Bone Morphogenetic Proteins 2 and 6, Expressed in Arthritic...

12
ARTHRITIS & RHEUMATISM Vol. 48, No. 10, October 2003, pp 2807–2818 DOI 10.1002/art.11389 © 2003, American College of Rheumatology Bone Morphogenetic Proteins 2 and 6, Expressed in Arthritic Synovium, Are Regulated by Proinflammatory Cytokines and Differentially Modulate Fibroblast-Like Synoviocyte Apoptosis Rik J. U. Lories, Inge Derese, Jan L. Ceuppens, and Frank P. Luyten Objective. To examine the expression, regulation, and potential roles of bone morphogenetic proteins (BMPs) in arthritic synovium. Methods. Expression of BMPs in arthritic syno- vium from patients with rheumatoid arthritis (RA) or spondylarthropathy (SpA) and in noninflamed syno- vium from patients undergoing diagnostic or therapeu- tic arthroscopies was studied by reverse transcription– polymerase chain reaction (RT-PCR), Western blot, immunohistochemistry, and 2-color immunofluores- cence. In vitro regulation of gene expression in fibroblast-like synoviocytes (FLS) was determined by real-time quantitative RT-PCR and immunohistochem- istry. We used 3 H-thymidine incorporation after serum deprivation–induced growth arrest to examine effects on FLS proliferation. FLS apoptosis was evaluated by flow cytometry cell cycle analysis. Apoptotic cells in synovial tissue were detected by TUNEL staining. Results. Transcripts of different BMPs, most strikingly BMP-2 and BMP-6, were detected in synovial tissues. By Western blot, BMP-2 and BMP-6 precursor protein was found in RA and SpA synovial tissue extracts, but not in extracts of noninflamed synovial tissue. By immunohistochemistry, BMP-2 and BMP-6 were detected in the hyperplastic lining and the sublin- ing layer of synovium from RA and SpA patients, both in CD90 cells (FLS) and in some CD68 cells (macrophages). Proinflammatory cytokines, such as interleukin-1 and tumor necrosis factor , but not interferon-, enhanced the expression of BMP-2 and BMP-6 transcripts in FLS in vitro. Neither BMP-2 nor BMP-6 affected FLS proliferation. BMP-2 promoted FLS apoptosis, whereas BMP-6 protected against nitric oxide–induced FLS apoptosis. BMP-2–positive apopto- tic cells were found in arthritic synovium. Conclusion. BMP-2 and BMP-6 are expressed in arthritic synovium and are strongly up-regulated by proinflammatory cytokines. Although BMP signaling has been proposed to be involved in cartilage and bone repair in arthritis, this pathway may be equally impor- tant in modulating FLS cell populations in inflamed synovium. Chronic inflammatory arthritides, including rheumatoid arthritis (RA) and the spondylarthropathies (SpA), are an important cause of morbidity and disabil- ity. Research efforts have mainly focused on inflamma- tory and immune pathways, leading to the identification of specific cytokines, chemokines, and tissue-destructive enzymes as key players in the pathologic process. Using this approach, successful targeted therapies, such as tumor necrosis factor (TNF) blockade, have been devel- oped for both RA and SpA patients (1,2). Which cells play the leading role in the develop- ment of chronic arthritis is still a subject of debate. The role of immune cells and, more particularly, T cells and autoantigens has been extensively studied (for review, see ref. 3). In RA, interactions between B cells and T cells in the synovium are critical (4). In recent years, it has become clear that other cell populations, including nonimmune cells, also contribute to the pathology and probably to the pathogenesis of chronic arthritis (5). The role of fibroblast-like synoviocytes (FLS) in particular has gained attention in both RA (5,6) and SpA (7). Activated and partially transformed FLS may escape current antiinflammatory and immune-mediated thera- Supported by grant G.0390.03 from the Fund for Scientific Research Flanders. Dr. Lories’ work was supported by an “Aspirant” fellowship from the Fund for Scientific Research Flanders. Rik J. U. Lories, MD, PhD, Inge Derese, Jan L. Ceuppens, MD, PhD, Frank P. Luyten, MD, PhD: Katholieke Universiteit Leuven, Leuven, Belgium. Address correspondence and reprint requests to Frank P. Luyten, MD, PhD, Laboratory for Skeletal Development and Joint Disorders, Herestraat 49, B-3000 Leuven, Belgium. E-mail: [email protected]. Submitted for publication May 24, 2002; accepted in revised form June 5, 2003. 2807

Transcript of Bone Morphogenetic Proteins 2 and 6, Expressed in Arthritic...

Page 1: Bone Morphogenetic Proteins 2 and 6, Expressed in Arthritic …ddegroot/SBO_STRETCH/35_Lories_2003.pdf · spondylarthropathy (SpA) and in noninflamed syno-vium from patients undergoing

ARTHRITIS & RHEUMATISMVol. 48, No. 10, October 2003, pp 2807–2818DOI 10.1002/art.11389© 2003, American College of Rheumatology

Bone Morphogenetic Proteins 2 and 6, Expressed in ArthriticSynovium, Are Regulated by Proinflammatory Cytokines and

Differentially Modulate Fibroblast-Like Synoviocyte Apoptosis

Rik J. U. Lories, Inge Derese, Jan L. Ceuppens, and Frank P. Luyten

Objective. To examine the expression, regulation,and potential roles of bone morphogenetic proteins(BMPs) in arthritic synovium.

Methods. Expression of BMPs in arthritic syno-vium from patients with rheumatoid arthritis (RA) orspondylarthropathy (SpA) and in noninflamed syno-vium from patients undergoing diagnostic or therapeu-tic arthroscopies was studied by reverse transcription–polymerase chain reaction (RT-PCR), Western blot,immunohistochemistry, and 2-color immunofluores-cence. In vitro regulation of gene expression infibroblast-like synoviocytes (FLS) was determined byreal-time quantitative RT-PCR and immunohistochem-istry. We used 3H-thymidine incorporation after serumdeprivation–induced growth arrest to examine effects onFLS proliferation. FLS apoptosis was evaluated by flowcytometry cell cycle analysis. Apoptotic cells in synovialtissue were detected by TUNEL staining.

Results. Transcripts of different BMPs, moststrikingly BMP-2 and BMP-6, were detected in synovialtissues. By Western blot, BMP-2 and BMP-6 precursorprotein was found in RA and SpA synovial tissueextracts, but not in extracts of noninflamed synovialtissue. By immunohistochemistry, BMP-2 and BMP-6were detected in the hyperplastic lining and the sublin-ing layer of synovium from RA and SpA patients, bothin CD90� cells (FLS) and in some CD68� cells(macrophages). Proinflammatory cytokines, such as

interleukin-1� and tumor necrosis factor �, but notinterferon-�, enhanced the expression of BMP-2 andBMP-6 transcripts in FLS in vitro. Neither BMP-2 norBMP-6 affected FLS proliferation. BMP-2 promotedFLS apoptosis, whereas BMP-6 protected against nitricoxide–induced FLS apoptosis. BMP-2–positive apopto-tic cells were found in arthritic synovium.

Conclusion. BMP-2 and BMP-6 are expressed inarthritic synovium and are strongly up-regulated byproinflammatory cytokines. Although BMP signalinghas been proposed to be involved in cartilage and bonerepair in arthritis, this pathway may be equally impor-tant in modulating FLS cell populations in inflamedsynovium.

Chronic inflammatory arthritides, includingrheumatoid arthritis (RA) and the spondylarthropathies(SpA), are an important cause of morbidity and disabil-ity. Research efforts have mainly focused on inflamma-tory and immune pathways, leading to the identificationof specific cytokines, chemokines, and tissue-destructiveenzymes as key players in the pathologic process. Usingthis approach, successful targeted therapies, such astumor necrosis factor (TNF) blockade, have been devel-oped for both RA and SpA patients (1,2).

Which cells play the leading role in the develop-ment of chronic arthritis is still a subject of debate. Therole of immune cells and, more particularly, T cells andautoantigens has been extensively studied (for review,see ref. 3). In RA, interactions between B cells and Tcells in the synovium are critical (4). In recent years, ithas become clear that other cell populations, includingnonimmune cells, also contribute to the pathology andprobably to the pathogenesis of chronic arthritis (5). Therole of fibroblast-like synoviocytes (FLS) in particularhas gained attention in both RA (5,6) and SpA (7).Activated and partially transformed FLS may escapecurrent antiinflammatory and immune-mediated thera-

Supported by grant G.0390.03 from the Fund for ScientificResearch Flanders. Dr. Lories’ work was supported by an “Aspirant”fellowship from the Fund for Scientific Research Flanders.

Rik J. U. Lories, MD, PhD, Inge Derese, Jan L. Ceuppens,MD, PhD, Frank P. Luyten, MD, PhD: Katholieke UniversiteitLeuven, Leuven, Belgium.

Address correspondence and reprint requests to Frank P.Luyten, MD, PhD, Laboratory for Skeletal Development and JointDisorders, Herestraat 49, B-3000 Leuven, Belgium. E-mail:[email protected].

Submitted for publication May 24, 2002; accepted in revisedform June 5, 2003.

2807

Page 2: Bone Morphogenetic Proteins 2 and 6, Expressed in Arthritic …ddegroot/SBO_STRETCH/35_Lories_2003.pdf · spondylarthropathy (SpA) and in noninflamed syno-vium from patients undergoing

pies. In addition, multipotential mesenchymal stem cellshave been identified in the synovium and in FLS culturesafter in vitro expansion (8,9). These cells are suggestedto contribute to tissue remodeling and repair (9) as wellas disease pathogenesis (8).

Modulation of the intrinsic tissue repair capacityis an emerging therapeutic option for the treatment ofboth degenerative and inflammatory joint diseases (10).In arthritis, such strategies are of critical importance inrestoring the damaged tissue that causes morbidity anddisability, even after control of the inflammation hasbeen achieved. Therefore, there is an emerging interestin embryonic signaling, more specifically in those growthfactors and morphogens known to be involved in theembryologic formation of the skeletal tissues and thejoint. Members of the transforming growth factor �(TGF�) superfamily, including TGF�1–3, bone mor-phogenetic proteins (BMPs), and activins, are amongthose key players (11). The TGF� superfamily is a largegroup of structurally related polypeptide cytokines withpleiotropic functions in regulating development, ho-meostasis, and repair of different tissues (12). Originallyidentified as polypeptides responsible for the in vivobone-inductive properties of demineralized bone matrix(13,14), BMPs have been recognized as key moleculesboth in early developmental patterning (15) and in latemorphogenesis in different organs including the skele-ton and the joints (11,16). A role for BMPs in skeletalrepair (e.g., fracture healing) has been demonstrated(17). An important role for both TGF� and BMPs hasbeen suggested in homeostatic and remodeling mecha-nisms in osteoarthritis (OA) (18–20).

Little is known about the potential role of BMPsin chronic inflammatory joint diseases. BMPs are cur-rently considered to be pleiotropic cytokines that influ-ence proliferation, growth, differentiation, and apopto-sis of many different cell types. Therefore, any role ofBMPs in arthritis may extend beyond those in bone andcartilage. BMPs mainly affect cells of mesenchymalorigin (e.g., chondrocytes and osteoblasts). Therefore,FLS may be a specific target of BMP signaling.

We examined the expression of BMP-2 andBMP-6 in arthritic synovium. These BMPs belong todifferent subgroups of the BMP family. In addition, weinvestigated the role of proinflammatory cytokines in theregulation of these morphogens and their potential rolein FLS proliferation and apoptosis. Our data suggestthat BMP signaling may prove to be a therapeutic targetfor controlling FLS behavior, thereby directing andmodulating the processes of repair and remodeling inarthritis.

PATIENTS AND METHODS

Reagents. Recombinant proteins were obtained fromthe following sources: recombinant human BMP-2 from Ge-netics Institute (Cambridge, MA), recombinant human TGF�1and recombinant human BMP-6 from R&D Systems (Abing-don, UK), recombinant human interleukin-1� (IL-1�) andrecombinant human TNF� from BioSource International(Nivelles, Belgium), and recombinant human interferon-�(IFN�) from Boehringer (Mannheim, Germany). Goat poly-clonal anti-human, anti-mouse, and anti-rat BMP-6 antibodywas purchased from Santa Cruz Biotechnology (Santa Cruz,CA). Affinity-purified polyclonal chicken anti-human BMP-2antibody was a gift from Pfizer Central Research (Groton,CT). Monoclonal mouse anti-human CD68 was purchasedfrom Dako (Glostrup, Denmark). Monoclonal mouse anti-human CD90 was purchased from Becton Dickinson (SanJose, CA).

Patient inclusion, needle arthroscopy, and cell isola-tion. Patients were treated at the Department of Rheumatol-ogy or the Department of Orthopedic Surgery, UniversityHospitals Leuven, and gave written informed consent. Thelocal Ethics Committee approved the procedure. All patientswith inflammatory arthritis who were included in the studyfulfilled either the American College of Rheumatology (ACR;formerly, the American Rheumatism Association) 1987 re-vised criteria for RA (21) or the European SpondylarthropathyStudy Group criteria for SpA (22). For a diagnosis of knee OA,the ACR criteria were used (23). Other diagnoses for thecontrol samples were made by combining clinical, radio-graphic, and arthroscopy data.

Needle arthroscopy of the knee, with random synovialbiopsies, was performed under sterile and standardized condi-tions. During needle arthroscopy, synovial hyperemia wasscored macroscopically using a semiquantitative scoring system(scarce, mild, moderate, strong) as reported previously (24).Similarly, synovial hypertrophy was described as mild or man-ifest hypertrophy or pannus formation (defined as hypertro-phic synovial tissue overlying the cartilage) by the arthroscopist(24). Biopsy samples of noninflamed synovium were obtainedfrom patients undergoing diagnostic arthroscopies.

For histologic assessments, 6 biopsy samples weresnap-frozen in TissueTek (Sakura, Zoeterwoude, The Nether-lands) and stored at –70°C for later use. Two biopsy sampleswere immediately used for RNA isolation (see below) orprotein extraction (see below). Two biopsy samples werewashed twice in Hanks’ balanced salt solution (Life Technol-ogies, Merelbeke, Belgium) supplemented with 2� antibiotic–antimycotic solution (100 units/ml penicillin, 100 �g/ml strep-tomycin, and 0.25 �g/ml amphotericin B; Life Technologies).

Cells were isolated using a previously described proto-col (9). Briefly, the biopsy samples were digested overnightwith 0.2% type IV collagenase (Life Technologies) in high-glucose Dulbecco’s modified Eagle’s medium (DMEM; LifeTechnologies) containing 10% fetal bovine serum (FBS; Bio-Whittaker, Verviers, Belgium) and 2� antibiotics (as above).Passage 0 cells from both biopsy samples were pooled andwashed twice in DMEM–10% FBS and antibiotics (growthmedium) and then resuspended in 3 ml of growth medium andplated in 3 different wells of a 6-well tissue culture plate.Nonadherent cells were removed after 72 hours by changing

2808 LORIES ET AL

Page 3: Bone Morphogenetic Proteins 2 and 6, Expressed in Arthritic …ddegroot/SBO_STRETCH/35_Lories_2003.pdf · spondylarthropathy (SpA) and in noninflamed syno-vium from patients undergoing

the growth medium. All tissue culture experiments wereperformed at 37°C in a humidified atmosphere of 5% CO2.

Cell expansion and stimulation. After reaching conflu-ence, cells were washed twice with Dulbecco’s calcium- andmagnesium-free phosphate buffered saline (DPBS; BioWhit-taker), harvested with trypsin–EDTA (0.25% trypsin, 1 mMEDTA; Life Technologies), and replated in a T75 flask.Further passaging was done with 1:3 dilutions. Growth me-dium was changed twice a week. All further experiments wereperformed between passages 4 and 9. For cytokine stimulationexperiments, harvested cells were diluted 1:3 in growth me-dium and plated in 6-well tissue culture plates. At confluence,cells were treated either with ligands (IL-1�, TNF�, or IFN�)or with carrier controls. In additional experiments, cells wereplated on tissue culture slides and incubated at confluence withligands as described above.

Total RNA extraction and reverse transcription–polymerase chain reaction (RT-PCR) analysis. Total RNAfrom stimulated cells or from fresh synovial biopsy specimenswas isolated using a commercially available kit (SNAP; Invitro-gen, Merelbeke, Belgium) according to the manufacturer’sinstructions. Complementary DNA (cDNA) was obtained byRT of 1–2 �g of total RNA (Thermoscript; Invitrogen) witholigo(dT)20 as primer. Conventional PCR was performed aspreviously described (9). Complementary DNA was mixedwith 0.5 units of Taq polymerase (Eurogentec, Seraing, Bel-gium), 0.2 mM dNTP, 0.5 �M specific primers, and 1.5 mMMgCl2. Primer pairs were designed using Vector NTI software(InforMax, North Bethesda, MD). The sequences of theprimers, the sizes of the PCR products, and the optimumannealing temperatures are shown in Table 1.

All conventional PCR reactions were carried out in aPerkin Elmer Thermal Cycler 9600 (Applied Biosystems, Len-nik, Belgium). After denaturation at 95°C for 2 minutes, cycleswere 10 seconds at 94°C, 10 seconds at the optimum annealingtemperature, and 30 seconds at 72°C. Cycling was followed bya 10-minute elongation at 72°C. For BMP-7/osteogenic protein1 (OP-1) PCR, a 2-step procedure with an annealing andelongation temperature of 68°C for 45 seconds was used. PCRproducts were electrophoresed in 1.2% agarose gels in Tris–borate–EDTA electrophoresis buffer, stained with ethidiumbromide, and visualized by ultraviolet transillumination.

For real-time quantitative analysis, PCR was per-formed using the ABI Prism 7700 sequence detection system(Applied Biosystems). PCRs were run in a 12.5-�l mixtureconsisting of 3.5 mM MgCl2, 200 �M of each dNTP, 0.5 �Mspecific primers, 0.025 units/�l HotGoldstar enzyme, 1 �l of

SYBR Green I dilution, and 1� reaction buffer (all fromEurogentec). After initial activation of the HotGoldstar en-zyme (10 minutes at 95°C), 40 cycles of denaturation at 95°Cfor 10 seconds and elongation at 60°C for 2 minutes were run.Nonspecific amplification was detected by melting curve ana-lysis. The identity of the PCR products was checked by DNAsequencing.

For quantitative analysis, the fluorescent signal wasplotted against the cycle number, and the threshold cycle (Ct;the cycle number at which an increase above backgroundfluorescence could be reliably detected) was determined. Se-rial dilutions of positive control cDNA templates were in-cluded in each PCR run, and standard curves for target geneand �-actin were generated by linear regression using log(Ct)versus log(cDNA relative dilution). Each PCR run also in-cluded no-template controls containing all reagents exceptcDNA. In preliminary experiments, the validity of �-actin as ahousekeeping gene was tested by comparing its expressionlevels with those of other housekeeping genes (data notshown). The use of an active reference gene (�-actin) allows usto normalize for differences in the amount of total RNA addedto a reaction and to compensate for different RT efficiencies.All PCR efficiencies were �0.87.

Protein extraction and Western blot analysis. Forsynovial tissue extraction, 2 biopsy samples (combined wetweight of tissue between 11 mg and 39 mg) were extracted in0.5 ml of guanidine HCl buffer (1M guanidine HCl, 50 mMsodium acetate buffered at pH 7.2) overnight at 4°C. Theextract was desalted and concentrated using Microcon YM-10centrifugal spin devices (Micropore, Lancaster, PA) and lyoph-ilized. Proteins were normalized to wet weight by resuspensionin an appropriate volume of sample buffer (50–176 �l) (Nu-PAGE LDS sample buffer; Invitrogen) containing 8M urea.

Aliquots of samples (10 �l) were analyzed underreduced conditions (2% mercaptoethanol and 0.1M dithiothre-itol). Samples were boiled for 5 minutes at 95°C, cooledinstantly, and loaded onto a 4–12% Bis-Tris gel (Invitrogen).Electrophoresis was carried out in a commercially availablerunning buffer (NuPAGE MES SDS running buffer [20�];Invitrogen) at 200V for 40 minutes. Proteins were transferredfrom the gel onto a polyvinylidene difluoride membrane for 90minutes at 30V in a buffer containing 0.4M glycine, 0.5M Trisbase, 0.01M sodium dodecyl sulfate, and 200 ml/liter methanol.Nonspecific binding sites were blocked overnight with 0.5%milk. Blots were probed with the above-described polyclonalantibodies against BMP-2 and BMP-6 and then incubated witha horseradish peroxidase (HRP)–conjugated rabbit anti-

Table 1. Sequences of the primers used for polymerase chain reaction, sizes of the products, and annealing temperatures*

Gene Forward primer Reverse primer

Productsize,bp

Annealingtemperature,

°C

BMP-2 5�-CAGAGACCCACCCCCAGCA-3� 5�-CTGTTTGTGTTTGGCTTGAC-3� 672 60BMP-4 5�-TTCCTGGTAACCGAATGCT-3� 5�-GGGGCTTCATAACCTCATAA-3� 555 60BMP-6 5�-CAGGAGCATCAGCACAGAGAC-3� 5�-ATGTGTGCGTTGAGTGGGAAG-3� 518 60BMP-7/OP-1 5�-GACATCACAGCCACCAGCAAC-3� 5�-GAAGTAGAGGACGGAGATGGC-3� 549 68TGF�1 5�-CAGAAATACAGCAACAATTCCTGG-3� 5�-TTGCAGTGTGTTATCCCTGCTGTC-3� 186 60TGF�2 5�-TCCAAAGATTTAACATCTCCAACC-3� 5�-TCCCACTGTTTTTTTTCCTAGTGG-3� 310 60TGF�3 5�-GCAAATGGCTGAGATGGAGG-3� 5�-GGAAGGCGTCTAACCAAGTG-3� 650 60

* BMP � bone morphogenetic protein; OP-1 � osteogenic protein 1; TGF�1 � transforming growth factor �1.

BMPs IN CHRONIC ARTHRITIS 2809

Page 4: Bone Morphogenetic Proteins 2 and 6, Expressed in Arthritic …ddegroot/SBO_STRETCH/35_Lories_2003.pdf · spondylarthropathy (SpA) and in noninflamed syno-vium from patients undergoing

chicken or rabbit anti-goat secondary antibody (Jackson Im-munoResearch, West Grove, PA). All washes were performedwith Tris buffered saline (TBS)–Tween/0.2% milk except inthe last washing step, when milk was omitted from the washingbuffer. For detection enhancement, 1 ml of chemiluminescentsubstrate (Super signal Pico; Pierce, Rockford, IL) was appliedto the membrane. Blots were visualized on X-Omat Blue XB-1imaging film (Eastman Kodak, Rochester, NY).

Immunohistochemistry. Frozen sections were cut fromTissue-Tek blocks. All 6 biopsy samples obtained at arthros-copy were analyzed on one slide. Silanated slides were air-dried and fixed with acetone for 10 minutes. Sections wereextensively quenched with 0.3% H2O2 in H2O. After washing 3times in TBS for 5 minutes, blocking donkey serum (1:5dilution in TBS) was added for 30 minutes at room tempera-ture. Sections were subsequently incubated with primarychicken anti-human BMP-2 antibody (5 �g/ml) or chicken IgG(Jackson ImmunoResearch) at the same concentration (alldiluted in TBS) at 4°C overnight. After washing 3 times in TBS,a second blocking step was added, and thereafter sections wereincubated for 30 minutes at room temperature with an HRP-conjugated rabbit anti-chicken antibody (1:100 dilution) (Jack-son ImmunoResearch). For BMP-6 staining, a polyclonal goatanti–BMP-6 antibody or goat IgG (Santa Cruz Biotechnology)was used (1:100 dilution). For a secondary antibody, we usedan HRP-conjugated rabbit anti-goat antibody (1:100 dilution)(Jackson ImmunoResearch). Diaminobenzidine (Sigma,

Bornem, Belgium) was used as a chromogen for 5 minutes inthe dark. Sections were washed in distilled water and counter-stained with hematoxylin. For tissue culture slides, the sameprocedure was applied.

Immunofluorescence. For 2-color immunofluores-cence, sections were quenched with 0.025 mg/ml NH4Cl.Blocking of nonspecific binding was done with Blocking Re-agent (Roche Molecular Biochemicals, Brussels, Belgium).For primary antibodies, we used the above-described anti–BMP-2 antibody and a monoclonal mouse anti-human CD68antibody or a monoclonal mouse anti-human CD90 antibody.For secondary antibodies, we used a Cy3-conjugated rabbitanti-chicken antibody (dilution 1:100) and a Cy2-conjugatedgoat anti-mouse antibody (dilution 1:100) (both from JacksonImmunoResearch). Chicken and mouse IgG (Santa CruzBiotechnology) were used as negative controls. Additionalcontrols were performed to exclude cross-species reactivity ofthe secondary antibodies. For nuclear counterstaining, 4�,6-diamidino-2-phenylindole (DAPI; Sigma) was used.

FLS proliferation assay. FLS proliferation rates wereanalyzed using a slightly modified protocol previously de-scribed (25). After trypsin release, FLS were resuspended ingrowth medium at a concentration of 104/ml. Two hundredmicroliters of the cell suspension was plated per well in a96-well flat-bottomed tissue culture plate, and cells wereallowed to attach and start growing for 24 hours. Subsequently,growth arrest was induced by replacing growth medium by

Table 2. Patient characteristics*

Patient/age/sex Disease

Diseaseduration

ESR,mm/hour

CRP level,mg/liter

Synovialfluid

cells/ml DMARDSynovial

vascularitySynovial

hypertrophy

1/22/M Meniscal trauma 9 months MD MD MD No MD MD2/19/F Cartilage injury 1 year MD MD MD No MD MD3/35/M Ligament injury 4 weeks MD MD MD No MD MD4/50/M Ligament injury 1.5 years MD MD MD No MD MD5/53/M Meniscal injury 1 year MD MD MD No MD MD6/48/M Cartilage injury 3 years MD MD MD No MD MD7/44/M Meniscal injury 3 months MD MD MD No MD MD8/45/M Meniscal injury 3 months MD MD MD No MD MD9/58/M RA 6 weeks 31 21.9 13,800 No Moderate Mild

10/52/M RA 6 years 13 8.8 33,300 No Strong Pannus11/54/M RA 6 weeks 100 65.8 900 No Moderate Manifest12/52/F RA 6 weeks 53 96.4 12,500 No Moderate Mild13/30/F RA 6 weeks 53 21 4,100 No Mild Absent14/73/F RA 6 years 87 140 12,100 Gold Strong Pannus15/62/M RA 2 years 2 3 1,200 SSZ Mild Pannus16/62/M RA 15 years MD MD MD MTX Moderate Manifest17/61/F RA 3 years 107 152 13,900 SSZ Moderate Mild18/31/F RA 2 years 49 60 14,400 MTX Moderate Pannus19/31/F SpA 2 months 45 65 3,200 No Mild Mild20/55/M SpA 6 weeks MD MD MD No Mild Mild21/46/M SpA 6 months 41 43 25,200 No Strong Manifest22/72/M SpA 6 weeks 70 156 12,500 No Moderate Manifest23/31/F SpA 5 months 29 19.7 4,200 No Moderate Mild24/35/F SpA 8 years 12 3 4,800 No Moderate Pannus25/32/M SpA 2 years 14 20 100 No Mild Mild26/36/M SpA 9 years 22 19 800 No Mild Mild27/25/F SpA 2 years 20 20 3,600 SSZ Moderate Mild28/55/M SpA 8 years 64 81 8,200 SSZ Moderate Absent

* ESR � erythrocyte sedimentation rate; CRP � C-reactive protein; DMARD � disease-modifying antirheumatic drug; MD � missing data; RA �rheumatoid arthritis; SSZ � sulfasalazine; MTX � methotrexate; SpA � spondylarthropathy.

2810 LORIES ET AL

Page 5: Bone Morphogenetic Proteins 2 and 6, Expressed in Arthritic …ddegroot/SBO_STRETCH/35_Lories_2003.pdf · spondylarthropathy (SpA) and in noninflamed syno-vium from patients undergoing

DMEM–1% FBS–1� antibiotics for 72 hours. The cultureswere supplemented with ligands for 24 hours or 48 hours. Onemicrocurie of 3H-thymidine (ICN Biomedicals, Costa Mesa,CA) was added immediately or after 24 hours. Cells werewashed with DPBS, trypsinized, and resuspended in 200 �l ofserum-free DMEM. The DNA-incorporated radioactivity wasdetermined in a liquid scintillation counter. Alternatively, cellswere lysed after 24 hours or 48 hours of ligand stimulation.DNA content was measured using SYBR Green I fluorescencein a 96-well plate.

FLS apoptosis assay–flow cytometric cell cycle analy-sis. After trypsin release, FLS were plated in 24-well tissueculture plates, grown to confluence in growth medium, andthen kept overnight in DMEM–1% FBS–1� antibiotics. Cellswere subsequently incubated with ligands or carrier controlsfor 4 hours, after which sodium nitroprusside (SNP; Sigma)was added at different concentrations in the appropriateexperimental conditions. Sixteen hours later, both adherentand nonadherent cells were collected and washed 3 times withPBS. Subsequently, 1% Triton X-100 (Sigma) and 100 �g/mlRNase A (Serva, Heidelberg, Germany) were added for 10minutes on ice. Cell DNA was stained with 50 ng/ml propidiumiodide (Sigma). Nuclear DNA content was analyzed on aFACScan (Becton Dickinson). The number of sub-G1 cells in

the DNA histogram was counted. Experiments were per-formed in duplicate or triplicate.

Staining of apoptotic cells. Apoptotic cells in synovialtissue were detected using TUNEL staining. Frozen sectionswere processed for TUNEL assays according to the manufac-turer’s instructions (In Situ Cell Death Detection kit; RocheDiagnostics, Mannheim, Germany). After TUNEL staining,sections were further processed for BMP staining as men-tioned above. Cells were counterstained with DAPI.

Statistical analysis. Comparisons among three ormore groups were made by a nonparametric Friedman analysisof variance (ANOVA), and those between two groups weremade by a nonparametric Wilcoxon test. P values less than 0.05were considered significant.

RESULTS

BMP gene expression in arthritic synovium. Weset out to study the expression of TGF� superfamilymembers from different subfamilies in arthritic syno-vium from patients with RA or SpA and in noninflamedsynovium from patients undergoing diagnostic or thera-peutic arthroscopies. Patient characteristics are shown inTable 2. In preliminary experiments, we found tran-scripts from different BMPs, including BMP-2, BMP-4,BMP-6, and BMP-7/OP-1, to be expressed in synovialtissues. Since BMP-2 and BMP-6 messenger RNA(mRNA) appeared to be most consistently expressed, wefurther studied levels of these BMPs by real-time quan-titative PCR on 15 samples (5 normal synovial tissues

Figure 1. Real-time quantitative reverse transcriptase–polymerasechain reaction analysis of synovial tissue gene expression levels of bonemorphogenetic proteins (BMPs) 2 and 6. Five patients with noninflam-matory joint diseases (normal), 5 with rheumatoid arthritis (RA), and5 with spondylarthropathy (SpA) were studied. Values are the meanand SD arbitrary units, reflecting the target gene expression levelsrelative to the amount of �-actin expressed. 2.0E�05 � 2 � 10�5.

Figure 2. Western blot detection of BMPs 2 and 6 in extracts ofhuman synovial tissue. Synovial tissues were extracted overnight in 0.5ml of 1M guanidine hydrochloride, desalted, lyophilized, dissolved in8M urea–1� sample buffer, and normalized to biopsy sample wetweight. BMP-2 and BMP-6 precursor protein was detected in extractsfrom RA and SpA patients, but not in those from control patientswithout arthritis. See Figure 1 for definitions.

BMPs IN CHRONIC ARTHRITIS 2811

Page 6: Bone Morphogenetic Proteins 2 and 6, Expressed in Arthritic …ddegroot/SBO_STRETCH/35_Lories_2003.pdf · spondylarthropathy (SpA) and in noninflamed syno-vium from patients undergoing

[from patients 1–5], 5 RA tissues [from patients 9–13],and 5 SpA tissues [from patients 19–23]). These BMPsare part of distinct subfamilies with different receptoraffinities (the BMP-2 and BMP-4 subfamily preferen-tially binding activin receptor–like kinase 3 [ALK-3] and

the subfamily that includes BMPs 5, 6, and 7, whichpreferentially bind ALK-2 and/or ALK-6) (10). Nodifferences in mRNA levels of these BMPs were seenbetween the normal and arthritic synovium after nor-malization to the housekeeping gene �-actin (Figure 1).

Figure 3. BMP-2 expression in arthritic synovium. A and B, Immunohistochemical staining showing many BMP-2–positive cells in the lining layerand the sublining layer in synovium from an SpA patient (A) and a few weakly BMP-2–positive cells in synovium from a patient with cartilage injury(B). Bars � 200 �m. C–H, Two-color immunofluorescence staining of the synovium from an RA patient. Shown are BMP-2 staining in thehyperplastic lining layer (C) and sublining zone (F), CD68 staining (D) and superimposed image (E) showing nuclear 4�,6-diamidino-2-phenylindole(DAPI) staining and CD68� cells positive for BMP-2, and CD90 staining (G) and superimposed image (H) showing nuclear DAPI staining andCD90� cells positive for BMP-2. Bars � 25 �m. See Figure 1 for other definitions.

2812 LORIES ET AL

Page 7: Bone Morphogenetic Proteins 2 and 6, Expressed in Arthritic …ddegroot/SBO_STRETCH/35_Lories_2003.pdf · spondylarthropathy (SpA) and in noninflamed syno-vium from patients undergoing

BMP protein expression in arthritic synovium.To study whether BMP proteins were also expressed inthe synovium, we used Western blot. Patient character-istics are shown in Table 2. BMP-2 and BMP-6 precursorproteins were found in synovium from RA patients(patients 14–18) and SpA patients (patients 24–28), butnot in noninflamed synovium from controls (patients6–8) (Figure 2). By extending the time of exposure ofthe chemiluminescent substrate to the film, smallamounts of processed protein could also be detected(results not shown).

We further studied synovial BMP-2 (Figure 3)and BMP-6 expression by immunohistochemistry. BMP-2–positive cells were demonstrated in both RA and SpAsynovium (Figures 3A, C, and F). BMP-2–positive cellswere found in the hyperplastic lining layer and ininfiltrating sublining and perivascular cells. In biopsysamples from the control patients with noninflamedsynovium, only a few scattered BMP-2–positive cellswere found (Figure 3B). Morphologic characteristics ofthe BMP-2–positive cells suggested expression in FLS(type B synoviocytes) as well as in macrophages (type Asynoviocytes). To further characterize BMP-2–expressing cells, we used 2-color immunofluorescencestaining, which showed that both CD90� cells (FLS)and CD68� cells (macrophages) stained for BMP-2(Figures 3C–H). Areas with large lymphoid infiltrateswere mostly BMP-2 negative. Similarly, BMP-6 expres-sion was found both in lining and in sublining layer cells(results not shown). Chicken, goat, or mouse IgG wasused as a negative control for all stainings.

Up-regulation of BMP gene expression in FLScultures by TNF� and IL-1�, but not by IFN�. Wefurther examined the influence of critical proinflamma-tory cytokines on gene expression of BMPs and TGF�sin FLS cultures. FLS from both nonarthritic and arthriticjoints were cultured in 6-well tissue culture plates ingrowth medium until confluence and were then stimu-lated for 24 hours with TNF� (10 ng/ml), IL-1� (10ng/ml), or IFN� (100 units/ml).

As shown by real-time quantitative RT-PCR ana-lysis, both BMP-2 and BMP-6 expression were up-regulated in the presence of TNF� or IL-1� stimulation,but not with IFN� (Figure 4). We consistently foundBMP-2 and BMP-6 up-regulation by IL-1� or TNF� inall FLS cultures, regardless of whether they originatedfrom nonarthritic, RA, or SpA synovium (Figure 4).(Data are from 12 different experiments in which FLSfrom 4 different donors were used.) Treatment with 10ng/ml IL-1� resulted in mean 35.87-fold and 14.50-foldincreases, respectively, in BMP-2 and BMP-6 expressionlevels (both P � 0.01), and incubation with 10 ng/ml

Figure 4. Up-regulation of bone morphogenetic protein 2 (BMP-2)and BMP-6 gene expression, but not transforming growth factor �1(TGF�1) gene expression, by tumor necrosis factor � (TNF�) andinterleukin-1� (IL-1�), but not by interferon-� (IFN�). Fibroblast-likesynoviocytes at confluence were further cultured for 24 hours withgrowth medium or were stimulated for 24 hours with TNF� (10 ng/ml),IL-1� (10 ng/ml), or IFN� (100 units/ml). Gene expression of BMP-2(A), BMP-6 (B), and TGF�1 (C) were studied by real-time quantitativereverse transcriptase–polymerase chain reaction. Gene expressionlevels were normalized to the expression of the housekeeping gene�-actin and compared with those of unstimulated controls. Water wasused as a template negative control. Values are the mean and SEMfold increase in gene expression as compared with nonstimulatedcontrols. Results are from 12 different experiments using cells from 4different donors. � � P � 0.01; �� � P � 0.05 versus unstimulatedcontrols.

BMPs IN CHRONIC ARTHRITIS 2813

Page 8: Bone Morphogenetic Proteins 2 and 6, Expressed in Arthritic …ddegroot/SBO_STRETCH/35_Lories_2003.pdf · spondylarthropathy (SpA) and in noninflamed syno-vium from patients undergoing

TNF� resulted in mean 17.24-fold and 5.21-fold in-creases, respectively, in BMP-2 and BMP-6 expressionlevels (P � 0.01 and P � 0.05, respectively) (Figure 4).BMP up-regulation by IL-1� and TNF� was dose de-pendent (data not shown). We also studied BMP-4 andBMP-7 regulation by TNF� and IL-1�. No up-regulation of BMP-4 or BMP-7 was seen (data notshown). Overall expression levels of BMP-7 were verylow. No significant effects on expression levels of TGF�1(Figure 4) or on expression levels of TGF�2 or TGF�3(data not shown) were seen.

To extend these findings, BMP-2 and BMP-6proteins were examined in FLS cultured on tissue cul-ture slides. FLS were treated with IL-1� (10 ng/ml) for24 hours. BMP-2 and BMP-6 proteins were clearlydetected using immunohistochemistry (Figure 5).

FLS proliferation stimulated by TGF�, but notby BMP. We studied the influence of BMPs and TGF�on FLS proliferation in vitro after serum deprivation–induced growth arrest. FLS from 5 different patients (1with OA, 2 with RA, and 2 with SpA) were studied, all

Figure 5. Up-regulation of BMP-2 and BMP-6 protein expression by IL-1� in fibroblast-like synoviocytes (FLS). Normal FLSwere cultured on tissue culture slides. At confluence, FLS were cultured in growth medium alone (A and C) or were stimulatedfor 24 hours with 10 ng/ml IL-1� (B and D). BMP-2 (A and B) and BMP-6 (C and D) were visualized by immunohistochemistry.Bars � 25 �m. See Figure 4 for other definitions.

Figure 6. Effects of TGF�1 and BMPs 2 and 6 on fibroblast-likesynoviocyte (FLS) proliferation. FLS proliferation was studied by3H-thymidine incorporation after serum deprivation–induced growtharrest. After 24 hours of serum deprivation (1% serum), cells werestimulated with BMP-2 (100 ng/ml), BMP-6 (100 ng/ml), or TGF�1 (10ng/ml) for 48 hours. During the last 24 hours, 1 �Ci 3H-thymidine wasadded to the cultures. Values are the mean and SD. FLS from 5different patients were studied. � � P � 0.05 versus unstimulatedcontrols. OA � osteoarthritis; RA � rheumatoid arthritis; SpA �spondylarthropathy (see Figure 4 for other definitions).

2814 LORIES ET AL

Page 9: Bone Morphogenetic Proteins 2 and 6, Expressed in Arthritic …ddegroot/SBO_STRETCH/35_Lories_2003.pdf · spondylarthropathy (SpA) and in noninflamed syno-vium from patients undergoing

between passages 4 and 7. We studied both early (first 24hours) and late (second 24 hours) effects on prolifera-tion. In contrast to TGF�1 (10 ng/ml), which consis-tently increased FLS proliferation after 48 hours (Figure6), but not after 24 hours (data not shown), no effect ofBMP-2 (100 ng/ml) or BMP-6 (100 ng/ml) was seen at 24hours (data not shown) or 48 hours (Figure 6). Nodifferences were seen with either dosage variation ofBMPs or with combinations of BMPs and TGF�1 (datanot shown). We confirmed these data obtained by3H-thymidine incorporation by measuring DNA contentafter 24 hours and 48 hours (data not shown).

FLS apoptosis promoted by BMP-2, protectionagainst nitric oxide (NO)–induced apoptosis conferredby BMP-6. We further tested the effects of BMP-2 andBMP-6 on FLS apoptosis in vitro. FLS cultures atconfluence were incubated with BMP-2 or BMP-6 for 20hours. Additionally, cells were pretreated with BMP-2 orBMP-6 for 4 hours, after which SNP (an NO liberator)was added to the cultures for 16 hours. FLS DNAhistograms were analyzed by flow cytometry. BMP-2(100 ng/ml) consistently increased the number of apo-ptotic cells in our in vitro cultures (P � 0.003 byFriedman ANOVA; P � 0.02 by nonparametric Wil-coxon test) (Figure 7A). In contrast, BMP-6 (100 ng/ml)

Figure 7. Different effects of bone morphogenetic protein 2 (BMP-2)and BMP-6 on fibroblast-like synoviocyte (FLS) apoptosis. FLS apo-ptosis was studied by flow cytometry with propidium iodide cell cycleanalysis. A, Cells were stimulated with or without BMP-2 (100 ng/ml)or BMP-6 (100 ng/ml) for 20 hours. � � P � 0.05 versus unstimulatedcontrols. B, Cells were pretreated with BMP-2 (100 ng/ml), BMP-6(100 ng/ml), or carrier control. After 4 hours, sodium nitroprusside(SNP) was added to the cultures for 16 hours. In each experiment, theconcentration of SNP used for analysis was chosen in such a way thata 15% increase in apoptotic cells was seen 16 hours after adding SNP.�� � P � 0.05 versus SNP-stimulated cells. Data from 7 differentexperiments are shown. Horizontal bars are medians.

Figure 8. Expression of bone morphogenetic protein 2 (BMP-2) insynoviocytes undergoing apoptosis. A, Double staining showing BMP-2–positive (brown) and TUNEL-positive (blue) cells. Several double-positive cells are seen. B, Superimposed image showing nuclear4�,6-diamidino-2-phenylindole staining, cytoplasmic BMP-2 positivity,and nuclear TUNEL positivity. Bars � 25 �m.

BMPs IN CHRONIC ARTHRITIS 2815

Page 10: Bone Morphogenetic Proteins 2 and 6, Expressed in Arthritic …ddegroot/SBO_STRETCH/35_Lories_2003.pdf · spondylarthropathy (SpA) and in noninflamed syno-vium from patients undergoing

stimulation had no direct effect on FLS apoptosis.However, BMP-6 pretreatment for 4 hours protectedFLS from NO-induced apoptosis (P � 0.004 by Fried-man ANOVA; P � 0.02 by nonparametric Wilcoxontest) (Figure 7B). Appropriate concentrations of SNP(the NO liberator) were chosen, resulting in an increaseof �15% in apoptotic cells compared with controls. Toshow specificity of the effect on FLS and to exclude anonspecific cytotoxic effect of our BMP-2 preparation,we also treated myogenic cell line C2C12 with BMP-2.As recently described, BMP-2 had no direct effect onC2C12 apoptosis, but protected against TNF�-inducedcell death (data not shown) (26).

Expression of BMP-2 in synoviocytes undergoingapoptosis. To examine whether the in vitro effect ofBMP-2 on synoviocyte apoptosis may be relevant in vivo,we used immunohistochemistry to study BMP-2 expres-sion in cells undergoing apoptosis (Figure 8). We foundBMP-2– and TUNEL-positive cells in both the liningand sublining layer of arthritic synovium. We also foundBMP-2–positive, TUNEL-negative cells and BMP-2–negative, TUNEL-positive cells. Our data suggest thatBMP-2 expression may precede the process of apoptosis.In the double-positive cells, TUNEL positivity was notcovering the whole nucleus. In later stages of apoptosis,the whole cell nucleus was TUNEL positive, and little orno BMP-2 staining was seen, probably due to progres-sion of apoptosis and shrinkage of the cell cytoplasm(Figure 8). Normal IgG were used as negative controlsfor the double staining.

DISCUSSION

In this study, we demonstrated the expressionand presence of specific BMPs in arthritic synovium byRT-PCR and immunohistochemistry. Real-time quanti-tative RT-PCR for BMP-2 and BMP-6 did not showsignificant differences between normal and arthriticsynovium. Although PCR techniques are extremely sen-sitive tools for detecting small amounts of mRNA orDNA, the need to normalize gene expression levels tothose of a housekeeping gene limits their value in theanalysis of complex tissues such as the synovium. Cellu-larity and cell types may be different from patient topatient due to differences in disease status, disease type,and random sampling. Housekeeping gene expressionlevels show a large variability between different celltypes and therefore often do not allow for a simplecomparison. Similarly, alternative normalization to totalRNA transcribed in the RT reaction is also influencedby cell types and cellularity as well as by the potentialpresence of inhibitors of the RT-PCR reaction. In

Western blot, however, both BMP-2 and BMP-6 precur-sor protein was present in extracts from arthritic syno-vium, but not in extracts obtained from the synovium ofcontrol patients. Our immunohistochemistry studies cor-roborate these findings, showing more BMP-2– andBMP-6–positive cells in arthritic synovium than in non-arthritic control synovium.

Two-color immunofluorescence demonstratedBMP-2 and BMP-6 expression both in type A synovio-cytes (macrophages) and in type B synoviocytes (FLS).The expression of BMPs in cells of mesenchymal origin(FLS) as well as in those of hematopoietic origin was notsurprising, since BMPs have been reported to play a rolein mesenchyme differentiation (15) and hematopoiesis(27). The spatial distribution of BMP-2– and BMP-6–producing cells in the inflamed synovium does notprovide specific clues about potential targets. In addi-tion, BMPs are secreted proteins and are thus expectedto work both in an autocrine and in a paracrine manner.

Up-regulation of BMP-2 mRNA by IL-1� in OAFLS has previously been reported (28). We extendedthese findings and added novel information by demon-strating a striking up-regulation of BMP-2 and BMP-6(but not of BMP-4) by the proinflammatory cytokinesTNF� and IL-1� (but not by IFN�). This selectivemodulation suggests a relevant function for these BMPsas autocrine or paracrine peptides in the pathologicmicroenvironment of chronic arthritis. It is clear thatBMPs and proinflammatory cytokines such as IL-1 andTNF� are part of a tightly controlled network, suggest-ing that local balances and autocrine or paracrine ef-fects, rather than the overall amount of protein, are thedeterminants of cell fate and function.

It remains unclear whether BMPs are part of thesynovial defense and repair mechanisms, or whetherthey play a role in disease pathogenesis. Specific BMPsare strongly up-regulated in response to proinflamma-tory cytokines in vitro. We hypothesize that any tissuedamage elicits a response from the body’s inherentdefense and repair capacity. Reactivation of embryonicsignaling pathways essential for tissue growth and differ-entiation during morphogenesis is likely to be such amechanism (11). Such phenomena, involving BMPs,have been clearly demonstrated in fracture healing (29).

Cartilage-derived morphogenetic proteins, whichare also members of the BMP family, are expressed inthe superficial layers of the articular cartilage in normalindividuals (30). In OA, however, their expression ex-tends into the deeper cartilage tissue layers of thedamaged areas. Their expression is also up-regulatedafter trypsin treatment, resulting in matrix depletion(30). Local overexpression of BMPs may provide a

2816 LORIES ET AL

Page 11: Bone Morphogenetic Proteins 2 and 6, Expressed in Arthritic …ddegroot/SBO_STRETCH/35_Lories_2003.pdf · spondylarthropathy (SpA) and in noninflamed syno-vium from patients undergoing

metabolic stimulus for chondrocytes and osteoblasts orosteocytes at the site of damage, increasing local resis-tance. Osteophyte formation in OA may be a partiallyBMP-driven repair/remodeling effort to increase jointstability after cartilage damage has occurred. Injectionof TGF�1 or BMP-2 into murine joints results inosteophyte formation (19).

BMP-2 has also been shown to up-regulate os-teoprotegerin in osteoblastic cells, thereby providing anegative signal for osteoclast differentiation and activa-tion (31,32). However, up-regulation of both receptoractivator of NF-�B ligand and cyclooxygenase 2 inosteoblast-like cells has been reported to contribute toosteoclastogenesis in coculture experiments ofosteoblast-like cells and hematopoietic cells (33). It isnoteworthy that BMP-4 has been reported to act as astrong chemotactic agent for mononuclear cells (34).Local BMP production within the synovium may there-fore also contribute to disease pathogenesis and progres-sion.

Although there is striking structural similaritybetween all members of the superfamily, biologic effectsare diverse. Different ligand–receptor affinities, targetcell receptor expression, specific intracellular pathwayactivation, and interactions of the DNA transcriptioncomplex with activators and inhibitors define specificityand diversity in signaling (35). In view of this, thecontrasting effects of BMP-2, BMP-6, and TGF�1 onFLS apoptosis and proliferation are not surprising.

Different BMPs have been associated with apo-ptosis in development (36,37). BMP-2 and the stronglyrelated BMP-4 have been described as proapoptoticmolecules in different in vitro settings (38–40). How-ever, in contrast to its effects on osteoblasts (38), BMP-2has also been reported to inhibit apoptosis in cardiacmyocytes (41). BMP-6 protects cortical neural cells invitro against H2O2-induced cell death and reduces brainischemia/reperfusion injury in rats (42). In arthritis,BMP-2–induced apoptosis may be a protective mecha-nism for controlling extensive and aggressive pannusproliferation. The antiapoptotic effect of BMP-6 is notinconsistent with this protective mechanism, since it mayprotect the synovium from excessive cell death due toaccumulation of NO or reactive oxygen molecules. Inthis respect, the identification of BMP receptor type IA(BMPRIA)–positive cells in arthritic synovium (8) andtheir potential role in RA pathogenesis may define aspecific target cell for BMP-2 signaling in arthritis. It isnoteworthy that such cells were not found in OA syno-vium (6). In the absence of BMPRIB (ALK-6) expres-sion in synovium, as demonstrated by RT-PCR (data notshown), BMP-6 signaling would preferably proceed

through activin receptor type Ia (ALK-2) (43). We havenot yet identified a specific target cell in the inflamedsynovium for BMP-6 signaling, although we are continu-ing research in this area.

In conclusion, we believe that this is the firstreport to describe synovial expression of BMP and someof its potential roles in the arthritic synovium. Furtherresearch is now warranted to assess the protective and/orpathogenetic role of distinct BMPs in specific forms ofarthritis. We propose that BMPs may further tilt thebalance between destructive and reparative mechanismsin the joint in favor of tissue repair, not only bypromoting cartilage and bone formation, but also bymodulating FLS cell populations in the inflamed syno-vium.

ACKNOWLEDGMENTS

The authors wish to thank all collaborators at theLaboratory for Skeletal Development and Joint Disorders forsupport and useful discussions. We also thank Drs. ReneWesthovens, Kurt De Vlam, Patrick Verschueren, and JohanVanlauwe for providing biopsy materials.

REFERENCES

1. Maini RN, Breedveld FC, Kalden JR, Smolen JS, Davis D,Macfarlane JD, et al. Therapeutic efficacy of multiple intravenousinfusions of anti–tumor necrosis factor � monoclonal antibodycombined with low-dose weekly methotrexate in rheumatoid ar-thritis. Arthritis Rheum 1998;41:1552–63.

2. Braun J, Brandt J, Listing J, Zink A, Alten R, Golder W, et al.Treatment of active ankylosing spondylitis with infliximab: arandomised controlled multicentre trial. Lancet 2002;359:1187–93.

3. Panayi GS, Corrigall VM, Pitzalis C. Pathogenesis of rheumatoidarthritis. The role of T cells and other beasts. Rheum Dis ClinNorth Am 2001;27:317–34.

4. Takemura S, Klimiuk PA, Braun A, Goronzy JJ, Weyand CM. Tcell activation in rheumatoid synovium is B cell dependent.J Immunol 2001;167:4710–8.

5. Firestein GS, Zvaifler NJ. How important are T cells in chronicrheumatoid synovitis? II. T cell–independent mechanisms frombeginning to end. Arthritis Rheum 2002;46:298–308.

6. Firestein GS. Invasive fibroblast-like synoviocytes in rheumatoidarthritis: passive responders or transformed aggressors? ArthritisRheum 1996;39:1781–90.

7. Kinne RW, Liehr T, Beensen V, Kunisch E, Zimmermann T,Holland H, et al. Mosaic chromosomal aberrations in synovialfibroblasts of patients with rheumatoid arthritis, osteoarthritis, andother inflammatory joint diseases. Arthritis Res 2001;3:319–30.

8. Marinova-Mutafchieva L, Taylor P, Funa K, Maini RN, ZvaiflerNJ. Mesenchymal cells expressing bone morphogenetic proteinreceptors are present in the rheumatoid arthritis joint. ArthritisRheum 2000;43:2046–55.

9. De Bari C, Dell’Accio F, Tylzanowski P, Luyten FP. Multipotentmesenchymal stem cells from adult human synovial membrane.Arthritis Rheum 2001;44:1928–42.

10. Luyten FP, Dell’Accio F, De Bari C. Skeletal tissue engineering:opportunities and challenges. Best Pract Res Clin Rheumatol2001;15:759–69.

11. Luyten FP, Lories RJ, De Bari C, Devalck D, Dell’Accio F. Bonemorphogenetic proteins and the synovial joint. In: Vukicevic S,

BMPs IN CHRONIC ARTHRITIS 2817

Page 12: Bone Morphogenetic Proteins 2 and 6, Expressed in Arthritic …ddegroot/SBO_STRETCH/35_Lories_2003.pdf · spondylarthropathy (SpA) and in noninflamed syno-vium from patients undergoing

Sampath KT, editors. Progress in inflammation research. Basel(Switzerland): Birckhauser Verlag; 2002. p. 223–48.

12. Massague J. TGF-beta signal transduction. Annu Rev Biochem1998;67:753–91.

13. Urist MR. Bone: formation by autoinduction. Science 1965;150:893–9.

14. Wozney JM, Rosen V, Celeste AJ, Mitsock LM, Whitters MJ, KrizRW, et al. Novel regulators of bone formation: molecular clonesand activities. Science 1988;242:1528–34.

15. Hogan BL. Bone morphogenetic proteins in development. CurrOpin Genet Dev 1996;6:432–8.

16. Wozney JM, Rosen V. Bone morphogenetic protein and bonemorphogenetic protein gene family in bone formation and repair.Clin Orthop 1998;26–37.

17. Nakase T, Nomura S, Yoshikawa H, Hashimoto J, Hirota S,Kitamura Y, et al. Transient and localized expression of bonemorphogenetic protein 4 messenger RNA during fracture healing.J Bone Miner Res 1994;9:651–9.

18. Yang X, Chen L, Xu X, Li C, Huang C, Deng CX. TGF-beta/Smad3 signals repress chondrocyte hypertrophic differentiationand are required for maintaining articular cartilage. J Cell Biol2001;153:35–46.

19. Van Beuningen HM, Glansbeek HL, van der Kraan PM, van denBerg WB. Differential effects of local application of BMP-2 orTGF-beta 1 on both articular cartilage composition and osteo-phyte formation. Osteoarthritis Cartilage 1998;6:306–17.

20. Scharstuhl A, Glansbeek HL, van Beuningen HM, Vitters EL, vander Kraan PM, van den Berg WB. Inhibition of endogenousTGF-� during experimental osteoarthritis prevents osteophyteformation and impairs cartilage repair. J Immunol 2002;169:507–14.

21. Arnett FC, Edworthy SM, Bloch DA, McShane DJ, Fries JF,Cooper NS, et al. The American Rheumatism Association 1987revised criteria for the classification of rheumatoid arthritis.Arthritis Rheum 1988;31:315–24.

22. Dougados M, van der Linden S, Juhlin R, Huitfeldt B, Amor B,Calin A, et al, and the European Spondylarthropathy StudyGroup. The European Spondylarthropathy Study Group prelimi-nary criteria for the classification of spondylarthropathy. ArthritisRheum 1991;34:1218–30.

23. Altman R, Asch E, Bloch D, Bole G, Borenstein D, Brandt K, etal. Development of criteria for the classification and reporting ofosteoarthritis: classification of osteoarthritis of the knee. ArthritisRheum 1986;29:1039–49.

24. Baeten D, van Den Bosch F, Elewaut D, Stuer A, Veys EM, deKeyser F. Needle arthroscopy of the knee with synovial biopsysampling: technical experience in 150 patients. Clin Rheumatol1999;18:434–41.

25. Zimmermann T, Kunisch E, Pfeiffer R, Hirth A, Stahl H, Sack U,et al. Isolation and characterization of rheumatoid arthritis syno-vial fibroblasts from primary culture—primary culture cells mark-edly differ from fourth-passage cells. Arthritis Res 2001;3:72–6.

26. Chen S, Guttridge DC, Tang E, Shi S, Guan K, Wang CY.Suppression of tumor necrosis factor-mediated apoptosis by nu-clear factor kappaB-independent bone morphogenetic protein/Smad signaling. J Biol Chem 2001;276:39259–63.

27. Bhatia M, Bonnet D, Wu D, Murdoch B, Wrana J, Gallacher L, etal. Bone morphogenetic proteins regulate the developmentalprogram of human hematopoietic stem cells. J Exp Med 1999;189:1139–48.

28. Fowler MJ Jr, Neff MS, Borghaei RC, Pease EA, Mochan E,Thornton RD. Induction of bone morphogenetic protein-2 byinterleukin-1 in human fibroblasts. Biochem Biophys Res Com-mun 1998;248:450–3.

29. Ferguson CM, Miclau T, Hu D, Alpern E, Helms JA. Commonmolecular pathways in skeletal morphogenesis and repair. Ann NY Acad Sci 1998;857:33–42.

30. Erlacher L, Ng C-K, Ullrich R, Krieger S, Luyten FP. Presence ofcartilage-derived morphogenetic proteins in articular cartilage andenhancement of matrix replacement in vitro. Arthritis Rheum1998;41:263–73.

31. Wan M, Shi X, Feng X, Cao X. Transcriptional mechanisms ofbone morphogenetic protein-induced osteoprotegerin gene ex-pression. J Biol Chem 2001;276:10119–25.

32. Hofbauer LC, Dunstan CR, Spelsberg TC, Riggs BL, Khosla S.Osteoprotegerin production by human osteoblast lineage cells isstimulated by vitamin D, bone morphogenetic protein-2, andcytokines. Biochem Biophys Res Commun 1998;250:776–81.

33. Koide M, Murase Y, Yamato K, Noguchi T, Okahashi N, Nishi-hara T. Bone morphogenetic protein-2 enhances osteoclast forma-tion mediated by interleukin-1alpha through upregulation of oste-oclast differentiation factor and cyclooxygenase-2. BiochemBiophys Res Commun 1999;259:97–102.

34. Cunningham NS, Paralkar V, Reddi AH. Osteogenin and recom-binant bone morphogenetic protein 2B are chemotactic for humanmonocytes and stimulate transforming growth factor beta 1mRNA expression. Proc Natl Acad Sci U S A 1992;89:11740–4.

35. Massague J. How cells read TGF-beta signals. Nat Rev Mol CellBiol 2000;1:169–78.

36. Graham A, Francis-West P, Brickell P, Lumsden A. The signallingmolecule BMP4 mediates apoptosis in the rhombencephalic neu-ral crest. Nature 1994;372:684–6.

37. Macias D, Ganan Y, Sampath TK, Piedra ME, Ros MA, HurleJM. Role of BMP-2 and OP-1 (BMP-7) in programmed cell deathand skeletogenesis during chick limb development. Development1997;124:1109–17.

38. Hay E, Lemonnier J, Fromigue O, Marie PJ. Bone morphogeneticprotein-2 promotes osteoblast apoptosis through a Smad-indepen-dent, protein kinase C-dependent signaling pathway. J Biol Chem2001;276:29028–36.

39. Kawamura C, Kizaki M, Yamato K, Uchida H, Fukuchi Y, HattoriY, et al. Bone morphogenetic protein-2 induces apoptosis inhuman myeloma cells with modulation of STAT3. Blood 2000;96:2005–11.

40. Kimura N, Matsuo R, Shibuya H, Nakashima K, Taga T. BMP2-induced apoptosis is mediated by activation of the TAK1-p38kinase pathway that is negatively regulated by Smad6. J Biol Chem2000;275:17647–52.

41. Izumi M, Fujio Y, Kunisada K, Negoro S, Tone E, Funamoto M,et al. Bone morphogenetic protein-2 inhibits serum deprivation-induced apoptosis of neonatal cardiac myocytes through activationof the Smad1 pathway. J Biol Chem 2001;276:31133–41.

42. Wang Y, Chang CF, Morales M, Chou J, Chen HL, Chiang YH, etal. Bone morphogenetic protein-6 reduces ischemia-induced braindamage in rats. Stroke 2001;32:2170–8.

43. Ebisawa T, Tada K, Kitajima I, Tojo K, Sampath TK, Kawabata M,et al. Characterization of bone morphogenetic protein-6 signalingpathways in osteoblast differentiation. J Cell Sci 1999;112(Pt20):3519–27.

2818 LORIES ET AL