaffhausen,B., andWhite, M. F. (1992) Proc. Natl.

 Acad. Sci. U. S. A.  89, 10350 –10354

11. Cross, D. A., Alessi, D. R., Cohen, P., Andjelkovich, M., and Hemmings,

B. A. (1995) Nature 378, 785–789

12. Srivastava, A. K., and Pandey, S. K. (1998)   Mol. Cell. Biochem.   182,

135–141

13. Bryant, N.J., Govers, R.,and James,D. E. (2002) Nat. Rev. Mol. Cell. Biol. 3,

267–277

14. Butler, M.,McKay, R. A.,Popoff,I. J.,Gaarde, W. A.,Witchell, D., Murray,

S. F., Dean, N. M., Bhanot, S., and Monia, B. P. (2002)   Diabetes   51,

1028–1034

15. Elchebly, M., Payette, P., Michaliszyn, E., Cromlish, W., Collins, S., Loy,

A. L., Normandin, D., Cheng, A., Himms-Hagen, J., Chan, C. C., Ram-

achandran, C., Gresser, M. J., Tremblay, M. L., and Kennedy, B. P. (1999)

Science 283, 1544–1548

16. Klaman, L. D., Boss, O., Peroni, O. D., Kim, J. K., Martino, J. L., Zabolotny,

J. M.,Moghal, N.,Lubkin,M., Kim,Y. B.,Sharpe, A. H.,Stricker-Krongrad,

A., Shulman, G. I., Neel, B. J., and Kahn, B. B. (2000)  Mol. Cell. Biol.  20,

5479–5489

17. Gum, R. J., Gaede, L. L., Koterski, S. L., Heindel, M., Clampit, J. E., Zinker,

B. A., Trevillyan J. M., Ulrich, R. G., Jirousek, M. R., and Rondinone C. M.

(2003) Diabetes 52, 21–28

18. Zinker, B. A.,Rondinone, C. M.,Trevillyan,J. M.,Gum, R. J.,Clampit J. E.,

Waring, J. F., Xie, N., Wilcox, D., Jacobson, P., Frost, L., Kroeger, P. E.,

Reilly, R. M.,Koterski, S.,Opgenorth, T. J.,Ulrich,R. G.,Crosby,S., Butler,

M., Murray, S. F., McKay, R. A., Bhanot, S., Monia, B. P., and Jirousek,

M. R. (2002) Proc. Natl. Acad. Sci. U. S. A.  99, 11357–11362

19. Sleeman, M. W.,Wortley, K.E., Lai, K. M.,Gowen, L. C.,Kintner, J.,Kline,

W. O., Garcia, K., Stitt, T. N., Yancopoulos, G. D., Wiegand, S. J., and

Glass, D. J. (2005) Nat. Med. 11, 199–205

20. Ramponi,G., andStefani, M. (1997) Int. J. Biochem. Cell Biol. 29, 279–292

21. Bottini, N., Bottini, E., Gloria-Bottini, F., and Mustelin, T. (2002) Arch.

 Immunol. Ther. Exp. 50, 95–104

22. Magherini, F., Giannoni, E., Raugei, G., Cirri, P., Paoli, P., Modesti, A.,

Camici, G., and Ramponi, G. (1998) FEBS Lett. 437, 263–266

23. Chiarugi, P., Cirri, P., Marra, F., Raugei, G., Camici, G., Manao, G., and

Ramponi, G. (1997) Biochem. Biophys. Res. Commun. 18, 676–682

24. Chiarugi, P., Cirri, P., Taddei, M. L., Giannoni, E., Fiaschi, T., Buricchi, F.,

Camici, G., Raugei, G., and Ramponi, G. (2002)   J. Biol. Chem.   277,

37331–37338

25. Taddei, M. L., Chiarugi, P., Cirri, P., Talini, D., Camici, G., Manao, G.,

Raugei, G.,and Ramponi, G. (2000) Biochem. Biophys. Res. Commun. 270,

564–569

26. Bottini, N., MacMurray, J., Peters, W., Rostamkhani, M., and Comings,

D. E. (2002) Mol. Gen. Met. 77, 226–229

27. Iannaccone, U., Bergamaschi, A., Magrini, A., Marino, G., Bottini, N.,

Lucarelli, P., Bottini, E., and Gloria-Bottini, F. (2005)  Metabolism   54,

891–894

28. Dean, N. M., Butler, M., Monia, B. P., and Manoharan, M. (2001) in Anti-

 sense Drug Technology: Principles, Strategies and Applications (Crooke,

S. T., ed) pp. 319–338, Dekker, New York

29. Yu, X. X., Murray, S. F., Pandey, S. K., Booten, S. L., Bao, D., Song, X. Z.,

Kelly, S., Chen, S., McKay, R., Monia, B. P., and Bhanot, S. (2005)  Hepa-

tology 42, 362–371

30. Pandey, S. K., Anand-Srivastava, M. B., and Srivastava, A. K. (1998) Bio-

chemistry 37, 7006–7014

31. Byon, J. C., Kusari, A. B., and Kusari, J. (1998) Mol. Cell. Biochem.  182,

101–108

32. Goldstein, B. J., Ahmad, F., Ding, W., Li, P. M., and Zhang, W. R. (1998)

 Mol. Cell. Biochem.  182, 91–99

Reductionof LMW-PTP Improves InsulinSensitivity 

MAY 11, 2007• VOLUME 282 • NUMBER 19   JOURNAL OF BIOLOGICAL CHEMISTRY    14299