12
Immunological Characterization of Tristetraprolin as a Low Abundance, Inducible, Stable Cytosolic Protein* Received for publication, January 27, 2004, and in revised form, March 1, 2004 Published, JBC Papers in Press, March 9, 2004, DOI 10.1074/jbc.M400900200 Heping Cao‡, Jane S. Tuttle‡, and Perry J. Blackshear‡§From the Laboratory of Signal Transduction and Office of Clinical Research, NIEHS, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina 27709 and the §Departments of Biochemistry and Medicine, Duke University Medical Center, Durham, North Carolina 27710 Tristetraprolin (TTP) is a zinc finger protein that can bind to AU-rich elements within certain mRNAs, result- ing in deadenylation and destabilization of those mRNAs. Its physiological targets include the mRNAs en- coding the cytokines tumor necrosis factor (TNF) and granulocyte-macrophage colony-stimulating factor. TTP was originally identified on the basis of its massive but transient increase in mRNA levels following mitogen stimulation of fibroblasts. It has been difficult to recon- cile this transient mRNA profile with the presumed con- tinuing “need” for TTP protein, for example, to reverse the effects of lipopolysaccharide (LPS)-stimulated TNF secretion. To investigate this and other questions con- cerning endogenous TTP protein in cells and tissues, we raised a high titer rabbit antiserum against full-length mouse TTP. TTP could be detected on immunoblots of mouse cytosolic tissue extracts; it was most highly ex- pressed in spleen, but its concentration in that tissue was only about 1.5 nM. TTP could be detected readily in splenic macrophages and stromal cells from LPS-in- jected rats. In both LPS-treated RAW 264.7 macrophages and fetal calf serum-treated mouse embryonic fibro- blasts, TTP protein was stable after induction, with min- imal degradation occurring for several hours after treatment of the cells with cycloheximide. The biosyn- thesis of TTP was accompanied by large changes in elec- trophoretic mobility consistent with progressive phos- phorylation. Confocal microscopy revealed that TTP accumulated in a vesicular pattern in the cytosol of the LPS-stimulated RAW 264.7 cells, and was occasionally seen in the cytosol of unstimulated dividing cells. Gel filtration of the endogenous protein suggested that its predominant structure was monomeric. TTP appears to be a low abundance, cytosolic protein in unstimulated cells and tissues, but once induced is relatively stable, in contrast to its very labile mRNA. Tristetraprolin (TTP) 1 is the best-understood member of a small family of tandem CCCH zinc finger proteins. In mam- mals, three members of this family have been characterized: TTP (also known as ZFP36, TIS11, G0S24, and NUP475), ZFP36L1 (also known as TIS11b, cMG1, ERF1, BRF1, and Berg36), and ZFP36L2 (also known as TIS11d, ERF2, and BRF2) (1). Although they are encoded by different genes and their patterns of cell- and tissue-specific expression and agonist- stimulated expression are quite different, they share certain properties: All have highly conserved tandem zinc finger do- mains, in which each C8xC5xC3xH zinc finger is preceded by the sequence (R/K)YKTEL, and the two fingers are separated by 18 amino acids (2); all are nuclear-cytoplasmic shuttling proteins (3, 4); and all are capable of binding AU-rich elements (ARE) within single-stranded RNA (2, 5–9) and promoting the deadenylation and subsequent destruction of those transcripts, both in transfection studies and in cell-free experiments (2, 7, 8, 10). In intact animals, TTP deficiency causes a profound inflammatory syndrome with erosive arthritis, autoimmunity, and myeloid hyperplasia, apparently due almost entirely to excessive production of tumor necrosis factor (TNF) and granulocyte-macrophage colony-stimulating factor (GM-CSF), both of whose mRNAs are direct targets of TTP and are stabi- lized in cells from the knockout (KO) mice (7, 10, 11). For these reasons, TTP can be thought of as an anti-inflammatory or arthritis-suppressor protein. The cDNAs encoding TTP were originally cloned by three groups by virtue of its very rapid and dramatic transcriptional induction in fibroblasts in response to insulin, phorbol esters, and serum (12–14). In a typical example, TTP mRNA was undetectable in serum-starved, insulin-responsive 3T3-L1 mouse fibroblasts, but the transcript became detectable within 10 min of insulin stimulation, peaked at 45 min, and returned to near baseline by 120 min (13). These dramatic but transient responses were also seen in cells in which TTP is thought to exert an effect in normal physiology, i.e. in the regulation of TNF mRNA stability in macrophages (7). Clearly, in most cell types there is a mechanism for the rapid transcription of the TTP gene (Zfp36 in the mouse) in response to various agonists, mediated by some well characterized and some relatively un- characterized enhancer elements (15), and a mechanism for the rapid turning off of transcription while transcript degradation is occurring, leading quickly to a return to baseline mRNA levels despite the continued presence of agonist (1). This rapid but transient increase in TTP mRNA levels is difficult to reconcile with the presumed continuing require- ment for TTP protein in situations in which it might be needed to destabilize TNF mRNA, for example. For these and other * 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. To whom correspondence should be addressed: A2-05, NIEHS, 111 Alexander Dr., Research Triangle Park, NC 27709. Tel.: 919-541-4899; Fax: 919-541-4571; E-mail: [email protected]. 1 The abbreviations used are: TTP, tristetraprolin; hTTP, human TTP; mTTP, mouse TTP; ARE, AU-rich element; BMM, mouse bone marrow-derived macrophages; BSA, bovine serum albumin; CHX, cy- cloheximide; CIAP, calf intestine alkaline phosphatase; FCS, fatal calf serum; GAR-HRP, goat anti-rabbit IgG (HL) horseradish peroxidase conjugate; GM-CSF, granulocyte-macrophage colony-stimulating fac- tor; KO, knockout; LPS, lipopolysaccharide; MBP, maltose binding pro- tein; MEF, mouse embryonic fibroblasts; SDS-PAGE, sodium dodecyl sulfate-polyacrylamide gel electrophoresis; PBS, phosphate-buffered saline; TNF, tumor necrosis factor alpha; TNFR, TNF receptor; TTBS, Tween (0.05%) in Tris-buffered saline; WT, wild-type. THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. 279, No. 20, Issue of May 14, pp. 21489 –21499, 2004 Printed in U.S.A. This paper is available on line at http://www.jbc.org 21489 by guest on April 21, 2020 http://www.jbc.org/ Downloaded from

THE JOURNAL OF BIOLOGICAL CHEMISTRY Printed in U.S.A. … · 2004-04-28 · Immunological Characterization of Tristetraprolin as a Low Abundance, Inducible, Stable Cytosolic Protein*

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: THE JOURNAL OF BIOLOGICAL CHEMISTRY Printed in U.S.A. … · 2004-04-28 · Immunological Characterization of Tristetraprolin as a Low Abundance, Inducible, Stable Cytosolic Protein*

Immunological Characterization of Tristetraprolin as aLow Abundance, Inducible, Stable Cytosolic Protein*

Received for publication, January 27, 2004, and in revised form, March 1, 2004Published, JBC Papers in Press, March 9, 2004, DOI 10.1074/jbc.M400900200

Heping Cao‡, Jane S. Tuttle‡, and Perry J. Blackshear‡§¶

From the ‡Laboratory of Signal Transduction and Office of Clinical Research, NIEHS, National Institutes of Health,Department of Health and Human Services, Research Triangle Park, North Carolina 27709 and the §Departments ofBiochemistry and Medicine, Duke University Medical Center, Durham, North Carolina 27710

Tristetraprolin (TTP) is a zinc finger protein that canbind to AU-rich elements within certain mRNAs, result-ing in deadenylation and destabilization of thosemRNAs. Its physiological targets include the mRNAs en-coding the cytokines tumor necrosis factor � (TNF) andgranulocyte-macrophage colony-stimulating factor. TTPwas originally identified on the basis of its massive buttransient increase in mRNA levels following mitogenstimulation of fibroblasts. It has been difficult to recon-cile this transient mRNA profile with the presumed con-tinuing “need” for TTP protein, for example, to reversethe effects of lipopolysaccharide (LPS)-stimulated TNFsecretion. To investigate this and other questions con-cerning endogenous TTP protein in cells and tissues, weraised a high titer rabbit antiserum against full-lengthmouse TTP. TTP could be detected on immunoblots ofmouse cytosolic tissue extracts; it was most highly ex-pressed in spleen, but its concentration in that tissuewas only about 1.5 nM. TTP could be detected readily insplenic macrophages and stromal cells from LPS-in-jected rats. In both LPS-treated RAW 264.7 macrophagesand fetal calf serum-treated mouse embryonic fibro-blasts, TTP protein was stable after induction, with min-imal degradation occurring for several hours aftertreatment of the cells with cycloheximide. The biosyn-thesis of TTP was accompanied by large changes in elec-trophoretic mobility consistent with progressive phos-phorylation. Confocal microscopy revealed that TTPaccumulated in a vesicular pattern in the cytosol of theLPS-stimulated RAW 264.7 cells, and was occasionallyseen in the cytosol of unstimulated dividing cells. Gelfiltration of the endogenous protein suggested that itspredominant structure was monomeric. TTP appears tobe a low abundance, cytosolic protein in unstimulatedcells and tissues, but once induced is relatively stable, incontrast to its very labile mRNA.

Tristetraprolin (TTP)1 is the best-understood member of asmall family of tandem CCCH zinc finger proteins. In mam-

mals, three members of this family have been characterized:TTP (also known as ZFP36, TIS11, G0S24, and NUP475),ZFP36L1 (also known as TIS11b, cMG1, ERF1, BRF1, andBerg36), and ZFP36L2 (also known as TIS11d, ERF2, and BRF2)(1). Although they are encoded by different genes and theirpatterns of cell- and tissue-specific expression and agonist-stimulated expression are quite different, they share certainproperties: All have highly conserved tandem zinc finger do-mains, in which each C8xC5xC3xH zinc finger is preceded bythe sequence (R/K)YKTEL, and the two fingers are separatedby 18 amino acids (2); all are nuclear-cytoplasmic shuttlingproteins (3, 4); and all are capable of binding AU-rich elements(ARE) within single-stranded RNA (2, 5–9) and promoting thedeadenylation and subsequent destruction of those transcripts,both in transfection studies and in cell-free experiments (2, 7,8, 10). In intact animals, TTP deficiency causes a profoundinflammatory syndrome with erosive arthritis, autoimmunity,and myeloid hyperplasia, apparently due almost entirely toexcessive production of tumor necrosis factor � (TNF) andgranulocyte-macrophage colony-stimulating factor (GM-CSF),both of whose mRNAs are direct targets of TTP and are stabi-lized in cells from the knockout (KO) mice (7, 10, 11). For thesereasons, TTP can be thought of as an anti-inflammatory orarthritis-suppressor protein.

The cDNAs encoding TTP were originally cloned by threegroups by virtue of its very rapid and dramatic transcriptionalinduction in fibroblasts in response to insulin, phorbol esters,and serum (12–14). In a typical example, TTP mRNA wasundetectable in serum-starved, insulin-responsive 3T3-L1mouse fibroblasts, but the transcript became detectable within10 min of insulin stimulation, peaked at �45 min, and returnedto near baseline by 120 min (13). These dramatic but transientresponses were also seen in cells in which TTP is thought toexert an effect in normal physiology, i.e. in the regulation ofTNF mRNA stability in macrophages (7). Clearly, in most celltypes there is a mechanism for the rapid transcription of theTTP gene (Zfp36 in the mouse) in response to various agonists,mediated by some well characterized and some relatively un-characterized enhancer elements (15), and a mechanism for therapid turning off of transcription while transcript degradationis occurring, leading quickly to a return to baseline mRNAlevels despite the continued presence of agonist (1).

This rapid but transient increase in TTP mRNA levels isdifficult to reconcile with the presumed continuing require-ment for TTP protein in situations in which it might be neededto destabilize TNF mRNA, for example. For these and other

* The costs of publication of this article were defrayed in part by thepayment of page charges. This article must therefore be hereby marked“advertisement” in accordance with 18 U.S.C. Section 1734 solely toindicate this fact.

¶ To whom correspondence should be addressed: A2-05, NIEHS, 111Alexander Dr., Research Triangle Park, NC 27709. Tel.: 919-541-4899;Fax: 919-541-4571; E-mail: [email protected].

1 The abbreviations used are: TTP, tristetraprolin; hTTP, humanTTP; mTTP, mouse TTP; ARE, AU-rich element; BMM, mouse bonemarrow-derived macrophages; BSA, bovine serum albumin; CHX, cy-cloheximide; CIAP, calf intestine alkaline phosphatase; FCS, fatal calfserum; GAR-HRP, goat anti-rabbit IgG (H�L) horseradish peroxidaseconjugate; GM-CSF, granulocyte-macrophage colony-stimulating fac-tor; KO, knockout; LPS, lipopolysaccharide; MBP, maltose binding pro-

tein; MEF, mouse embryonic fibroblasts; SDS-PAGE, sodium dodecylsulfate-polyacrylamide gel electrophoresis; PBS, phosphate-bufferedsaline; TNF, tumor necrosis factor alpha; TNFR, TNF receptor; TTBS,Tween (0.05%) in Tris-buffered saline; WT, wild-type.

THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. 279, No. 20, Issue of May 14, pp. 21489–21499, 2004Printed in U.S.A.

This paper is available on line at http://www.jbc.org 21489

by guest on April 21, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 2: THE JOURNAL OF BIOLOGICAL CHEMISTRY Printed in U.S.A. … · 2004-04-28 · Immunological Characterization of Tristetraprolin as a Low Abundance, Inducible, Stable Cytosolic Protein*

reasons, it was important to establish the normal patterns ofTTP protein expression in cells and tissues, as well as theprotein response patterns in cells such as macrophages inwhich the protein is thought to play an important physiologicalrole. However, studies of this type have been hampered by thelack of good antibodies to TTP, a deficiency that is partly due todifficulties in expressing the intact protein in soluble form intypical expression systems (6). In the present studies, we de-scribe the development of a high titer antibody to mouse TTP,and its use in characterizing the normal patterns of TTP ex-pression in mouse tissues and its pattern of induction in cul-tured cells. Among the interesting findings were that TTPseems to be an extraordinarily low abundance protein in nor-mal mouse tissues; that its levels do not necessarily parallel thesteady-state levels of its transcript in these tissues; that it is avery stable cytoplasmic protein once induced, at least in macro-phages and fibroblasts; and finally, that it undergoes progres-sive phosphorylation during biosynthetic induction over a scaleof hours.

MATERIALS AND METHODS

MBP-mTTP Purification and Antibody Production—Maltose bindingprotein-mouse TTP fusion protein (MBP-mTTP) and MBP were purifiedas described (6) and used to produce rabbit antisera according to stand-ard procedures (Covance Research Products, Inc., Denver, PA). Theantiserum that contained the highest titer anti-TTP activity was cen-trifuged at 10,000 � g for 10 min, sodium azide was added to a finalconcentration of 0.02% (w/v), and the serum was stored at �70 °C.

Preparation of Tissue Extracts—Wild-type (WT) C57B16 mice, andmice deficient in TTP, TTP/TNF� receptor 1 (TNFR1), or TTP/TNF�receptor 2 (TNFR2) (11, 16) were maintained in microisolator cages ina barrier facility according to institutional guidelines. Tissues werecollected from mice at �6 months of age after euthanasia with CO2. Thetissues were frozen in liquid nitrogen, stored at �70 °C, pulverized witha mortar and pestle under liquid nitrogen, and then homogenized withan Overhead Strirrer (Wheaton Instruments, Millville, NJ) in 50 mM

Tris-HCl, pH 7.4, 1 mM EDTA, 1.5 mM MgCl2, 250 mM sucrose, 100 mM

NaF, 5 mM dithiothreitol, 2 mM sodium phosphate buffer, 2 mM ZnCl2,1 mM phenylmethylsulfonyl fluoride, and 2 �M leupeptin. Supernatantsfrom centrifugations at 20,000 � g for 10 min at 4 °C were stored at�70 °C after addition of 20% glycerol (v/v). A cytosolic fraction wasseparated from a membrane fraction by further centrifugation at100,000 � g for 1 h at 4 °C.

Cell Culture—Mouse bone marrow-derived macrophages (BMM)were prepared from 6- to 8-month-old WT and TTP KO mice as de-scribed previously (17), then stimulated with 0.1 or 1 �g/ml lipopolysac-charide (LPS, Sigma Chemical Co., St. Louis, MO) for various times.Primary mouse embryonic fibroblasts (MEF) were prepared from mouseembryos at embryonic day 14 and cultured in Dulbecco’s modifiedEagle’s medium (Invitrogen, Gaithersburg, MD) as described (18). Theywere serum-deprived overnight and then stimulated with 10% FCS forvarious times. Mouse RAW 264.7 cells (American Type Culture Collec-tion) were cultured as MEF, except in Eagle’s minimum essential me-dium (Invitrogen), and treated with LPS (0.1 �g/ml) for various times.In one experiment, cells were stimulated with LPS for 2.5 h, followed byLPS “washout” as described (19). In some experiments, 50 �M cyclohex-imide (CHX) was added to inhibit protein synthesis.

Cell Extracts—After washing twice with PBS, cells were gentlyscraped with a rubber policeman into 5 ml of ice-cold PBS, and centri-fuged at 1,000 � g for 5 min at 4 °C. Cells from each 10-cm dish werethen resuspended and lysed in 0.5 ml of a lysis buffer containing 50 mM

NaH2PO4, pH 7.6, 250 mM NaCl, 50 mM NaF, 10 mM imidazole, 0.5%Nonidet P-40, 1 �g/ml leupeptin, and 1 mM phenylmethylsulfonyl fluo-ride. The cell lysate was left on ice for about 20 min and then centri-fuged at 1,000 � g for 5 min at 4 °C. The supernatant was thencentrifuged again at 10,000 � g for 10 min at 4 °C. The 10,000 � gsupernatant was mixed with glycerol to a final concentration of 20%(v/v), frozen in liquid nitrogen, and stored at �20 °C for future use. Thepellet from the 1,000 � g centrifugation was used for the preparation ofnuclear extracts as described previously (7). Alternatively, nuclear ex-tract was prepared from the pellet by sonication and then centrifuga-tion at 10,000 � g for 10 min at 4 °C. To concentrate proteins in themouse fibroblast extracts, proteins in the 10,000 � g supernatant from2–4 dishes were precipitated with 25% (v/v) of trichloroacetic acid on icefor 2 h, washed twice with cold acetone, dried with a Speed-Vac (Thermo

Electron Corp., Waltham, MA), and dissolved in 40 �l of 10 mM Tris-HCl at pH 8.5. Dephosphorylation of mTTP from LPS-stimulated RAW264.7 cellular extracts used calf intestine alkaline phosphatase (CIAP)(Stratagene) as described in the legend to Fig. 6D.

Size Exclusion Chromatography of TTP from Mouse RAW 264.7Cells—Proteins in the 10,000 � g supernatant from RAW 264.7 cellsstimulated with 0.1 �g/ml LPS for 2 h were precipitated with ammo-nium sulfate at 50% saturation, followed by separation on a Superose 6HR 10/30 size exclusion column (Amersham Biosciences, Uppsala, Swe-den) using a similar procedure to that described previously (20). Pro-teins were eluted with 50 mM NaH2PO4, 300 mM NaCl, 20 mM imidaz-ole, 0.05% Tween 20, pH 8.0, and column fractions were analyzed byimmunoblotting using anti-MBP-mTTP serum. The molecular mass ofmTTP was determined by comparing its elution volume to a standardcurve generated with protein standards separated on the same column.The protein standards (Amersham Biosciences) used were bovine pan-creas ribonuclease A (13.7 kDa), bovine pancreas chymotrypsinogen (25kDa), hen egg ovalbumin (43 kDa), bovine serum albumin (67 kDa),rabbit muscle aldolase (158 kDa), bovine liver catalase (232 kDa), horsespleen ferritin (440 kDa), and bovine thyroid thyroglobulin (669 kDa).The void volume of the column was determined with blue dextran (2000kDa).

SDS-PAGE and Immunoblotting—Protein concentrations were de-termined by the Bradford method (Bio-Rad Laboratories, Hercules,CA), with bovine serum albumin (BSA) as the standard, with NaOHtreatment as described (21). Proteins were separated by SDS-PAGEfollowing standard protocols (22) and visualized with either CoomassieBlue (22) or silver staining (23). For Western blotting, proteins weretransferred onto nitrocellulose membranes in transfer buffer containing0.1% SDS and visualized with Ponceau S staining. The membraneswere blocked with 5% nonfat dry milk in 0.05% Tween 20 in Tris-buffered saline (TTBS) for 30–60 min and incubated with the primaryantiserum at a 1:10,000 dilution in the blocking buffer for 1–18 h. Afterbeing washed with TTBS three times for 10 min each, the membraneswere incubated with a 1:10,000 dilution of secondary antibody in TTBSfor 1–6 h. Following three, 10-min washes with TTBS, membranes wereincubated with SuperSignal West Pico Chemiluminescent Substrate(Pierce, Rockford, IL) and exposed to x-ray film. The primary antibodiesused were anti-MBP serum (New England Biolabs, Beverly, MA) andthe anti-MBP-mTTP antiserum described here and its pre-immunecontrol serum. The secondary antibodies were affinity-purified goatanti-rabbit IgG (H�L) horseradish peroxidase conjugate with humanIgG absorbed (GAR-HRP) (Bio-Rad).

Immunocytochemistry and Confocal Microscopy of RAW 264.7Cells—Mouse RAW 264.7 cells were grown on glass slides or glasscoverslips under the conditions described above. Following the additionof LPS (0.1 �g/ml) for various times, the cells were washed with ice-coldPBS three times before being fixed at room temperature for 10 min in4% (w/v) paraformaldehyde in PBS. Cells were again washed with PBStwice and then permeabilized with 0.2% (v/v) Triton X-100 in TBS atroom temperature for 10 min. The cells were then incubated at 4 °Covernight in anti-MBP-mTTP serum (1:8–10,000 dilution) and 1% nor-mal goat serum in 0.1 M sodium phosphate buffer (pH 7.4). Followingtwo washes with 0.2% (w/v) Triton X-100 in TBS, the cells were incu-bated at room temperature for 30 min in goat anti-rabbit Alexa Fluor488 (1:300 dilution, Molecular Probes) and 25% normal goat serum inTBS. The cells were rinsed twice with PBS for 10 min each time beforebeing mounted with Prolong anti-fade (Molecular Probes). After dryingovernight, the slides were examined and imaged with an LSM510 UVconfocal microscope (Zeiss, Thornwood, NY).

Immunostaining of Rat Spleen Sections—We took advantage of thefact that mouse and rat TTP are 96% identical (compare GenBankTM

accession numbers NP_035886 and NP_579824) to examine the pat-terns of immunoreactive TTP expression in rat spleen. Male Sprague-Dawley rats at �12 weeks of age were maintained as described (24) andinjected intraperitoneally with LPS (5 mg/kg in 5 ml/kg PBS) or thesame volume of PBS as a control. 2 h later, spleens were collected, fixedin Bouin’s fixative for 48 h, clarified in 70% ethanol, then embedded inparaffin and used for immunohistochemistry essentially as described(25). The sections were incubated with the anti-MBP-mTTP serum orpreimmune serum (1:1000) in PBS containing 1% BSA at 37 °C for 45min, washed in PBS three times for 5 min each, and incubated with abiotin-labeled anti-rabbit antibody (1:200) (Vector Laboratories, Bur-lingame, CA) in PBS containing 1% BSA at 37 °C for 20 min. Thesections were washed as above, and the avidin/biotin complex wasdeveloped with 3,3�-diaminobenzidine staining, counterstaining, anddehydration as described (25) using the Vectastain ABC Elite kit (Vec-tor Laboratories).

Immunological Characterization of Tristetraprolin21490

by guest on April 21, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 3: THE JOURNAL OF BIOLOGICAL CHEMISTRY Printed in U.S.A. … · 2004-04-28 · Immunological Characterization of Tristetraprolin as a Low Abundance, Inducible, Stable Cytosolic Protein*

RESULTS

Production and Characterization of Anti-MBP-mTTP Se-rum—For antigen production, MBP-mTTP was expressed inEscherichia coli and purified to near-homogeneity as described(6). The results of this purification are shown after stainingwith Coomassie Blue (Fig. 1A) and immunoblotting with ananti-MBP antibody (Fig. 1B). The Mono Q fractions with thehighest purity of MBP-mTTP (Fig. 1, lane 6) were pooled,concentrated, and used as antigen.

The resulting anti-MBP-mTTP serum at a 1:10,000 dilutionwas able to detect as little as 1 ng of the purified MBP-mTTP,using the SuperSignal detection reagent and a 5-s exposure ofthe blot (Fig. 1C). We also tested the antiserum using extractsfrom 293 cells expressing mouse TTP (3, 4). The antiserum at1:10,000 dilution was able to detect mouse TTP when as littleas 1 �g of total protein from the transfected 293 cells wasloaded into the gel (Fig. 1C). This antiserum recognized mouseTTP but not human TTP or mouse Zfp36L1 under these immu-noblotting conditions (data not shown). However, it could rec-ognize human TTP when much more protein was used or whenthe blot was exposed to film for longer times (see below).

Identification of TTP in Mouse Cells and Tissues—Using thepresent antiserum at 1:10,000 dilution, TTP could be detectedin Western blots of normal spleen when 5 mg of protein per gellane and the SuperSignal detection reagent were used (Fig.2A). TTP in cells and tissues occurs as multiple bands of Mr

about 40–50,000, probably a mixture of differentially phospho-rylated species and degradation products (1, 7). The TTP bandswere absent in spleen taken from TTP KO mice (Fig. 2A). As apositive control, we used 2 �g of protein extract from trans-fected 293 cells (Fig. 2A).

The 20,000 � g spleen supernatant was separated into su-pernatant and pellet fractions by centrifugation at 100,000 � g.TTP was found primarily in the 100,000 � g supernatant, withvery little immunoreactivity in the membrane pellet (Fig. 2B).No TTP signal was detected in the 20,000 � g pellet (data notshown). Because purified recombinant MBP-mTTP (10 ng, or0.134 pmol of the 74.9-kDa fusion protein) was probed on thesame blot as the spleen cytosol (1.7 mg of total protein), we usedthis blot to estimate the approximate concentration of TTP innormal mouse spleen. In these studies, the average spleenweight was 95.2 mg (n � 14), the average soluble proteincontent per spleen was 16.5 mg/spleen (n � 14), and adultmouse spleen water content was �86% (26). As shown in Fig.2B, the TTP immunoreactivity in 1.7 mg of spleen extractprotein appeared to be about 10% of that of 10 ng (0.134 pmol)of the recombinant protein, or 0.0134 pmol. Therefore, TTP wasexpressed at �0.0134 pmol per 1.7 mg/16.5 mg/spleen, or0.1265 pmol/spleen. Because average spleen water content was86% of an average wet weight of 95.2 mg/spleen, the finalestimated TTP concentration was 0.1265 pmol/81.87 �l, orabout 1.5 nM.

FIG. 1. Purification of MBP-mTTP from E. coli and characterization of antibodies. MBP-mTTP was purified from E. coli transformedwith plasmid pMBP-mTTP. MBP-mTTP was initially purified from a 10,000 � g supernatant by amylose resin chromatography, followed bySuperose 12 size exclusion and Mono Q anion exchange chromatography. A, purification of MBP-mTTP stained with Coomassie Blue: lane 1,protein size standards; lane 2, homogenate (50 �g of protein); lane 3, supernatant (50 �g); lane 4, amylose resin column peak fraction (5 �g); lane5, Superose 12 column peak fraction (1 �g); lane 6, Mono Q column peak fraction (1 �g); and lane 7, MBP eluted from the amylose resin column(5 �g). The positions of MBP-mTTP and MBP are indicated. B, detection of MBP-mTTP and MBP by anti-MBP serum. The samples were identicalto those shown in A except that about 10% of the amount of protein was used in each lane. C, characterization of anti-MBP-mTTP serum by Westernblotting. The indicated amounts of MBP-mTTP eluted from the amylose resin column, and HA-mTTP in soluble extracts of transfected 293 cells,were probed with the anti-MBP-mTTP serum (1:10,000) and GAR-HRP (1:10,000) for 30 min each before being incubated in the detection reagentfor 5 min and exposed to x-ray film for 30 s. The smaller immunoreactive bands seen both with the E. coli protein and the protein expressed in 293cells are presumed to be proteolytic fragments.

Immunological Characterization of Tristetraprolin 21491

by guest on April 21, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 4: THE JOURNAL OF BIOLOGICAL CHEMISTRY Printed in U.S.A. … · 2004-04-28 · Immunological Characterization of Tristetraprolin as a Low Abundance, Inducible, Stable Cytosolic Protein*

Distribution of TTP in Mouse Tissues—The anti-MBP-mTTPserum (1:10,000 dilution) cross-reacted with proteins of theappropriate size in spleen, lung, liver, large intestine, andthymus, and with proteins of different sizes from several othertissues (Fig. 3A). To distinguish between true positive and falsepositive signals, we analyzed tissues from mice deficient ineither TNFR1 and WT for TTP (Fig. 3B) or deficient in bothTNFR1 and TTP (Fig. 3C). These genotypes were used, becausethe absence of the TNFR1 largely prevents the development ofthe inflammatory TTP-deficiency syndrome, which might havealtered the expression of nonspecific bands in the tissues.When compared with tissues from the TTP KO mice (Fig. 3C),true positive TTP signals were seen in spleen, lung, liver, andlarge intestine (Fig. 3B), whereas nonspecific immunoreactivebands of various sizes were detected in brain, heart, pancreas,and skin from both the TTP WT and KO mice (Fig. 3, compareB and C). These data showed that spleen expressed the highestconcentration of TTP of any tissue examined.

Induction of TTP in Cultured Cells—To address the specific-ity of this antiserum in cultured cells, BMM from WT and TTPKO mice were exposed to LPS (1 �g/ml) for 2 h, and proteinsfrom 10,000 � g supernatants were separated by SDS-PAGEand probed with the antiserum at a 1:10,000 dilution. Themultiple bands of TTP were clearly detected when 200 �g ofcellular protein was used per gel lane from the LPS-inducedWT BMM, but were not seen in the macrophages from the TTPKO mice (Fig. 4A). Similar results were obtained using 500 �gof protein per lane in the 10,000 � g supernatants from WT butnot KO MEF, which had been stimulated with 10% FCS for 2and 3 h (Fig. 4B). As also shown in Fig. 4B, TTP was readilydetected in RAW 264.7 cell extracts when only 50 �g of protein

was used per lane from the 10,000 � g supernatant of cellsstimulated with 0.1 �g/ml LPS.

Time Course of Induction and Stability of TTP in CulturedCells—To investigate the patterns of TTP protein accumulationin cultured cells, BMM were collected following LPS (0.1 �g/ml)stimulation. TTP was undetectable at time 0 but readily de-tectable after 2 h, reaching a peak of expression at about 4 h(Fig. 5A). Immunoreactive TTP in 500 �g of BMM protein wascompared with only 50 �g of protein from RAW 264.7 cellsstimulated with LPS for 1.5 and 2 h (Fig. 5A).

Similar induction kinetics were seen in MEF stimulated with10% FCS (Fig. 5B). Again, TTP was undetectable in the serum-deprived cells at time 0, but accumulated dramatically to reachpeak levels at �2–3 h. These peak levels, detected with 200 �gof cellular protein per lane, were comparable to the levels seenin LPS-stimulated RAW 264.7 cells when only 10 �g of proteinwas loaded into the gel lane (Fig. 5B). Interestingly, there wasa continued shift upward in the apparent molecular weight ofthe protein with still longer times of exposure to FCS (Fig. 5B),compatible with increasing phosphorylation. Protein was stillreadily detectable at 5–6 h.

To estimate the stability of the newly synthesized protein inthese cells, the cells were stimulated for 2 h with 10% FCS andthen treated with CHX (50 �M) for a further 4 h (Fig. 5B).Remarkably, there was very little apparent disappearance ofthe protein over 4 h following treatment with CHX (Fig. 5B), atconcentrations that completely inhibited protein synthesis inthis cell type (data not shown).

Because RAW 264.7 cells produced much more TTP than theother cell types we tested, we analyzed the induction kinetics inmore detail in these cells. As shown in Fig. 5 (C and D), TTP

FIG. 2. Identification of endogenous TTP in mouse spleen. A, 2 �g of protein from 293 cell extracts expressing HA-mTTP, and 5 mg ofprotein from 20,000 � g supernatants of spleen homogenate from WT and TTP KO mice, were separated by SDS-PAGE and transferred ontonitrocellulose membranes for Western blotting. The membrane was incubated in anti-MBP-mTTP serum (1:10,000) overnight and with GAR-HRP(1:10,000) for 1 h and exposed to x-ray film for 1 min. B, the 20,000 � g supernatant from mouse spleen was further centrifuged at 100,000 � gfor 1 h to separate microsomal membranes (Membranes) from the cytosol (Soluble). Equal amounts of protein (1.7 mg) from the soluble andmembrane fractions were used for Western blotting as above, along with MBP-mTTP (10 ng) from the amylose resin column and HA-mTTP in thetransfected 293 cell extracts (1 �g of total soluble protein). In this case, the membrane was exposed to x-ray film for 10 min.

Immunological Characterization of Tristetraprolin21492

by guest on April 21, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 5: THE JOURNAL OF BIOLOGICAL CHEMISTRY Printed in U.S.A. … · 2004-04-28 · Immunological Characterization of Tristetraprolin as a Low Abundance, Inducible, Stable Cytosolic Protein*

was undetectable in unstimulated cells and in cells stimulatedwith LPS for 15 and 30 min, using 50 �g of cytosolic protein anda relatively short autoradiographic exposure (Fig. 5, C and D).TTP could be detected in the 0.5-h samples if much moreprotein and/or much longer autoradiographic exposure wereused (data not shown). TTP was detectable following inductionfor 45 min (Fig. 5C). At 1 h, TTP was detected as two majorbands of about Mr 38,000 and 42,000 (Fig. 5D). As TTP contin-ued to accumulate, there were continued increases in the ap-parent molecular weight to 40–50,000, which stabilized by 2 h(Fig. 5, C and D). Significant amounts of immunoreactive TTPwere still detectable in samples collected after 24 h of LPSinduction (Fig. 5D). We also calculated TTP concentration inthe stimulated RAW 264.7 cells with 0.1 �g/ml LPS for 2 husing known concentrations of the purified MBP-mTTP (1–100ng). The immunoreactivity in 50 �g of RAW 264.7 cell 10,000 �g supernatant was approximately the same as that of 10 ng(0.134 pmol) of the recombinant protein. This represents about340 times the concentration in normal spleen, or �0.5 �M.

The stability of TTP in RAW 264.7 cells was further evalu-ated following LPS stimulation for 2 h (0.1 �g/ml), followed bytreatment with CHX (50 �M). No TTP was detected in RAW264.7 cell extracts 1, 1.5, 2, 3, 4, or 5 h after both LPS and CHXwere added to the culture medium at the same time, indicatingthat this concentration of CHX was effective at inhibiting pro-tein synthesis under these experimental conditions (data notshown). Despite the inhibition of protein synthesis, there wasonly a modest decrease of TTP immunoreactivity over the next4 h, although there was a continued shift to apparently greatermolecular weight between 2 and 3 h after LPS, even in thepresence of CHX (Fig. 5E). Similar slow rates of protein disap-

pearance were observed in cells stimulated with LPS for 2.5 hfollowed by LPS “washout” and CHX treatment (data notshown).

Monomeric Nature of TTP in RAW 264.7 Cells—The10,000 � g supernatant from RAW 264.7 cells stimulated withLPS (0.1 �g/ml) for 2 h was subjected to size exclusion chroma-tography following ammonium sulfate concentration. TTPcould be detected in a range of column fractions, but the peakof TTP was detected in fraction #31 (Fig. 6A), which corre-sponded to a monomer size of about 40 kDa when comparedwith a standard curve (Fig. 6B). Small amounts of TTP weredetected in earlier fractions (Fig. 6A), suggesting that a smallproportion of the protein might exist in RAW 264.7 cells asoligomers or in complexes with other proteins under theseconditions.

When LPS-stimulated RAW 264.7 cells were fractionatedinto cytosolic and nuclear fractions, most of the TTP was in thecytosolic fraction, with little if any associated with the nuclearfraction (Fig. 6C); there was no evidence of higher molecularweight oligomers or aggregates under these conditions. Therewas also some degree of cross-reactivity with the human TTPprotein expressed in and purified from E. coli (Fig. 6C, lane 5).This figure also illustrates the extent of the inhibited SDS-PAGE migration caused by, presumably, phosphorylation inthe RAW 264.7 cells. Human TTP has a predicted molecularweight of 34,086, and the protein purified from E. coli migratedas a single band at Mr �36,000, as estimated with proteinstandards (lane 5), as we have shown previously (6). However,the mouse protein in LPS-stimulated RAW 264.7 cells, with acalculated molecular weight of 33,613, migrated as multiplebands of apparent molecular mass 40–50 kDa. The migration

FIG. 3. Expression of TTP in mouse tissues. A supernatant (10,000 � g) was prepared from tissues from WT and TTP KO mice and was usedfor immunoblotting. Transfected 293 cell extracts expressing HA-mTTP (1 �g of total protein) were used as positive controls. A, in each lane, 1.7mg of total protein in the supernatant from WT mouse tissue was used, and the blot was exposed to x-ray film for 4 min. The broad band or doubletcorresponding to TTP is indicated. B, TNFR1 KO (TTP WT); C, TNFR1/TTP double knockout (TTP KO) tissue extracts. 1 mg of total protein in thetissue supernatants was used, and the blots were exposed to x-ray film for 1 h in the same cassette. The bands seen in B but not in C representspecific TTP immunoreactivity; the bands shown in both blots represent nonspecific protein reactivity.

Immunological Characterization of Tristetraprolin 21493

by guest on April 21, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 6: THE JOURNAL OF BIOLOGICAL CHEMISTRY Printed in U.S.A. … · 2004-04-28 · Immunological Characterization of Tristetraprolin as a Low Abundance, Inducible, Stable Cytosolic Protein*

of TTP was increased following dephosphorylation of mTTP inthe RAW 264.7 cell 10,000 � g supernatant with CIAP treat-ment (Fig. 6D). Incubation with longer time or with more CIAPresulted in a faster migration of the protein, although the sizesof those dephosphorylated proteins were still larger than thatof hTTP purified from E. coli cells (data not shown). Thischange in SDS-PAGE migration appears to be largely due tophosphorylation, because the multiband complex collapses intoa single protein band of lower apparent molecular weight upondephosphorylation with alkaline phosphatase (27).

Immunostaining of TTP in Intact Cells and Tissues—To fur-ther localize TTP in RAW 264.7 cells, they were treated with0.1 �g/ml LPS for 3 h, followed by immunostaining with anti-MBP-mTTP (1:8,000 dilution). As shown in Fig. 7A, confocalmicroscopy revealed bright cytosolic immunofluorescence inLPS-treated RAW 264.7 cells, whereas minimal fluorescencewas detected in controls treated in parallel with LPS inductionand pre-immune serum; without LPS induction but with anti-MBP-mTTP serum; or without LPS treatment but with pre-immune serum. The cytosolic staining was in a vesicular pat-tern (Fig. 7A). There were occasional foci of nuclear staining,but these were seen also in the cells not treated with LPS; theywere more prominent with immune serum than in the othertwo negative controls (Fig. 7A), suggesting that they may rep-resent true foci of nuclear TTP. Serial sections of a single RAW264.7 cell following LPS induction showed that the vesicularpattern of fluorescence was almost exclusively cytosolic, withlittle if any signal in the nucleus (Fig. 7B).

Consistent with the immunoblotting results, TTP was barelydetectable under unstimulated conditions but peaked in thecytosol by 2–3 h following LPS stimulation (Fig. 7C). Signifi-cant immunostaining was still visible in the RAW 264.7 cellsafter 5 h, although the signal was somewhat decreased relativeto the earlier time points (Fig. 7C). Although there was mini-mal detectable fluorescence in the unstimulated cells in Fig. 7(A and C), we occasionally noticed increased fluorescence in the

cytosol of cells undergoing division, even in the absence of LPS(Fig. 7D).

We also evaluated the use of the antibody in rat spleen, afterthe intraperitoneal injection of LPS (5 mg/kg). 2 h after theinjection, there was prominent TTP staining in macrophagesand stromal cells in the spleen periphery (Fig. 8A), whereas nospecific staining was seen in spleen from a PBS-injected rat (C)or with preimmune serum staining of spleen from either anLPS-injected (B) or PBS-injected (D) rat. The light microscopicimages in A–D were obtained and processed under identicalconditions. Of interest was the negative staining of the whitepulp in the center of Fig. 8A (arrow) (see “Discussion”). Thisimmunostaining was seen with the use of Bouin’s fixed, paraf-fin-embedded sections, but not with spleens fixed in 4%paraformaldehyde and processed for frozen sections (data notshown).

DISCUSSION

Despite more than 13 years of information about the tran-scriptional induction of TTP in response to a variety of growthfactors, cytokines, and mitogens, relatively little is knownabout the behavior of the TTP protein itself under similarcircumstances (1). This lack of information is due in part todifficulties in expressing the recombinant protein, makingpreparation of high affinity antibodies more difficult (6). How-ever, the most difficult problem to overcome has been theapparent extraordinary scarcity of the protein in most cells andtissues. We have begun to approach some of these issues bydeveloping a high titer antibody to the recombinant mouse TTPprotein expressed in E. coli as a MBP fusion protein.

One of the fundamental questions we hoped to address withthis new antiserum was whether TTP protein was relativelystable in cells after its induction by mitogens and cytokines.This question arose because of the archetypal immediate-earlyresponse gene characteristics of the TTP mRNA after its induc-tion by insulin, serum, or other mitogens in cultured fibroblasts

FIG. 4. TTP expression in primary mouse macrophages, RAW 264.7 cells, and MEF. A, primary mouse macrophages from TTP WT andKO mice were treated with LPS (1 �g/ml) for 2 h, and soluble cellular extracts were prepared. Equal amounts of protein from these extracts (200�g) were used for immunoblotting, using the anti-MBP-mTTP serum (1:10,000). The blot was exposed to x-ray film for 30 s. The position ofimmunoreactive TTP is shown in the WT but not the KO cells. B, RAW 264.7 cells and MEF. RAW 264.7 cells (RAW) were stimulated with LPS(0.1 �g/ml) for 1.5 and 2 h, and used for the preparation of 10,000 � g supernatants. Similarly, MEF from WT and TTP KO mice were stimulatedwith 10% FCS for 2 and 3 h. Supernatants from RAW (50 �g) and MEF (500 �g) were used for immunoblotting with the anti-MBP-mTTP serum(1:10,000). The blot was exposed to x-ray film overnight. The position of TTP is indicated; note its absence in the KO MEF.

Immunological Characterization of Tristetraprolin21494

by guest on April 21, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 7: THE JOURNAL OF BIOLOGICAL CHEMISTRY Printed in U.S.A. … · 2004-04-28 · Immunological Characterization of Tristetraprolin as a Low Abundance, Inducible, Stable Cytosolic Protein*

(1). For example, in mouse 3T3-L1 adipocytes, TTP mRNA wasessentially undetectable by Northern blot in the basal, serum-deprived G0 state. However, within 10 min of exposure toinsulin, there was a detectable level of mature TTP transcript,and this continued to accumulate for the first 45 min, afterwhich there was a rapid decline in transcript levels to nearbasal concentrations by 120 min (13). Similar induction andturnover kinetics were observed in what is believed to be aphysiologically relevant cell type and response, i.e. the induc-tion in macrophages by agents such as LPS or TNF (7). In bothsituations, the rapid and dramatic induction of transcriptionwas followed by a similarly rapid shut-off of transcription ac-companied by mRNA lability, such that steady-state levels ofmRNA had returned to near-basal levels despite the continuedpresence of the inducing stimulus (1).

This rapid and transient transcript response is characteristicof prototypical immediate early response genes such as c-fos.However, this type of response did not fit with the proposedphysiological function of the TTP protein, i.e. to bind to anddestabilize mRNAs such as those encoding TNF and GM-CSF(1). For example, TNF mRNA is rapidly and dramatically in-duced by LPS in macrophages, followed by the production ofsecreted TNF (7); how can we reconcile the rapid but transient

TTP transcript response with the presumed continued require-ment for TTP protein to cause degradation of the TNF tran-script and reversal of TNF secretion?

In the present study, we addressed this apparent discrep-ancy by evaluating the induction patterns of TTP protein andits stability in cultured MEF after stimulation with serum, andin cultured mouse RAW 264.7 macrophages after stimulationby LPS. In the latter case, TTP protein was readily detectableas multiple bands on Western blots of a crude cytoplasmicfraction from the cells after induction, although the protein wasbarely detectable in the unstimulated cells, as noted previously(28). These multiple bands were shown previously to be multi-ple phosphorylated species, because treatment of the protein inextracts from 293 cells with alkaline phosphatase caused col-lapse of the multiple bands into a single, presumably dephos-phorylated species (27). In LPS-stimulated RAW 264.7 cells,TTP protein accumulated to reach near steady-state levelsafter 90–120 min; remarkably, these high levels of proteinremained nearly constant for many hours thereafter, and werestill readily detectable after 24 h. In keeping with this pattern,treatment of the cells with CHX 2 h after LPS stimulation, atconcentrations shown to completely inhibit protein synthesis inthese cells, revealed a very stable protein, with minimal decline

FIG. 5. Time course of induction and stability of TTP in mouse cells. RAW 264.7 cells and primary mouse macrophages were stimulatedwith 0.1 �g/ml LPS, and MEF were stimulated with 10% FCS, for various times as indicated. To measure protein stability, the cells werestimulated for 2 h before being treated with CHX (50 �M) for the indicated times. A supernatant (10,000 � g) was prepared from the cells and usedfor immunoblotting using the anti-MBP-mTTP serum (1:10,000). A, data from RAW 264.7 cells (RAW) (50 �g of protein per lane) were comparedwith data from primary macrophages (500 �g per lane). The blot was incubated in the primary antiserum for 2 h and the secondary antibody for1 h, then exposed to x-ray film overnight. B, data from RAW 264.7 cells stimulated by LPS for 2 h (10 �g per lane) were compared with data fromMEF (fibroblasts) stimulated with FCS with or without the later addition of CHX (200 �g of protein per lane). The blot was incubated in theprimary antiserum overnight and the secondary antibody for 2 h, then exposed to x-ray film for 2 min. C, time course of TTP induction in RAW264.7 cells (50 �g of protein per lane). The blot was incubated in the primary antiserum for 18 h and the secondary antibody for 1 h. The blot wasexposed to x-ray film for 5 s. D, time course of TTP induction in RAW 264.7 cells without CHX (50 �g of protein per lane). The blot was incubatedin the primary antiserum for 1.5 h and the secondary antibody for 1 h. The blot was exposed to x-ray film for 30 s. E, TTP stability in RAW 264.7cells (50 �g of protein per lane). The cells were first stimulated with LPS for 2 h, followed by treatment with CHX for the indicated times. The blotwas incubated in the primary antiserum for 1.5 h and the secondary antibody for 1 h, then exposed to x-ray film for 30 s.

Immunological Characterization of Tristetraprolin 21495

by guest on April 21, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 8: THE JOURNAL OF BIOLOGICAL CHEMISTRY Printed in U.S.A. … · 2004-04-28 · Immunological Characterization of Tristetraprolin as a Low Abundance, Inducible, Stable Cytosolic Protein*

in protein levels observed for several hours, until generalizedCHX toxicity began to develop. Similar patterns were observedin serum-treated mouse fibroblasts, again with the formation ofa very stable protein. Taken together, these data make physi-ological sense, in that the continued presence of the newlysynthesized protein would be expected to continue to exert itsnegative regulatory effects on ARE-containing transcripts forseveral hours after the initial stimulation.

Another interesting aspect of this protein induction patternis the gradual increase in apparent phosphorylation that ac-companied the increase in steady-state protein levels. Bothmaximal phosphorylation, as indicated by maximal increase inapparent molecular weight on SDS gels, and maximal steady-state protein levels were achieved by �4 h after stimulation inboth macrophages and fibroblasts. This interesting pattern isdifferent from a more typical protein phosphorylation para-digm, in which previously synthesized protein is phosphoryl-

ated within a few minutes by an activated protein kinase. Inthis unusual case, phosphorylation and biosynthesis appearedto occur in parallel, raising interesting questions about thetypes of protein kinases involved, their regulation, and theireffects on the protein. To date, sites for the broad family ofmitogen-activated protein kinases have been identified in theprotein (18), as well as sites for the MKK2 kinase that appearto be involved in binding of the protein to 14-3-3 protein (29,30). Nonetheless, many more phosphorylation sites remain tobe identified on this protein. It is interesting to speculate thatthe increase in phosphorylation that accompanied the in-creased biosynthesis of the protein might be involved in con-ferring stability to the protein, as noted previously for p53, forexample (31, 32). Obviously, other regulatory possibilities ofthis phosphorylation exist, including the regulation of the nu-clear to cytoplasmic shuttling of TTP, its binding affinity for itsRNA targets, its association with other proteins, and others.

FIG. 6. Size exclusion chromatography and dephosphorylation of endogenous TTP from RAW 264.7 cells. A, proteins in the 10,000 �g supernatant fraction from RAW 264.7 cells following LPS induction and ammonium sulfate concentration were separated by size exclusionchromatography with a Superose 6 column. TTP was detected by immunoblotting with the anti-MBP-mTTP antiserum (1:10,000). The position ofTTP is indicated. B, the size of TTP was determined with a standard curve generated with protein standards separated on the same column underthe identical conditions, in which Kav � (Ve � Vo)/(Vt � Vo), where Ve, Vo, and Vt are the elution volume of the protein determined by the experiment,the void volume determined with blue dextran, and the bed volume of the column provided by the manufacturer, respectively. The peak fractioncontaining TTP (#31) corresponded to a molecular size of 40 kDa (diamond). C, cytosolic localization of TTP in RAW 264.7 cells. RAW 264.7 cellswere fractionated into cytosolic and nuclear fractions in two separate experiments after stimulation with LPS (0.1 �g/ml) for 4 and 6 h. Equalamounts of protein from the cytosolic and nuclear fractions (50 �g) were used for immunoblotting with anti-MBP-mTTP serum (1:10,000). HumanTTP (no fusion partner) purified from E. coli (20 ng) was used to demonstrate the electrophoretic mobility shift of TTP in RAW 264.7 cells followingLPS induction. This blot was incubated in the primary antiserum for 1.5 h and the secondary antibody for 1 h. The blot was exposed to x-ray filmfor 10 s. D, dephosphorylation of mTTP in LPS-stimulated RAW 264.7 cell extracts. RAW 264.7 cells were stimulated with LPS (0.1 �g/ml) for 0,1, 2, 3, 4, and 6 h. The 10,000 � g supernatant (100 �g of total protein) was incubated with or without CIAP (35 units) at 23 °C (room temperature)for 1 h. Half of the mixture was used for immunoblotting detection. This blot was incubated in the primary antiserum for 18 h and the secondaryantibody for 1 h. The blot was exposed to x-ray film for 10 s. “hTTP” refers to the same protein as in C, lane 5, above.

Immunological Characterization of Tristetraprolin21496

by guest on April 21, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 9: THE JOURNAL OF BIOLOGICAL CHEMISTRY Printed in U.S.A. … · 2004-04-28 · Immunological Characterization of Tristetraprolin as a Low Abundance, Inducible, Stable Cytosolic Protein*

The studies described here are in general agreement withprevious studies of TTP expression and subcellular localiza-tion. For example, Taylor et al. (18) used a mouse amino-terminal anti-peptide antibody and immunoprecipitation todemonstrate the induction of TTP in mouse fibroblasts and thedecreased gel mobility upon cell stimulation with various mi-togens, due in part to phosphorylation by one or more mitogen-activated protein kinases. They also used the same antiserumto show that TTP in stably transfected serum-deprived fibro-blasts was mostly in the nucleus but translocated to the cytosolwithin 5 min of serum stimulation (33). Carballo et al. (7) founda massive increase of TTP in the cytosol of primary mousemacrophages 4 h after LPS or TNF stimulation, using radioac-tive labeling and immunoprecipitation with the same anti-peptide antiserum. Lai et al. (8) were able to immunoprecipi-tate mouse macrophage TTP cross-linked to radiolabeled TNFRNA and found that most overexpressed TTP was present inthe cytosol of 293 cells. Rigby and colleagues (34) used an

affinity-purified polyclonal antibody directed against a carbox-yl-terminal peptide from human TTP to show that most immu-noreactive TTP was in the cytosol of LPS-stimulated THP-1cells and could be induced rapidly and transiently in those cellsafter LPS stimulation (35). Interestingly, their data fromTHP-1 cells suggested a more transient response of the TTPprotein to LPS stimulation than seen in the mouse cells de-scribed here. The protein turnover rate was not commented onin those studies, nor was the apparent size change upon stim-ulation noted, probably because most of their protein assaysinvolved fluorescence-activated cell sorting.

In the present study, TTP was essentially undetectable inunstimulated RAW 264.7 macrophages, although slightlyhigher levels of punctate nuclear staining were observed inthese cells stained in parallel with the immune serum com-pared with pre-immune serum. This result is compatible with aprevious study in serum-deprived cultured fibroblasts stablyexpressing TTP driven by a metallothionein promoter, in which

FIG. 7. Confocal microscopy detection of TTP in RAW 264.7 cells. A, confocal microscopy of RAW 264.7 cells. Cells were treated with eitherLPS (0.1 �g/ml) or PBS for 3 h, then fixed and stained with either the anti-MBP-mTTP serum (I) or preimmune serum (PI) (1:8,000 dilution).Confocal exposures and image processing were the same for all four panels. B, cytosolic localization of TTP in RAW 264.7 cells. RAW 264.7 cellswere stimulated with LPS (0.1 �g/ml) for 2 h and stained with anti-MBP-mTTP serum (1:10,000 dilution) as described in A. Serial optical sectionsof the same cell were collected at 0.5-�m intervals; the upper left image is approximately in the middle of the cell, whereas the bottom right imageis closest to the glass surface. C, time course of TTP induction in RAW 264.7 cells. RAW 264.7 cells were stimulated with LPS (0.1 �g/ml) for 0,2, 3, and 5 h as indicated and stained with anti-MBP-mTTP serum (1:10,000 dilution) as described in A. D, TTP immunostaining during celldivision in RAW 264.7 cells. Unstimulated RAW 264.7 cells were stained with the anti-MBP-mTTP serum (1:10,000 dilution), as described in A.Immunoreactive TTP was visible in the cytoplasm of the dividing cell indicated by the arrowheads in D, panel 1, whereas no cytoplasmic stainingwas visible in the other cells in the field. A light microscopic image of the same field of cells is also shown (D, panel 2).

Immunological Characterization of Tristetraprolin 21497

by guest on April 21, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 10: THE JOURNAL OF BIOLOGICAL CHEMISTRY Printed in U.S.A. … · 2004-04-28 · Immunological Characterization of Tristetraprolin as a Low Abundance, Inducible, Stable Cytosolic Protein*

unstimulated cells expressed predominantly nuclear TTP, butthe protein had shifted into the cytosol within 5 min of serumstimulation (33). We also noted in the current study that therewas detectable cytoplasmic expression in dividing cells, evenwithout LPS stimulation. Within an hour or so of LPS stainingin the cultured macrophages, clear-cut cytoplasmic stainingwas observed, which remained readily detectable in an inter-esting vesicular pattern for several hours. Cellular fraction-ation of similarly treated cells confirmed the cytoplasmic local-ization of almost all immunoreactivity in the LPS-treated cells.The nature of the vesicular pattern observed in the cytoplasmis not clear, but most immunoreactivity did not sediment witha membrane fraction centrifuged at 100,000 � g for 1 h. Co-staining with markers of other cellular organelles previouslyassociated with TTP, such as exosomes (36) and stress granules(37), as well as other organelles will be of interest in thisregard.

We also used this antiserum to probe the expression of TTPprotein in normal tissues of adult animals. This required ex-traordinarily large amounts of protein to be loaded into the gelwells so that Western blots could be performed, with compara-ble samples from TTP KO mice used to ensure the specificity ofthe observed bands. When this was done, protein was detecta-ble as multiple bands in similar tissues to those previouslycharacterized as expressing the most mRNA by Northern blot-

ting; these include spleen, thymus, lung, and liver, with loweror essentially undetectable expression in other tissues (13).One somewhat surprising finding was the relatively high levelexpression in large intestine, with much lower levels in smallintestine; this is in contrast to previous Northern blot data, inwhich high level mRNA expression was observed in small in-testine.2 This is also interesting given the fact that the TTP KOmice on a C56Bl6 background do not seem to develop a Crohn’s-like colitis, in contrast to mice in which the ARE of the TNFtranscript has been removed, resulting in a stable transcriptand TNF overexpression (38). Identification of the cell typesexpressing TTP under normal circumstances and in models ofexperimental colitis will be of interest in this regard.

In a very recent report in which a commercial antibody wasused, the authors described a very different pattern of TTPtissue distribution in the mouse (39). For example, they de-tected the highest level of expression in liver, with significantexpression in testis and ovary, with TTP being represented asa single sharp band of Mr 43,000; in contrast, we could notdetect immunoreactivity in the testis. In addition, they re-ported minimal to undetectable expression in spleen, thymus,lung, and intestine, in contrast to our results showing readily

2 G. A. Taylor and P.J. Blackshear, unpublished data.

FIG. 8. TTP immunostaining in rat spleen. Rats were injected intraperitoneally with LPS (5 mg/kg; A and B) or PBS (5 ml/kg, C and D), andspleens were removed and used for immunostaining of TTP with either the anti-MBP-mTTP serum (I) or pre-immune serum (PI) as describedunder “Materials and Methods.” The reddish brown staining in A represents TTP immunoreactivity, whereas none was seen in the other threepanels. The arrows in A point to the white pulp of the spleen, which is unstained by the TTP antibody. See the text for other details.

Immunological Characterization of Tristetraprolin21498

by guest on April 21, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 11: THE JOURNAL OF BIOLOGICAL CHEMISTRY Printed in U.S.A. … · 2004-04-28 · Immunological Characterization of Tristetraprolin as a Low Abundance, Inducible, Stable Cytosolic Protein*

detectable expression in those tissues. However, certain criticalcontrols were omitted from their report, including the use ofpre-immune serum, overexpressing cells as a positive control,and tissues from KO mice as negative controls, casting somedoubt on the reliability of their expression data.

We used these Western blot data to calculate an approximateintracellular concentration of TTP in spleen of normal adultmouse and in LPS-stimulated RAW 264.7 cells. This was doneby comparing the expression levels of a known amount ofrecombinant protein to that contained in a known amount ofspleen cytosol and RAW 264.7 cells. This calculation revealedan overall concentration in spleen of about 1.5 nM, and inmaximally stimulated RAW 264.7 cells of about 0.5 �M. Theextraordinarily low value in spleen may account for previousdifficulties in determining protein levels in normal tissues andhigh background of nonspecific bands on Western blots re-ported here. The dramatic stimulation of TTP in RAW 264.7cells by LPS may explain why TTP could be easily detected inthe stimulated cells and the Western blots are much cleanerthan those using normal tissues. Obviously, concentrations inspecific cell types within the spleen are likely to be higher. Oneparadoxical finding is that, in previous studies of TTP tran-script localization by in situ hybridization histochemistry,there was apparent high level transcript expression in thewhite pulp containing almost exclusively lymphocytes,whereas the rest of the spleen expressed much lower levels.3

However, in the present studies, no protein immunoreactivitywas detected in the white pulp of rat spleen, either undernormal circumstances or 2 h after injection of LPS. Instead, theimmunoreactive cells were in the periphery of the spleen andappeared to be macrophages and stromal cells; even in thesecells, there was minimal detectable specific immunoreactivityin the saline-injected rat spleen. Obviously, there are differ-ences in sensitivity of the two techniques, but the finding ofrelatively high transcript levels in the white pulp of the spleen(and other lymphoid organs of the adult mouse) in the settingof undetectable protein levels raises the possibility of a trans-lational block under such circumstances. Alternative explana-tions include the possibility that the protein is labile in theunphosphorylated and/or nuclear state, resulting in the failureto accumulate significant amounts of protein in the unstimu-lated state. These uncertainties should be resolvable in cul-tured lymphocytes or lymphoid cell lines. In any case, the LPSstimulus in intact rats did not seem to overcome this failure oflymphoid protein expression, because the white splenic pulpremained antibody-negative.

A final point concerns the oligomeric nature of the inducedendogenous protein in RAW 264.7 cells. When TTP is overex-pressed in 293 cells, it is common to observe apparent dimerson SDS gels, either by Western blotting or by cross-linking with32P-labeled ARE probes (7, 8). This finding, as well as ourrepeated identification of TTP as a binding partner for itself inyeast two-hybrid analyses,4 suggest that a substantial fractionof TTP might exist in the cell as dimeric or oligomeric species.The current data suggest that the predominant species of en-dogenous, induced TTP in RAW 264.7 cells is a monomer; thiswas supported by the Western blotting data, which did notidentify the same types of putative dimers as seen in theoverexpression studies. Nonetheless, although there was noobvious higher molecular weight peak of immunoreactivity onthe gel filtration column, substantial immunoreactive proteinwas detected at larger sizes. This could conceivably represent

dimers or oligomers of TTP, or possibly hetero-oligomers withputative TTP-interacting proteins.

Acknowledgments—We thank Ester Carballo-Jane and DebbieStumpo for the TTP knockout mouse tissues and some of the bonemarrow-derived macrophage extracts, Wi Lai for the 293 cell extracts,Alex Merrick for the rat spleens, Jeff Reece for assistance with confocalmicroscopy, and Betsy Kennington for her valuable advice. We alsothank Anton Jetten and Kathleen Smoak for helpful comments on themanuscript.

REFERENCES

1. Blackshear, P. J. (2001) Biochem. Soc. Trans. 30, 945–9522. Lai, W. S., Carballo, E., Thorn, J. M., Kennington, E. A., and Blackshear, P. J.

(2000) J. Biol. Chem. 275, 17827–178373. Phillips, R. S., Ramos, S. B., and Blackshear, P. J. (2002) J. Biol. Chem. 277,

11606–116134. Murata, T., Yoshino, Y., Morita, N., and Kaneda, N. (2002) Biochem. Biophys.

Res. Commun. 293, 1242–12475. Blackshear, P. J., Lai, W. S., Kennington, E. A., Brewer, G., Wilson, G. M.,

Guan, X., and Zhou, P. (2003) J. Biol. Chem. 278, 19947–199556. Cao, H., Dzineku, F., and Blackshear, P. J. (2003) Arch. Biochem. Biophys.

412, 106–1207. Carballo, E., Lai, W. S., and Blackshear, P. J. (1998) Science 281, 1001–10058. Lai, W. S., Carballo, E., Strum, J. R., Kennington, E. A., Phillips, R. S., and

Blackshear, P. J. (1999) Mol. Cell Biol. 19, 4311–43239. Worthington, M. T., Pelo, J. W., Sachedina, M. A., Applegate, J. L., Arseneau,

K. O., and Pizarro, T. T. (2002) J. Biol. Chem. 277, 48558–4856410. Carballo, E., Lai, W. S., and Blackshear, P. J. (2000) Blood 95, 1891–189911. Taylor, G. A., Carballo, E., Lee, D. M., Lai, W. S., Thompson, M. J., Patel, D. D.,

Schenkman, D. I., Gilkeson, G. S., Broxmeyer, H. E., Haynes, B. F., andBlackshear, P. J. (1996) Immunity 4, 445–454

12. DuBois, R. N., McLane, M. W., Ryder, K., Lau, L. F., and Nathans, D. (1990)J. Biol. Chem. 265, 19185–19191

13. Lai, W. S., Stumpo, D. J., and Blackshear, P. J. (1990) J. Biol. Chem. 265,16556–16563

14. Varnum, B. C., Lim, R. W., Sukhatme, V. P., and Herschman, H. R. (1989)Oncogene 4, 119–120

15. Lai, W. S., Thompson, M. J., Taylor, G. A., Liu, Y., and Blackshear, P. J. (1995)J. Biol. Chem. 270, 25266–25272

16. Carballo, E., and Blackshear, P. J. (2001) Blood 98, 2389–239517. Carballo, E., Gilkeson, G. S., and Blackshear, P. J. (1997) J. Clin. Invest. 100,

986–99518. Taylor, G. A., Thompson, M. J., Lai, W. S., and Blackshear, P. J. (1995) J. Biol.

Chem. 270, 13341–1334719. Lidington, D., Ouellette, Y., and Tyml, K. (2000) J. Cell Physiol. 185, 117–12520. Cao, H., James, M. G., and Myers, A. M. (2000) Arch. Biochem. Biophys. 373,

135–14621. Cao, H., Sullivan, T. D., Boyer, C. D., and Shannon, J. C. (1995) Physiol. Plant

95, 176–18622. Sambrook, J., Fritsch, E. F., and Naniatis, T. (1989) Molecular Cloning: A

Laboratory Manual, 2nd. Ed., Cold Spring Harbor Laboratory Press, ColdSpring Harbor, NY

23. Shevchenko, A., Wilm, M., Vorm, O., and Mann, M. (1996) Anal. Chem. 68,850–858

24. Bruno, M. E., Borchers, C. H., Dial, J. M., Walker, N. J., Hartis, J. E.,Wetmore, B. A., Carl Barrett, J. C., Tomer, K. B., and Merrick, B. A. (2002)Arch. Biochem. Biophys. 406, 153–164

25. Flores, K. G., Li, J., and Hale, L. P. (2001) Hum. Pathol. 32, 926–93426. Zebrowska, G., Lewa, C. J., Ramee, M. P., Husson, F., and De Certaines, J. D.

(2001) Anticancer Res. 21, 1213–121727. Carballo, E., Cao, H., Lai, W. S., Kennington, E. A., Campbell, D., and Black-

shear, P. J. (2001) J. Biol. Chem. 276, 42580–4258728. Zhu, W., Brauchle, M. A., Di Padova, F., Gram, H., New, L., Ono, K., Downey,

J. S., and Han, J. (2001) Am. J. Physiol. 281, L499–L50829. Johnson, B. A., Stehn, J. R., Yaffe, M. B., and Blackwell, T. K. (2002) J. Biol.

Chem. 277, 18029–1803630. Chrestensen, C. A., Schroeder, M. J., Shabanowitz, J., Hunt, D. F., Pelo, J. W.,

Worthington, M. T., and Sturgill, T. W. (2004) J. Biol. Chem. 279,10176–10184

31. Zacchi, P., Gostissa, M., Uchida, T., Salvagno, C., Avolio, F., Volinia, S., Ronai,Z., Blandino, G., Schneider, C., and Del Sal, G. (2002) Nature 419, 853–857

32. Zheng, H., You, H., Zhou, X. Z., Murray, S. A., Uchida, T., Wulf, G., Gu, L.,Tang, X., Lu, K. P., and Xiao, Z. X. (2002) Nature 419, 849–853

33. Taylor, G. A., Thompson, M. J., Lai, W. S., and Blackshear, P. J. (1996) Mol.Endocrinol. 10, 140–146

34. Brooks, S. A., Connolly, J. E., Diegel, R. J., Fava, R. A., and Rigby, W. F. (2002)Arthritis Rheum. 46, 1362–1370

35. Fairhurst, A. M., Connolly, J. E., Hintz, K. A., Goulding, N. J., Rassias, A. J.,Yeager, M. P., Rigby, W., and Wallace, P. K. (2003) Arthritis Res. Ther. 5,R214–R225

36. Chen, C. Y., Gherzi, R., Ong, S. E., Chan, E. L., Raijmakers, R., Pruijn, G. J.,Stoecklin, G., Moroni, C., Mann, M., and Karin, M. (2001) Cell 107, 451–464

37. Kedersha, N., and Anderson, P. (2001) Biochem. Soc. Trans. 30, 963–96938. Kontoyiannis, D., Boulougouris, G., Manoloukos, M., Armaka, M., Apostolaki,

M., Pizarro, T., Kotlyarov, A., Forster, I., Flavell, R., Gaestel, M., Tsichlis,P., Cominelli, F., and Kollias, G. (2002) J. Exp. Med. 196, 1563–1574

39. Lu, J. Y., and Schneider, R. J. (2004) J. Biol. Chem. 279, 12974–12979

3 P. J. Blackshear and W. S. Young III, unpublished data.4 D. J. Stumpo and P. J. Blackshear, unpublished data.

Immunological Characterization of Tristetraprolin 21499

by guest on April 21, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 12: THE JOURNAL OF BIOLOGICAL CHEMISTRY Printed in U.S.A. … · 2004-04-28 · Immunological Characterization of Tristetraprolin as a Low Abundance, Inducible, Stable Cytosolic Protein*

Heping Cao, Jane S. Tuttle and Perry J. BlackshearStable Cytosolic Protein

Immunological Characterization of Tristetraprolin as a Low Abundance, Inducible,

doi: 10.1074/jbc.M400900200 originally published online March 9, 20042004, 279:21489-21499.J. Biol. Chem. 

  10.1074/jbc.M400900200Access 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/279/20/21489.full.html#ref-list-1

This article cites 36 references, 17 of which can be accessed free at

by guest on April 21, 2020

http://ww

w.jbc.org/

Dow

nloaded from