+ All Categories
Home > Documents > Proteasome Inhibition with Bortezomib Depletes Plasma Cells and Autoantibodies in Experimental...

Proteasome Inhibition with Bortezomib Depletes Plasma Cells and Autoantibodies in Experimental...

Date post: 29-Apr-2023
Category:
Upload: independent
View: 0 times
Download: 0 times
Share this document with a friend
12
of October 4, 2011 This information is current as http://www.jimmunol.org/content/186/4/2503 doi:10.4049/jimmunol.1002539 January 2011; 2011;186;2503-2513; Prepublished online 14 J Immunol E. Voll, Rudolf A. Manz, Marc H. De Baets and Mario Losen Eline van der Esch, Hans Duimel, Fons Verheyen, Reinhard Martínez-Martínez, Peter C. Molenaar, Marko Phernambucq, Alejandro M. Gomez, Kathleen Vrolix, Pilar Autoimmune Myasthenia Gravis Experimental Plasma Cells and Autoantibodies in Proteasome Inhibition with Bortezomib Depletes References http://www.jimmunol.org/content/186/4/2503.full.html#ref-list-1 , 16 of which can be accessed free at: cites 44 articles This article Subscriptions http://www.jimmunol.org/subscriptions is online at The Journal of Immunology Information about subscribing to Permissions http://www.aai.org/ji/copyright.html Submit copyright permission requests at Email Alerts http://www.jimmunol.org/etoc/subscriptions.shtml/ Receive free email-alerts when new articles cite this article. Sign up at Print ISSN: 0022-1767 Online ISSN: 1550-6606. Immunologists, Inc. All rights reserved. by The American Association of Copyright ©2011 9650 Rockville Pike, Bethesda, MD 20814-3994. The American Association of Immunologists, Inc., is published twice each month by The Journal of Immunology on October 4, 2011 www.jimmunol.org Downloaded from
Transcript

of October 4, 2011This information is current as

http://www.jimmunol.org/content/186/4/2503doi:10.4049/jimmunol.1002539January 2011;

2011;186;2503-2513; Prepublished online 14J Immunol E. Voll, Rudolf A. Manz, Marc H. De Baets and Mario LosenEline van der Esch, Hans Duimel, Fons Verheyen, ReinhardMartínez-Martínez, Peter C. Molenaar, Marko Phernambucq, Alejandro M. Gomez, Kathleen Vrolix, Pilar Autoimmune Myasthenia Gravis

ExperimentalPlasma Cells and Autoantibodies in Proteasome Inhibition with Bortezomib Depletes

References http://www.jimmunol.org/content/186/4/2503.full.html#ref-list-1

, 16 of which can be accessed free at:cites 44 articlesThis article

Subscriptions http://www.jimmunol.org/subscriptions

is online atThe Journal of ImmunologyInformation about subscribing to

Permissions http://www.aai.org/ji/copyright.html

Submit copyright permission requests at

Email Alerts http://www.jimmunol.org/etoc/subscriptions.shtml/

Receive free email-alerts when new articles cite this article. Sign up at

Print ISSN: 0022-1767 Online ISSN: 1550-6606.Immunologists, Inc. All rights reserved.

by The American Association ofCopyright ©2011 9650 Rockville Pike, Bethesda, MD 20814-3994.The American Association of Immunologists, Inc.,

is published twice each month byThe Journal of Immunology

on October 4, 2011

ww

w.jim

munol.org

Dow

nloaded from

The Journal of Immunology

Proteasome Inhibition with Bortezomib Depletes Plasma Cellsand Autoantibodies in Experimental Autoimmune MyastheniaGravis

Alejandro M. Gomez,* Kathleen Vrolix,* Pilar Martınez-Martınez,* Peter C. Molenaar,*

Marko Phernambucq,* Eline van der Esch,*,† Hans Duimel,† Fons Verheyen,†

Reinhard E. Voll,‡ Rudolf A. Manz,x Marc H. De Baets,*,{ and Mario Losen*

Bortezomib, an inhibitor of proteasomes, has been reported to reduce autoantibody titers and to improve clinical condition in mice

suffering from lupus-like disease. Bortezomib depletes both short- and long-lived plasma cells; the latter normally survive the

standard immunosuppressant treatments targeting T and B cells. These findings encouraged us to test whether bortezomib is ef-

fective for alleviating the symptoms in the experimental autoimmune myasthenia gravis (EAMG) model for myasthenia gravis,

a disease that is characterized by autoantibodies against the acetylcholine receptor (AChR) of skeletal muscle. Lewis rats were

immunized with saline (control, n = 36) or Torpedo AChR (EAMG, n = 54) in CFA in the first week of an experimental period

of 8 wk. After immunization, rats received twice a week s.c. injections of bortezomib (0.2 mg/kg in saline) or saline injections.

Bortezomib induced apoptosis in bone marrow cells and reduced the amount of plasma cells in the bone marrow by up to 81%. In

the EAMG animals, bortezomib efficiently reduced the rise of anti-AChR autoantibody titers, prevented ultrastructural damage

of the postsynaptic membrane, improved neuromuscular transmission, and decreased myasthenic symptoms. This study thus

underscores the potential of the therapeutic use of proteasome inhibitors to target plasma cells in Ab-mediated autoimmune

diseases. The Journal of Immunology, 2011, 186: 2503–2513.

The resistance of long-lived plasma cells against immu-nosuppressive medication poses a serious problem for thetreatment of Ab-mediated autoimmune diseases. Currently

used immunosuppressive drugs, including corticosteroids, mito-mycin C, cyclosporine A, azathioprine, and cyclosphosphamide,affect mainly activated and dividing B and/or T cells (1). Plasmacells are the terminally differentiated, nondividing effector cells ofthe B cell lineage that have lost many surface markers. In theirsurvival niches in the spleen and in particular in the bone marrow(2), resident long-lived plasma cells are resistant to most therapies,

including immunosuppressive drugs and anti-CD20 Abs that areaimed to inhibit the activation and/or proliferation of lymphocytesor to deplete certain lymphocyte subpopulations (3, 4). Thus, re-sistance to available therapies might be due to persistent long-lived plasma cells that continue to produce autoantibodies not-withstanding immunosuppressive treatment (5).However, because of their high rate of Ig production, both short-

and long-lived plasma cells are particularly sensitive to inhibitionof the proteasome (6, 7). Indeed, proteasome inhibition causesaccumulation of nondegraded, misfolded proteins within theendoplasmic reticulum of plasma cells and, subsequently, to acti-vation of the terminal unfolded protein response, ultimately lead-ing to apoptosis (8). The proteasome inhibitor bortezomib, alsoknown under the trade name Velcade, is a boronic acid dipeptide(phenylalanine-leucine) derivative, which binds reversibly to the 26Sproteasome (9). After injection, bortezomib is distributed widely andquickly to the blood and most tissues (10). Currently, bortezomib isapproved for the treatment of multiple myeloma and mantle celllymphoma. In addition to the treatment of B cell malignancies,proteasome inhibition could be a useful therapeutic strategy for Ab-mediated autoimmunity such as lupus (7).In the present study we tested the effect of proteasome inhibition

in a model for myasthenia gravis (MG), a well-characterized dis-ease that is found to be critically dependent on the level of auto-antibodies against the acetylcholine receptors (AChRs) of muscle.In 85% of MG patients the disease is caused by autoantibodiesagainst the muscle AChR (11). The remaining patients haveautoantibodies against the muscle-specific kinase (12) (∼5% ofall MG patients) or no detectable autoantibodies (idiopathicMG, accounting for ∼10% of all patients) (13). Both AChR andmuscle-specific kinase proteins are located in the postsynapticmembrane of the neuromuscular junction (NMJ), which is spe-cialized to respond to the neurotransmitter ACh released from the

*NeuroimmunologyGroup, Department ofNeuroscience, School ofMental Health andNeuroscience, Maastricht University, Maastricht, The Netherlands; †Electron Micros-copy Unit, Department of Molecular Cell Biology, Maastricht University, Maastricht,The Netherlands; ‡Department of Internal Medicine 3, Nikolaus Fiebiger-Center ofMolecular Medicine, University Hospital Erlangen, Erlangen, Germany; xInstitute forSystemic Inflammation Research, University of Lubeck, Lubeck, Germany; and {Neu-roimmunologyGroup, Biomedical Research Institute, Hasselt University, Diepenbeek,Belgium

Received for publication July 30, 2010. Accepted for publication December 1, 2010.

This work was supported by the European Union Sixth Framework Program (FP6)MYASTAID LSHM-CT-2006-037833. A.M.G. was supported by a Marie-Curie fel-lowship by the European Union and a grant from the Prinses Beatrix Fonds (ProjectWAR08-12). P.M.-M. was supported by grants from the Prinses Beatrix Fonds (Pro-ject MAR03-0115), L’Association Francaise contre les Myopathies, and Genmab.M.L. was supported by a Veni fellowship of The Netherlands Organization forScientific Research and a fellowship of the Brain Foundation of The Netherlands.

Address correspondence and reprint requests to Dr. Mario Losen, Department ofNeuroscience, School of Mental Health and Neuroscience, Maastricht University,P.O. Box 616, 6200 MD Maastricht, The Netherlands. E-mail address: [email protected]

Abbreviations used in this article: AChR, acetylcholine receptor; AMC, 7-amino-4-methylcoumarin; CMAP, compound muscle action potential; EAMG, experimentalautoimmune myasthenia gravis; EM, electron microscopy; EMG, electromyography;IF, immunofluorescence; MG, myasthenia gravis; NMJ, neuromuscular junction; PI,propidium iodide; tAChR, acetylcholine receptor from Torpedo californica.

Copyright� 2011 by The American Association of Immunologists, Inc. 0022-1767/11/$16.00

www.jimmunol.org/cgi/doi/10.4049/jimmunol.1002539

on October 4, 2011

ww

w.jim

munol.org

Dow

nloaded from

overlying nerve ending. These proteins are thus essential formuscle contraction, and MG is potentially fatal, since autoanti-bodies can cause respiratory failure by impairing neuromusculartransmission. In such an event of acute myasthenic crisis, the firstchoice of treatment is plasma exchange, mechanical ventilation(14), and i.v. treatment with high doses of IgG (15). Plasma ex-change typically leads to significant improvement or remissionwithin a few days in most patients, including those with the idi-opathic MG, but the effect is, of course, not long-lasting. Immu-nosuppressive drugs such as prednisone and azathioprine aregenerally used for long-term therapy (1, 16–18). Because thesedepend mostly on preventing the activation, proliferation, anddifferentiation of developing B and T cells, the autoantibodytiters only drop over a period of months. Using an establishedimmunotherapy protocol with prednisone and azathioprine, itmay take as much as 18 mo before patients improve (16). For theintermediate time interval, during which plasma cells continueto produce autoantibodies, proteasome inhibition might bea useful therapy. Moreover, because some MG patients do notrespond well to any currently available treatment in terms ofpoor reduction of autoantibodies or the occurrence of side ef-fects, proteasome inhibition might provide a therapeutic al-ternative.In the experimental autoimmune myasthenia gravis (EAMG)

model the disease is induced by immunizing rats with the AChRfrom the electric organ of the electric ray Torpedo californica (19,20). A small proportion of Abs against the Torpedo AChR cross-reacts with the AChR of the muscle (21). As in human MG withanti-AChR autoantibodies, the disease symptoms in EAMG arecaused by Ab-mediated destruction of the neuromuscular junction(22, 23).In this study, we examined the effect of bortezomib in EAMG

rats using two different treatment regimes. The first treatmentconsisted of bortezomib injections for 8 wk, starting directly afterimmunization (herein referred to as 8w-Bz). For the secondtreatment regimen, rats were injected with bortezomib startingonly 4 wk after immunization, when autoantibody titers were al-ready detectable, until 8 wk after immunization (4w-Bz). Using thissetup we investigated the potential therapeutic effect of bortezomibafter onset of the disease. The results show that both treatment re-gimes reduce autoantibody levels by depleting bone marrow plasmacells, but only the 8 wk bortezomib-treatment led to a significantimprovement of the clinical condition of the EAMG rats and to areduction of postsynaptic damage.

Materials and MethodsAnimals

Inbred female Lewis rats (n = 90) were obtained from the Department ofExperimental Animal Services, Maastricht University (The Netherlands).Permission to perform this experiment was granted by the Committee onAnimal Welfare, according to Dutch governmental rules. At 6 wk of age,animals were weighed and divided into six experimental groups (Table I)with an equal average weight.

Induction of EAMG

EAMG was induced in 7-wk-old rats (n = 54) by active immunization withAChR purified from the electric organ of T. californica (tAChR) in CFA. Inbrief, 20 mg tAChR (20) was dissolved in 100 ml PBS and emulsified withan equal volume of CFA with 0.1% of Mycobacterium tuberculosis H37(Difco Laboratories, Detroit, MI). Animals were initially anesthetized ina cylindrical tube through which 5% isoflurane in air was supplied. Sub-sequently, 3% isoflurane was supplied by a cap over the head and 200 mlCFA/tAChR emulsion was injected s.c. at the base of the tail at threedifferent spots, as described by Lennon and colleagues (24). Control ani-mals (n = 36) were injected with an equal volume of emulsified PBS andCFA. Rats were sacrificed 8 wk after immunization or earlier, if they hadreached the humane endpoints as described below. They were sacrificed byinhalation of CO2 in air and subsequent cervical dislocation.

Experimental design and administration of drugs

Bortezomib was purchased as a lyophilized powder (Velcade; Janssen-Cilag, Beerse, Belgium) and dissolved in sterile saline solution at a finalconcentration of 0.1 mg/ml. Two weekly doses of 0.2 mg/kg bortezomibsolution were administered s.c., which is considered the highest dose to usewithout having increased mortality rates or severe side effects in rats (10,25, 26). Control and EAMG groups were subdivided into three treatmentregimes each (Table I). “Saline” groups received two weekly s.c. injectionsof 0.9% NaCl solution (2 ml/kg) at the neck for 8 wk. The 4w-Bz groupsreceived two weekly saline injections for the first 4 wk after immunization(by which time autoantibody levels were highly elevated in the plasma)and subsequently two weekly injections of bortezomib for another 4 wk.The 8w-Bz groups received two weekly injections of bortezomib for 8 wk,starting directly after immunization.

For practical reasons the experiment was conducted three times in sets of30 animals each, including an equal number of all the aforementioned groups.The animals’ tissues were analyzed using electron microscopy (EM), elec-tromyography (EMG), immunofluorescence (IF), and FACS, and the numberof animals used for each method is indicated in Table I.

Weight and clinical scoring

The weights of animals were measured on a weekly basis as a generalindicator of health and for dose calculations. The severity of clinical signs ofEAMGwas scored weekly bymeasuring muscular weakness by two blindedinvestigators (A.M.G. and M.P.). The animals’ muscle strength and fati-gability was assessed by their ability to grasp and lift repeatedly a 300-gmetal rack from the table, while suspended manually by the base of the tailfor 30 s (27–29). Clinical scoring was based on the presence of tremor,hunched posture, muscle strength, and fatigability. Disease severity wasexpressed as follows: 0, no obvious abnormalities; 1, no abnormalitiesbefore testing, but reduced strength at the end; 2, clinical signs presentbefore testing, that is, tremor, head down, hunched posture, weak grip,difficulty in breathing; 3, severe clinical signs present before testing, nogrip, moribund (24).

Animals that reached a clinical score of level 3, or lost more than 20% oftheir weight in the course of 1 wk, were sacrificed within 24 h.

Tissue preparation

For EM analysis, bone marrow was extracted from the femurs by cutting offtheir upper and lower endings and flushing the shaft with 10 ml sterile salinesolution through the bone marrow cavity using a syringe with a 25-gaugeneedle. For FACS analysis of bone marrow, PBS containing 2% FCS and0.1% NaN3 (FACS buffer) was used instead. Thymus and spleen tissuewere entirely removed from the animals by dissection and the organ weightwas measured. Single-cell suspensions were prepared using a gentleMACStissue dissociator (Miltenyi Biotec, Bergisch Gladbach, Germany) ac-cording to the manufacturer’s instructions. Cells of different tissues wereresuspended in FACS buffer and run through a 70-mm nylon cell strainer to

Table I. Treatment groups

Saline Groups(No Bortezomib)

4w-Bz Groups (BortezomibStarted 4 wk afterImmunization)

8w-Bz Groups (BortezomibStarted Directly after

Immunization)

Control (immunizedwith CFA)

n = 12 (IF and FACS, n = 5;EM and EMG, n = 7)

n = 12 (IF and FACS, n = 5;EM and EMG, n = 7)

n = 12 (IF and FACS, n = 5;EM and EMG, n = 7)

EAMG (immunizedwith AChR in CFA)

n = 18 (IF and FACS, n = 8;EM and EMG, n = 10)

n = 18 (IF and FACS, n = 8;EM and EMG, n = 10)

n = 18 (IF and FACS, n = 8;EM and EMG, n = 10)

2504 PLASMA CELL DEPLETION WITH BORTEZOMIB IN EAMG

on October 4, 2011

ww

w.jim

munol.org

Dow

nloaded from

remove large cell clumps. Numbers of viable cells were determined bycounting trypan blue negative cells on hemocytometers. Cells were dividedinto microcentrifuge tubes (105 cells/100 ml/tube) and then stained fordead cells, T cells, and B cells.

Heparinized blood samples were taken from the vena saphena weekly,starting on the week before immunization (week 0) until the last week ofexperiment (week 8). PBMCs were isolated by lysing RBCs using FACSlysing buffer (BD Biosciences). PBMCs from 80 ml blood were divided inmicrocentrifuge tubes in a ratio of 20:20:40 ml to stain for dead cells,T cells, and B cells, respectively.

Proteasome activity assay

The proteolytic activity of the proteasome was evaluated in whole blood(30) by means of a 20S proteasome activity kit (APT 280; Millipore), asdescribed by the manufacturer. Taking into account the pharmacokineticand pharmacodynamic profiles of s.c. bortezomib administration (31),blood samples were obtained between 3 and 6 h after administration ofbortezomib or saline. In brief, 80 mg whole blood protein extract wasincubated in the provided buffer with 3.8 mg fluorophore-linked peptidesubstrate (LLVY-7-amino-4-methylcoumarin [AMC]) for 120 min at 37˚C.Proteasome activity was measured by quantification of relative fluorescentunits from the release of the fluorescent-cleaved product AMC usinga 380/460 nm filter set in a fluorometer (Victor X3 multilabel reader;PerkinElmer). A solution of the 20S proteasome subunit and the protea-some inhibitor lactacystin were used as controls for the assay.

Autoantibody titers

Ab titers against rat AChR were measured in plasma samples with animmunoprecipitation RIA. In brief, 2.5 ml plasma was incubated at 4˚Covernight with 100 ml rat muscle cell-membrane extract (containing ∼5nmol/l AChR; isolated from denervated rat muscles). The AChR was la-beled with an excess of [125I]-a-bungarotoxin (NEX126, 5 TBq/mmol;PerkinElmer). The immune complexes were precipitated by addition of150 ml goat anti-rat IgG serum and incubation for 4 h at 4˚C. Pellets werewashed three times in PBS with 0.5% Triton X-100 and centrifuged at25,0003 g for 5 min. Radioactivity was measured on an automatic gammacounter (2470 Wizard2; PerkinElmer). Autoantibody titers were expressedas nanomoles of a-bungarotoxin binding sites per liter.

Total IgG ELISA

A sandwich ELISA was used to measure total IgG content in plasmasamples. Briefly, ELISA plates (Microlon 655092; Greiner Bio-One,Frickenhausen, Germany) were coated with 50 ml catching Ab (goat Iganti-rat IgG, ab6252; Abcam, Cambridge, U.K.), diluted 1:200 in coatingbuffer (50 mM sodium carbonate [pH 9.6]), for 1 h at 37˚C. Plates werewashed with PBS containing 0.05% Tween 20 and blocked for 30 min with100 ml 4% nonfat dry milk in PBS (blotting grade blocker, catalogue no.1706404; Bio-Rad). Afterwards, 50 ml plasma samples, diluted 1:20,000 inincubation buffer (PBS plus 1% BSA plus 0.02% Tween 20), were in-cubated for 1 h at 37˚C. A standard curve was made using serially dilutedsamples of purified rat IgG (catalogue no. I8015; Sigma-Aldrich). Afterwashing, 50 ml HRP-conjugated Ab (ab6257; Abcam), diluted 1:5000 inincubation buffer, was added and plates were incubated for another hour at37˚C. Following a washing step, 100 ml tetramethylbenzidine substratesolution (s(HS)TMB; SDT Reagents, Baesweiler, Germany) was used todevelop HRP-labeled Abs bound to the plates. The color reaction wasallowed to develop for 10 min and stopped with 50 ml 2 M sulfuric acid.The OD was measured at 450 nm filter using a microplate reader (VictorX3 multilabel reader). Results were expressed as milligrams of total IgGper milliliter of plasma.

Apoptosis assays

Early apoptotic and dead cells were identified by flow cytometric analysisaccording to surface binding of FITC-labeled Annexin V to exposedmembrane phosphatidylserine and propidium iodide (PI) staining (annexinV-FITC apoptosis detection kit; BD Biosciences, Breda, The Netherlands).The cells (105/100 ml) were washed with annexin V-binding buffer andincubated with 5 ml annexin Vand 5 ml PI for 15 min at room temperature.Without washing, cells were immediately measured. Annexin V+/PI2 cellswere regarded as early apoptotic, while annexin V+/PI+ cells were con-sidered dead cells.

Extracellular staining for T cell markers

The cells were washed once with FACS buffer by centrifugation at 2503 gat 4˚C and incubated for 30 min at 4˚C with Abs directed against CD3(FITC-labeled), CD4 (PE-labeled), and CD8 (PerCP-labeled) (all from BD

Biosciences), diluted 1:50 in FACS buffer. The samples were washed twiceand kept at 4˚C in the dark until measurement within 2 h.

Intracellular staining for B cell markers

Cells were washed once with FACS buffer by centrifugation at 250 3 g at4˚C and incubated with PE-conjugated Ab against CD45RA (BD Bio-sciences), diluted 1:20 in FACS buffer. After one washing step, cells werefixed with 2% paraformaldehyde in FACS buffer for 10 min at 37˚C. Aftertwo more washing steps, cells were permeabilized with cold 90% methanolfor 30 min on ice. Cells were washed twice and incubated with FITC-conjugated Ab directed against intracellular Igk (BD Biosciences), diluted1:20 in FACS buffer. The samples were washed twice and kept at 4˚C inthe dark until measurement within 2 h. Because no appropriate CDmarkers were available to specifically detect rat plasma cells, we measuredrat plasma cells by using high levels of intracellular Igk expression.

FIGURE 1. Proteasome activity in blood lysates. Samples were in-

cubated with a labeled peptide substrate (LLVY-AMC) and the amount of

cleaved fluorophore AMC was measured in a fluorometer. A, Proteasome

activities at 4 wk after immunization were normalized using the average

relative fluorescence units (RFU) value of the control saline group at week

4. B, Proteasome activities at 8 wk after immunization were normalized

using the average RFU value of the control saline group at week 8. One-

way ANOVA and Bonferoni post hoc testing were used for statistical

analyses.

The Journal of Immunology 2505

on October 4, 2011

ww

w.jim

munol.org

Dow

nloaded from

FACS analysis

All cytofluorometric analyses were performed on a FACSCalibur (BDBiosciences) and analyzed using the CellQuest software (BD Biosciences).Forward and sideward light angle scatters were collected. Using these plots,samples were gated to exclude cell debris and cellular aggregates for furtheranalysis. For each marker, the percentage of positive cells stained abovebackground was measured for all gated cells. The cutoff was defined usingunstained cells processed alongside the experimental samples.

Plasma cell quantification by EM

Bone marrow cell suspensions were fixed by mixing with an equal volumeof 5% glutaraldehyde in PBS and then centrifuged at 8003 g. Pellets wereresuspended in PBS and embedded in 10% gelatin, then centrifuged againat 1000 3 g to form a compact pellet. Cell pellets were postfixed with 1%osmium tetroxide in 0.1 M phosphate buffer (pH 7.4) dehydrated througha graded ethanol series and embedded in Epon 812 (Electron MicroscopySciences, Fort Washington, PA). Ultrathin sections from whole pelletswere contrasted with uranyl acetate and lead citrate and viewed witha Philips CM 100 electron microscope. Plasma cell recognition was basedon morphological features that are particular to this cell type, such as thetypical “cartwheel” chromatin configuration in the nucleus and the pres-ence of extensive rough endoplasmic reticulum in the cytoplasm, in-dicative of an intense protein production. Quantification was performed atsix different portions of the sample to have a systematic representation ofthe cell gradient in the sample that resulted from the centrifugation.Results are expressed as percentage of plasma cells; a total of 800 bonemarrow cells were counted per sample.

Electromyography

Animals were initially anesthetized as described above and subsequentlyintubated in the trachea. Anesthesia was maintained with 3% isoflurane inair. Compoundmuscle action potential (CMAP) decrement was measured inthe tibialis anterior muscle using the EMG system Viking IV (NicoletBiomedicals, Madison, WI) at the end of the experimental period. Forstimulation, two small monopolar needle electrodes were used. The cathodewas inserted near the peroneal nerve at the level of the knee and the anode

more proximal and lateral at a distance of 3–4 mm. For recording, a thirdmonopolar needle electrode was inserted s.c. over the tibialis anteriormuscle. The reference electrode was inserted s.c. near the ankle. A ringelectrode around the tail served as ground electrode. To detect a decrementalresponse of the CMAP, series of eight supramaximal stimuli were given at 3Hz with 0.2 ms duration. The test was considered positive for decrementwhen both the amplitude and the area of the CMAP-negative peak showeda decrease of at least 10% (32). To demonstrate reproducibility, at least threeconsecutive decrement recordings were made of all investigated muscles.During the measurements, skin temperature was kept between 35˚C and37˚C by means of an infrared heating lamp. If initially no decrement waspresent in the tibialis anterior muscle, neuromuscular transmission waschallenged by a continuous i.v. infusion of curare [(+)-tubocurarine, T2379;Sigma-Aldrich]. A solution of 20 mg/ml curare was injected into the venasaphena using a Terfusion syringe pump (model STC-521; Terumo, Tokyo,

FIGURE 2. Electron micrographs of bone marrow plasma cells. After 4

wk of bortezomib treatment, the rough endoplasmic reticulum cisternae

have a vesicular appearance; after 8 wk of treatment, pronounced dilatation

of the rough endoplasmic reticulum is visible. Cells were stained with

osmium tetroxide and contrasted with uranyl acetate and lead citrate. Scale

bars, 1 mm.

FIGURE 3. Analysis of plasma cells in the bone marrow 8 wk after

immunization. A, Electron microscopical analysis. Bortezomib treatment

decreases the number of plasma cells in the bone marrow. B, Flow cyto-

metric analyses of Igkhigh cells. Data are shown as percentages of cells

with respect to gated living bone marrow cells. The proportion of Igkhigh

cells was significantly lower in 4w-Bz and 8w-Bz groups compared with

the corresponding saline-treated groups. Horizontal bars represent the

mean percentage of each group. Two-way ANOVA and Bonferoni post hoc

testing were used for statistical analyses.

2506 PLASMA CELL DEPLETION WITH BORTEZOMIB IN EAMG

on October 4, 2011

ww

w.jim

munol.org

Dow

nloaded from

Japan) at a rate of 1 ml/h (0.33 mg curare/min). During curare infusion,CMAPmeasurements were repeated with intervals of 1 min until a repeateddecrement was observed. The resistance against curare was used as an in-direct, albeit nonlinear, measure for the safety factor of neuromusculartransmission and thus for the performance of neuromuscular transmission(33, 34).

EM of muscle tissue

Electron micrographs were taken from endplates of the tibialis anteriormuscles. Anesthetized rats were transcardially perfused as previously de-scribed (21, 32). Ultrathin sections were viewed with a Philips CM 100electron microscope. At least five endplate regions were photographedfrom each muscle. Quantitative morphometry of the folding index (lengthof postsynaptic membrane/length of presynaptic membrane) was per-formed as previously described (34, 35). For the analysis, the followingnumber of animals was used per group: control saline (n = 3); EAMGsaline (n = 4); EAMG 4w-Bz (n = 4); EAMG 8w-Bz (n = 4). Between 5and 25 endplate regions were analyzed per animal.

Statistics

GraphPad Prism 4 was used to perform statistical analyses. Comparisonbetween normally distributed values was performed using one- or two-wayANOVA, wherever appropriate. Bonferroni post hoc tests were used tocompare groups to each other. A two-sided probability value of 0.05 orlower was considered significant. Values are expressed as means 6 SEMunless stated otherwise. Clinical scores were analyzed by the x2 test fortrend, and survival was analyzed using the log-rank test.

ResultsBortezomib reduces plasma cells in bone marrow

To investigate whether proteasome inhibition affects plasma cellsin vivo, rats were injected with bortezomib or saline. Subcutaneousinjections of bortezomib significantly reduced the proteasomeactivity in rat whole blood (Fig. 1). The effect of bortezomib onplasma cells from the bone marrow was analyzed by EM and byFACS. In the bone marrow of bortezomib-treated rats, plasmacells with altered morphology were frequently observed (Fig. 2),

which was characterized by a vesicular appearance of the roughendoplasmic reticulum cisternae or pronounced dilatation of therough endoplasmic reticulum. The percentage of plasma cells inbone marrow was markedly reduced in bortezomib-treated groups(Fig. 3A). Animals that received bortezomib only between 4 and8 wk after immunization showed a significant decrease in theirpercentage of bone marrow plasma cells (57% reduction in the4w-Bz control group [p , 0.05] and 82% reduction in the 4w-BzEAMG group [p , 0.01] compared with the corresponding saline-treated groups). Rats in the 8w-Bz EAMG group also showed astrong and significant depletion of bone marrow plasma cells (p ,0.05; 70% reduction compared with the saline-treated EAMGgroup). Very similar results were obtained by FACS analysis us-ing intracellular staining of the Igk L chain in bone marrow cells(Fig. 3B).

Bortezomib affects the lymphoid organs

We investigated the overall effect of bortezomib treatment on theimmune system by measuring the weight of thymus and spleentissue and analyzing the proportions of B cells and T cell sub-sets in the thymus, spleen, blood, and bone marrow by FACS(Table II).Bortezomib treatment significantly reduced the mean thymus

weight, both in the 4w-Bz (46% reduction; p , 0.01) and 8w-Bzgroups (50% reduction; p, 0.01), compared with the saline group(data not shown). In contrast, the mean spleen weight significantlyincreased by bortezomib treatment in both 4w-Bz (36% increase;p , 0.05) and 8w-Bz groups (32% increase; p , 0.05) comparedwith the corresponding saline-treated groups (data not shown). Nosignificant difference was observed between control and EAMGanimals.To assess the effects of bortezomib treatment on the leukocyte

viability, we measured the proportion of early apoptotic (annex-inV+/PI2) and dead cells (annexinV+/PI+).

Table II. FACS analysis of lymphoid organs

Tissue Saline 4w-Bz 8w-Bz

Dead cells, Ann+/PI+ Thymus 11.25 6 1.43 13.63 6 1.17 14.19 6 1.94Spleen 25.38 6 2.34 28.00 6 3.28 31.26 6 4.25

Bone marrow 12.93 6 0.86 14.57 6 1.20 15.82 6 1.16PBMC 5.72 6 0.66 2.84 6 0.17*** 2.92 6 0.18***

Early apoptotic cells, Ann+/PI2 Thymus 8.23 6 0.96 12.11 6 0.63 10.22 6 1.60Spleen 17.02 6 1.27 16.36 6 1.62 15.80 6 1.63

Bone marrow 12.40 6 1.76 10.40 6 1.19 13.08 6 1.56PBMC 2.25 6 0.18 1.23 6 0.12*** 1.91 6 0.22

CD45RA+/Igk+ Thymus 0.48 6 0.05 0.89 6 0.12 0.63 6 0.10Spleen 19.85 6 0.36 15.85 6 1.05* 16.13 6 1.24*

Bone marrow 4.16 6 0.16 2.56 6 0.30** 2.44 6 0.33***PBMC 5.64 6 0.43 3.16 6 0.26*** 2.57 6 0.18***

CD3+/CD4+ CD82 Thymus 7.72 6 0.39 10.53 6 0.65** 9.62 6 0.61Spleen 39.44 6 0.25 43.92 6 1.53* 44.72 6 0.71*

Bone marrow 1.27 6 0.22 1.27 6 0.31 1.49 6 0.29PBMC 56.52 6 0.68 63.21 6 0.63*** 62.18 6 0.79***

CD3+/CD8+ CD42 Thymus 1.78 6 0.11 3.49 6 0.30*** 2.80 6 0.31*Spleen 12.89 6 0.26 9.82 6 0.39*** 9.13 6 0.63***

Bone marrow 1.67 6 0.30 0.82 6 0.08* 1.29 6 0.20PBMC 19.48 6 0.40 20.91 6 0.47 20.85 6 0.43

CD4+/CD8+ Thymus 83.57 6 1.24 77.41 6 1.87* 78.99 6 1.38Spleen 1.33 6 0.06 1.27 6 0.07 1.46 6 0.07

Bone marrow 1.60 6 0.39 1.03 6 0.11 0.99 6 0.12PBMC 1.39 6 0.12 1.62 6 0.13 2.16 6 0.19**

Using flow cytometry, the effects of bortezomib treatment on apoptosis and on lymphocytes were measured in the thymus, spleen, bone marrow, andperipheral blood. Results are shown as average percentages 6 SEM. One-way ANOVA analysis and Bonferroni post hoc testing. For analyzing the effectof bortezomib treatment, the data of control and EAMG groups were combined since no significant differences were observed between them in any of theparameters studied.

*p , 0.05; **p , 0.01; ***p , 0.001.Ann, annexin V.

The Journal of Immunology 2507

on October 4, 2011

ww

w.jim

munol.org

Dow

nloaded from

In the spleen, thymus, and bone marrow, a trend toward a higherproportion of early apoptotic and dead cells after bortezomib ad-ministration could be observed, but the differences did not reachstatistical significance. However, the lymphocyte subpopulation inthe bone marrow contained a significantly increased number ofapoptotic or dead cells after bortezomib-treatment (data not shown).In the peripheral blood, unexpectedly, bortezomib treatment

significantly decreased the proportion of dead cells in PBMCs inthe 4w-Bz group and 8w-Bz groups compared with the salinegroups. The proportion of early apoptotic cells in PBMCs wassignificantly lower in the 4w-Bz groups (p , 0.001) but not in the8w-Bz groups.In the spleen, the bone marrow, and the blood, the proportion

of CD45RA+/Igk+ B cells was significantly decreased after borte-zomib treatment (Table II), both in the 4w-Bz and the 8w-Bzgroups.After treatment with bortezomib, the proportion of immature

CD4+/CD8+ cells in the thymus was decreased whereas the pro-portions of CD3+/CD4+/CD82 Th cells and cytotoxic CD3+/CD8+/CD42 T cells were increased.In the blood and the spleen, CD3+/CD4+/CD82 cells were

significantly increased; the proportion of CD3+/CD8+/CD42 cellswas significantly decreased in the spleen and bone marrow ofbortezomib-treated rats.In general, the two bortezomib treatment regimes led to similar

changes of the rat immune system, with the exception of the pro-portion of apoptotic cells, as aforementioned.

Total IgG concentration is decreased by bortezomib

The effect of bortezomib on total IgG content in plasma sampleswas measured by ELISA (Fig. 4A). In comparison with the saline-treated group, total IgG at week 8 was significantly reduced inboth the 4w-Bz and the 8w-Bz group (p , 0.001). Importantly,this immunosuppressive effect of bortezomib was observed inboth control and EAMG animals, although IgG reduction wasmore pronounced in 8w-Bz EAMG animals than in the 4w-BzEAMG rats (p , 0.05). A slightly but significantly higher con-centration of IgG was observed in all EAMG groups in compar-ison with the corresponding control groups. Compared to thesaline-treated groups, a highly significant reduction in IgG con-centrations (p , 0.001) was already achieved after 4 wk of bor-tezomib treatment in the 8w-Bz group (data not shown).

Bortezomib reduces autoantibody titers in EAMG

The plasma concentration of autoantibodies to the rat AChR wasmeasured by RIA. Autoantibodies were detectable 4 wk afterimmunization in all EAMG animals and reached very high levelsafter 8 wk (Fig. 4B). The variability of autoantibody levels be-tween animals is typical for the EAMG model, but it should beborne in mind that already a titer of 1 nM autoantibodies is suf-ficient to cause substantial damage to the NMJ (33). As expected,no anti-AChR Ab titers could be detected in sham/CFA-immunizedanimals (control group; data not shown). In contrast, animals thatreceived bortezomib injections from the moment of immunizationshowed a significantly lower production of autoantibodies (p ,0.01; corresponding to a 72% reduction of average autoantibodytiter) compared with the saline-treated EAMG group 8 wk afterimmunization (Fig. 4C). Interestingly, rats that received bortezo-mib 4 wk after immunization had an autoantibody productionprofile similar to that observed in the 8w-Bz group. After injectionof bortezomib the production of anti-AChR Abs was reducedsignificantly compared with saline-treated animals (correspondingto a 60% reduction; p , 0.01). This demonstrated that bortezomibeffectively diminished Ab production not only when administrated

FIGURE 4. Total IgG and autoantibody titers in plasma. A, Plasma IgG

titers 8 wk after immunization. Bortezomib significantly reduced plasma

IgG levels. Two-way ANOVA and Bonferoni post hoc testing were used

for statistical analyses. B, Average anti-rat AChR-titer; error bars corre-

spond to the SEM. **p , 0.01; ***p ,0.001 compared with the saline-

treated groups. Autoantibody titers of the 4w-Bz and the 8w-Bz groups

were not significantly different from each other at any time point. C, Anti-

rat AChR titers 8 wk after immunization with tAChR. One-way ANOVA

and Bonferoni post hoc testing were used for statistical analyses.

2508 PLASMA CELL DEPLETION WITH BORTEZOMIB IN EAMG

on October 4, 2011

ww

w.jim

munol.org

Dow

nloaded from

at the moment of immunization but also once the immune re-sponse was already ongoing.

Bortezomib ameliorates clinical conditions in EAMG

To assess the effect of bortezomib on the overall condition ofexperimental animals, we weighed them and scored their clinicalstatus on a weekly basis. During the first 5 wk of treatment, controlanimals that had received bortezomib from the moment of im-munization (8w-Bz group) showed a slower increase in their total

body weight compared with saline-treated control animals (Fig. 5A,5B; p , 0.001). After 5 wk, the growth of these animals nor-malized again. A similar reduction of growth was observed 4 wk

FIGURE 5. Average rat weights. A, Body weights of bortezomib-treated

animals increased slower compared with saline-treated animals. Weight

loss occurred frequently in EAMG animals. B and C, Normalized weights

were calculated using the average weight of the saline-treated control

group. Error bars correspond to the SEM.

FIGURE 6. MG symptoms and muscle function. A, Survival. B, Clinical

scores of muscle weakness. Each group contained 18 animals (indicated

with 100%). C, EMG after curare infusion. The curare dose that induces at

least 10% decrement of the CMAP was used as a measure of the neuro-

muscular safety factor.

The Journal of Immunology 2509

on October 4, 2011

ww

w.jim

munol.org

Dow

nloaded from

later in the 4w-Bz control group. At the end of the experimentthere were no significant differences between the average weightsof the 4w-Bz and the 8w-Bz control groups. As expected, controlanimals did not present muscle weakness or any other clinical signof EAMG.Animals immunized with Torpedo AChR developed clinical

symptoms of EAMG starting 5 wk after immunization, when Abtiters reached considerable levels. Frequently, weight loss pre-ceded the observation of other myasthenic symptoms in EAMGanimals, indicating weakness of bulbar muscles and difficulties inchewing and swallowing. The disease in some of these animalsprogressed rapidly to score 3 within 2 d and they had to be sac-rificed (Fig. 6A). By the end of the experiment 50% of animals (9of 18) treated with saline solution reached a clinical score of 3 orlost .20% body weight and had to be sacrificed for ethical rea-sons. This percentage was reduced to 33% (6 of 18) in the 4w-Bzgroup and to 11% (2 of 18) in the 8w-Bz group (Fig. 6B). Thesurvival rate of the 8w-Bz EAMG group was significantly highercompared with the saline-treated EAMG group (p , 0.01; Fig.6A).

From the AChR-immunized animals, 78% developed muscleweakness in the saline-treated EAMG group, 72% in the 4w-BzEAMG group, and 39% in the 8w-Bz EAMG group. The clini-cal score of the 8w-Bz EAMG group was significantly lowercompared with the 4w-Bz group (p , 0.05) and the saline-treatedEAMG group (p , 0.01; Fig. 6B). Despite the reduced amount ofautoantibodies, the muscle weakness and the survival rate of the4w-Bz EAMG group were not significantly different from thesaline-treated EAMG group. The onset of weight loss in the 4w-Bz EAMG group even occurred somewhat earlier compared withthe saline-treated EAMG group (Fig. 5C), indicating that weightloss was partly caused by the (side) effects of bortezomib.We evaluated the safety factor of neuromuscular transmission,

which is a function of the postsynaptic density of AChRs, byperforming EMG studies in the presence of the AChR-blockingagent (+)-tubocurarine (curare). The amount of curare needed toinduce a decrement in CMAP is related to the safety factor ofneuromuscular transmission. Bortezomib had slight effects on thecurare sensitivity of the NMJ in control animals in the 4w-Bz group(Fig. 6C). The neuromuscular transmission was significantly

FIGURE 7. Electron micrographs of

synaptic boutons of the NMJ. Nerve termi-

nals are indicated by asterisks. In control

animals (A–C), the postsynaptic membrane

contains secondary clefts (postsynaptic folds),

which are indicated by arrows. In EAMG

animals (D–G) pathologic changes of the

postsynaptic membrane are indicated: degen-

erating folds (arrowhead), simplified and

without folds (dagger), and widening of the

primary and secondary synaptic clefts (double

dagger). The postsynaptic damage of an ani-

mal with an anti-AChR titer of 3 nM (F) was

mild compared with an animal with a titer of

20 nM (G). The muscle tissue was stained

with osmium tetroxide and contrasted with

uranyl acetate and lead citrate.

2510 PLASMA CELL DEPLETION WITH BORTEZOMIB IN EAMG

on October 4, 2011

ww

w.jim

munol.org

Dow

nloaded from

impaired in all EAMG groups compared with the correspondingcontrol groups. In the 8w-Bz EAMG group, neuromuscular trans-mission was significantly improved compared with the saline-treated and the 4w-Bz EAMG groups (p , 0.05).Ultrastructural analysis of the NMJ revealed damage of the

postsynaptic membrane morphology, with degenerating or absentsecondary clefts in EAMG animals (Fig. 7). The damage in end-plates of animals with low titers in the 8w-Bz group (Fig. 7F) wasless severe compared with animals with higher titers (Fig. 7D, 7E,7G). Quantitative morphometric analysis of the synapse ultra-structure was performed to measure the loss of postsynaptic folds(Fig. 8). The folding index was significantly reduced in the saline-treated EAMG animals compared with the saline-treated controlanimals (a reduction of 55%, p , 0.001). In the 8w-Bz EAMGanimals only a 20% reduction of the folding index compared withsaline-treated control animals was observed, and a considerableproportion of the endplates had a relatively high folding index.Compared to the saline-treated EAMG group, the folding index inthe 8w-Bz group was significantly higher (p , 0.001). However,this treatment effect was not observed in the 4w-Bz group. Inconclusion, bortezomib could not improve synaptic ultrastructureif treatment was started 4 wk after immunization, but it efficientlyprevented damage of the postsynaptic membrane when adminis-trated for 8 wk starting directly after immunization.

DiscussionIn this study we analyzed the effect of proteasome inhibition on theautoimmune response in the EAMG model for MG. Bortezomibreduced the amount of plasma cells, leading to a significant re-duction of total serum IgG and autoantibody levels and caused anamelioration of myasthenic symptoms compared with a saline-treated EAMG group. Both the 4- and 8-wk bortezomib treat-ment regimes (4w-Bz and 8w-Bz) were generally well tolerated,but some side effects were noticed, which are discussed below.There were no significant differences in autoantibody production

between the 4w-Bz and the 8w-Bz group. Because bortezomib inthe 4w-Bz group was only administered starting 4 wk after im-munization with autoantigen, this suggests that mainly the late(effector) phase of the autoimmune response is affected by pro-teasome inhibition.In the EAMG model, the production of Abs against the muscle

AChR is induced by immunization with tAChR. EAMG in Lewisrats is characterized by chronic muscle weakness starting 5 wkafter immunization. A transient acute phase of muscle weaknessstarting 1 wk after immunization is seen in EAMG models usingCFA with Bordetella pertussis and M. tuberculosis (20). In theimmunization protocol used in this study with CFA containingonly M. tuberculosis, the acute phase of muscle weakness doesnot occur (20). Therefore, we could investigate both a therapeuticas well as a preventive treatment regimen. Bortezomib efficientlyreduced the production of autoantibodies and also ameliorated MGsymptoms in the 8w-Bz group. The average levels of anti-AChRautoantibodies in this study were high, comparable to a previousstudy using the same protocol with 20 mg tAChR (36, 37) andmuch higher than another study using 10 mg tAChR (33), rangingbetween 3 and 80 nM even in the 8w-Bz groups. Because 1 nMserum anti-muscle AChR antibodies can already reduce the amountof total muscle AChR by 50% in rats (33), the limited improvementof muscle strength in this study is understandable. It is thereforeconceivable that if we could have used EAMG animals witha lower titer, the effects of bortezomib would have been morepronounced. Nevertheless, the resistance against curare, and thusthe amount of functional AChR at the NMJ and the folding indexof the postsynaptic membrane, was significantly increased bybortezomib in the 8w-Bz treatment group, resulting in highersurvival rates. In human MG patients, an autoantibody titer re-duction of 50% after plasma exchange is generally sufficient toachieve clinical remission (38, 39), and therefore in this respectthe bortezomib-induced reduction of autoantibody production by.65% within 4 wk is therapeutically promising.However, despite the reduced autoantibody levels, the 4w-Bz

treatment did not result in a significant improvement ofhealth in EAMG animals, in contrast to the 8w-BZ treatment,where bortezomib caused amelioration in the condition of theanimals. Two side effects of bortezomib might have influenced thisresult. First, bortezomib negatively affected body weight during thefirst 4 wk of administration in control animals and presumably alsoin EAMG animals. In the 4w-Bz EAMG group, this coincided withthe weight loss as a result of muscle weakness and the resultingproblems with eating and drinking. Because a 20% weight loss waschosen as a criterion to sacrifice animals for ethical reasons, thebortezomib treatment could have reduced survival time to someextent. Second, we observed a mild impairment of neuromusculartransmission in the 4w-Bz control group (but not in the 8w-BzControl group) in comparison with the saline-treated controlgroup. This effect possibly indicates transient nerve damage similarto the bortezomib-induced polyneuropathy (26, 40). Arguably, theweight loss in control animals was caused by an effect of pro-teasome inhibition on the gastrointestinal tract, which has beenseen in patients (41). However, our study was not designed forinvestigating the side effects of bortezomib, and therefore wecannot conclusively attribute the effect on weight to any particularside effect. In the 8w-Bz control group, weight gain, neuromus-cular transmission, and apoptosis were normalized 8 wk afterimmunization, suggesting that by that time compensatory mech-anisms limited these adverse effects of bortezomib.Despite comparable autoantibody titers, the ultrastructural post-

synaptic damage of the 4w-Bz EAMG group was significantlyhigher compared with the 8w-Bz EAMG group. Because the repair

FIGURE 8. Analysis of folding index (length of postsynaptic mem-

brane/length of presynaptic membrane) by quantitative morphometry of

electron micrographs. Each point represents one endplate region with one

or two synaptic boutons as shown in Fig. 7. Due to loss of postsynaptic

folds, the folding index is significantly reduced in EAMG saline animals

compared with control saline animals. In the EAMG 8w-Bz, the reduction

of postsynaptic folding was prevented by bortezomib. One-way ANOVA

and Bonferoni post hoc testing were used for statistical analyses.

The Journal of Immunology 2511

on October 4, 2011

ww

w.jim

munol.org

Dow

nloaded from

of the postsynaptic membrane takes at least 10 d (42), it seemspossible that the observed loss of postsynaptic folding in the 4w-Bzgroup is the result of earlier damage, in particular in the periodbetween 4 and 7 wk after immunization, when titers were highercompared with the 8w-Bz EAMG group.Apart from the intended killing of plasma cells, bortezomib af-

fected the immune system in a more general fashion. In particular,the thymus was affected by bortezomib, an effect that might not beharmful, since thymectomy is a frequently used treatment in MG,albeit with unproven efficacy so far. In this respect it is relevant thatin human thymocyte cultures from thymectomized MG patients weobserved that bortezomib induced cell death and reduction of au-toantibody production (A. Gomez, K. Vrolix, and M. Losen, un-published observations). Similar to a previous study in mice (43)we found in our rat model that bortezomib mainly affects immaturethymocytes. In the spleen, bone marrow, and the blood of rats, theproportion of CD45RA+/Igk+ B cells was significantly decreased,whereas the proportion of CD3+ T cells of total lymphocyteswas increased. There was a trend for increased proportion ofdead and apoptotic cells in the thymus, bone marrow, and spleenof bortezomib-treated animals. In contrast, bortezomib induced asignificant reduction of apoptotic cells in the blood. Because thePBMCs only represent a minor proportion of leukocytes, thiseffect could be attributed to migration of cells into other lymphoidorgans, for example, the spleen, which was significantly enlarged inbortezomib-treated rats.In the past the EAMG model has been instrumental for testing

the efficacy of new therapies that are now used for the treatment ofMG (reviewed in Ref. 44), for example, cyclophosphamide (45),pixantrone (46), linomide (47), azathioprine, and hydrocortisone(48). Also, mycophenolate mofetil efficiently reduces autoanti-bodies in a rat model of EAMG (33). This study now indicates thatbortezomib might be useful to complement these drugs for thetreatment of MG since it can additionally target plasma cells thatproduce autoantibodies. Because of the observed plasma cell re-duction in rats it is reasonable to presume that a course of bor-tezomib treatment might well eliminate short- and long-livedplasma cells also in humans and therefore induce a long-lastingtreatment response in Ab-mediated autoimmune diseases.

AcknowledgmentsWe thank Joost Van den Broeck, Caroline Hammels, Jo Stevens, and Jonas

Hummel for valuable help and Pauline Wouters, Richard Frijnts, and Rik

Tinnemans for excellent technical assistance and for performing the animal

experiments.

DisclosuresR.A.M. and R.E.V. have applied for a patent on the use of proteasome inhib-

itors for plasma cell depletion. The other authors have no financial conflicts

of interest.

References1. Sanders, D. B., and A. Evoli. 2010. Immunosuppressive therapies in myasthenia

gravis. Autoimmunity 43: 428–435.2. Shapiro-Shelef, M., and K. Calame. 2005. Regulation of plasma-cell de-

velopment. Nat. Rev. Immunol. 5: 230–242.3. Miller, J. J., III, and L. J. Cole. 1967. Resistance of long-lived lymphocytes and

plasma cells in rat lymph nodes to treatment with prednisone, cyclophospha-mide, 6-mercaptopurine, and actinomycin D. J. Exp. Med. 126: 109–125.

4. DiLillo, D. J., Y. Hamaguchi, Y. Ueda, K. Yang, J. Uchida, K. M. Haas,G. Kelsoe, and T. F. Tedder. 2008. Maintenance of long-lived plasma cells andserological memory despite mature and memory B cell depletion during CD20immunotherapy in mice. J. Immunol. 180: 361–371.

5. Arce, S., G. Cassese, A. Hauser, T. Dorner, M. Odendahl, R. Manz, A. Radbruch,and F. Hiepe. 2002. The role of long-lived plasma cells in autoimmunity.Immunobiology 206: 558–562.

6. Meister, S., U. Schubert, K. Neubert, K. Herrmann, R. Burger, M. Gramatzki,S. Hahn, S. Schreiber, S. Wilhelm, M. Herrmann, et al. 2007. Extensive im-

munoglobulin production sensitizes myeloma cells for proteasome inhibition.Cancer Res. 67: 1783–1792.

7. Neubert, K., S. Meister, K. Moser, F. Weisel, D. Maseda, K. Amann, C. Wiethe,T. H. Winkler, J. R. Kalden, R. A. Manz, and R. E. Voll. 2008. The proteasomeinhibitor bortezomib depletes plasma cells and protects mice with lupus-likedisease from nephritis. Nat. Med. 14: 748–755.

8. Obeng, E. A., L. M. Carlson, D. M. Gutman, W. J. Harrington, Jr., K. P. Lee, andL. H. Boise. 2006. Proteasome inhibitors induce a terminal unfolded proteinresponse in multiple myeloma cells. Blood 107: 4907–4916.

9. Groll, M., C. R. Berkers, H. L. Ploegh, and H. Ovaa. 2006. Crystal structure ofthe boronic acid-based proteasome inhibitor bortezomib in complex with theyeast 20S proteasome. Structure 14: 451–456.

10. Hemeryck, A., R. Geerts, J. Monbaliu, S. Hassler, T. Verhaeghe, L. Diels,W. Verluyten, L. van Beijsterveldt, R. N. Mamidi, C. Janssen, and R. De Coster.2007. Tissue distribution and depletion kinetics of bortezomib and bortezomib-related radioactivity in male rats after single and repeated intravenous injectionof 14C-bortezomib. Cancer Chemother. Pharmacol. 60: 777–787.

11. Vrolix, K., J. Fraussen, P. C. Molenaar, M. Losen, V. Somers, P. Stinissen,M. H. De Baets, and P. Martınez-Martınez. 2010. The auto-antigen repertoire inmyasthenia gravis. Autoimmunity 43: 380–400.

12. Hoch, W., J. McConville, S. Helms, J. Newsom-Davis, A. Melms, andA. Vincent. 2001. Auto-antibodies to the receptor tyrosine kinase MuSK inpatients with myasthenia gravis without acetylcholine receptor antibodies. Nat.Med. 7: 365–368.

13. Vrolix, K., E. H. Niks, R. Le Panse, M. M. van Ostaijen-ten Dam, A.-H. Muris,C. M. Jol-van der Zijde, M. J. van Tol, M. Losen, P. C. Molenaar, E. J. J. vanZoelen, et al. 2010. Reduced thymic expression of ErbB receptors without auto-antibodies against synaptic ErbB in myasthenia gravis. J. Neuroimmunol. DOI:10.1016/j.jneuroim.2010.10.024.

14. Gajdos, P., S. Chevret, and K. Toyka. 2002. Plasma exchange for myastheniagravis. Cochrane Database Syst. Rev. (4):):CD002275.

15. Gajdos, P., S. Chevret, and K. Toyka. 2008. Intravenous immunoglobulin formyasthenia gravis. Cochrane Database Syst. Rev. (1):):CD002277.

16. Palace, J., J. Newsom-Davis, B. Lecky; Myasthenia Gravis Study Group. 1998.A randomized double-blind trial of prednisolone alone or with azathioprine inmyasthenia gravis. Neurology 50: 1778–1783.

17. Skeie, G. O., S. Apostolski, A. Evoli, N. E. Gilhus, I. Illa, L. Harms, D. Hilton-Jones, A. Melms, J. Verschuuren, and H. W. Horge. 2006. Guidelines for thetreatment of autoimmune neuromuscular transmission disorders. Eur. J. Neurol.13: 691–699.

18. Trivedi, J. R., and G. I. Wolfe. 2010. Myasthenia gravis therapy and thymec-tomy. In Myasthenia Gravis: Disease Mechanisms and Immune Intervention.P. Christadoss, ed. Linus Publications, Deer Park, NY, p. 55–79.

19. De Baets, M., M. Stassen, M. Losen, X. Zhang, and B. Machiels. 2003. Im-munoregulation in experimental autoimmune myasthenia gravis: about T cells,antibodies, and endplates. Ann. N. Y. Acad. Sci. 998: 308–317.

20. Lindstrom, J. 1980. Experimental autoimmune myasthenia gravis. J. Neurol.Neurosurg. Psychiatry 43: 568–576.

21. Martınez-Martınez, P., M. Losen, H. Duimel, P. Frederik, F. Spaans, P. Molenaar,A. Vincent, and M. H. De Baets. 2007. Overexpression of rapsyn in rat muscleincreases acetylcholine receptor levels in chronic experimental autoimmunemyasthenia gravis. Am. J. Pathol. 170: 644–657.

22. Gomez, A. M., J. Van Den Broeck, K. Vrolix, S. P. Janssen, M. A. Lemmens,E. Van Der Esch, H. Duimel, P. Frederik, P. C. Molenaar, P. Martınez-Martınez,et al. 2010. Antibody effector mechanisms in myasthenia gravis: pathogenesis atthe neuromuscular junction. Autoimmunity 43: 353–370.

23. Losen, M., P. Martınez-Martınez, M. Phernambucq, J. Schuurman, P. W. Parren,and M. H. De Baets. 2008. Treatment of myasthenia gravis by preventing ace-tylcholine receptor modulation. Ann. N. Y. Acad. Sci. 1132: 174–179.

24. Lennon, V. A., J. M. Lindstrom, and M. E. Seybold. 1975. Experimental auto-immune myasthenia: a model of myasthenia gravis in rats and guinea pigs. J.Exp. Med. 141: 1365–1375.

25. Sinn, D. I., S. T. Lee, K. Chu, K. H. Jung, E. H. Kim, J. M. Kim, D. K. Park,E. C. Song, B. S. Kim, S. S. Yoon, et al. 2007. Proteasomal inhibition in in-tracerebral hemorrhage: neuroprotective and anti-inflammatory effects of bor-tezomib. Neurosci. Res. 58: 12–18.

26. Cavaletti, G., A. Gilardini, A. Canta, L. Rigamonti, V. Rodriguez-Menendez,C. Ceresa, P. Marmiroli, M. Bossi, N. Oggioni, M. D’Incalci, and R. De Coster.2007. Bortezomib-induced peripheral neurotoxicity: a neurophysiological andpathological study in the rat. Exp. Neurol. 204: 317–325.

27. Verschuuren, J. J., F. Spaans, and M. H. De Baets. 1990. Single-fiber electro-myography in experimental autoimmune myasthenia gravis. Muscle Nerve 13:485–492.

28. Hoedemaekers, A., Y. Graus, P. van Breda Vriesman, and M. de Baets. 1997.Age- and sex-related resistance to chronic experimental autoimmune myastheniagravis (EAMG) in Brown Norway rats. Clin. Exp. Immunol. 107: 189–197.

29. Hoedemaekers, A. C., J. J. Verschuuren, F. Spaans, Y. F. Graus, S. Riemersma,P. J. van Breda Vriesman, and M. H. De Baets. 1997. Age-related susceptibilityto experimental autoimmune myasthenia gravis: immunological and electro-physiological aspects. Muscle Nerve 20: 1091–1101.

30. Lightcap, E. S., T. A. McCormack, C. S. Pien, V. Chau, J. Adams, andP. J. Elliott. 2000. Proteasome inhibition measurements: clinical application.Clin. Chem. 46: 673–683.

31. Moreau, P., V. Coiteux, C. Hulin, X. Leleu, H. van de Velde, M. Acharya, andJ. L. Harousseau. 2008. Prospective comparison of subcutaneous versus in-travenous administration of bortezomib in patients with multiple myeloma.Haematologica 93: 1908–1911.

2512 PLASMA CELL DEPLETION WITH BORTEZOMIB IN EAMG

on October 4, 2011

ww

w.jim

munol.org

Dow

nloaded from

32. Losen, M., M. H. Stassen, P. Martınez-Martınez, B. M. Machiels, H. Duimel,P. Frederik, H. Veldman, J. H. Wokke, F. Spaans, A. Vincent, and M. H. DeBaets. 2005. Increased expression of rapsyn in muscles prevents acetylcholinereceptor loss in experimental autoimmune myasthenia gravis. Brain 128: 2327–2337.

33. Janssen, S. P., M. Phernambucq, P. Martinez-Martinez, M. H. De Baets, andM. Losen. 2008. Immunosuppression of experimental autoimmune myastheniagravis by mycophenolate mofetil. J. Neuroimmunol. 201-202: 111–120.

34. Martınez-Martınez, P., M. Phernambucq, L. Steinbusch, L. Schaeffer, S. Berrih-Aknin, H. Duimel, P. Frederik, P. Molenaar, M. H. De Baets, and M. Losen.2009. Silencing rapsyn in vivo decreases acetylcholine receptors and augmentssodium channels and secondary postsynaptic membrane folding. Neurobiol. Dis.35: 14–23.

35. Engel, A. G., and T. Santa. 1971. Histometric analysis of the ultrastructure of theneuromuscular junction in myasthenia gravis and in the myasthenic syndrome.Ann. N. Y. Acad. Sci. 183: 46–63.

36. Manz, R. A., A. E. Hauser, F. Hiepe, and A. Radbruch. 2005. Maintenance ofserum antibody levels. Annu. Rev. Immunol. 23: 367–386.

37. Slifka, M. K., and R. Ahmed. 1998. Long-lived plasma cells: a mechanism formaintaining persistent antibody production. Curr. Opin. Immunol. 10: 252–258.

38. Hawkey, C. J., J. Newsom-Davis, and A. Vincent. 1981. Plasma exchange andimmunosuppressive drug treatment in myasthenia gravis: no evidence for syn-ergy. J. Neurol. Neurosurg. Psychiatry 44: 469–475.

39. Olarte, M. R., R. S. Schoenfeldt, A. S. Penn, R. E. Lovelace, and L. P. Rowland.1981. Effect of plasmapheresis in myasthenia gravis 1978–1980. Ann. N. Y.Acad. Sci. 377: 725–728.

40. Bruna, J., E. Udina, A. Ale, J. J. Vilches, A. Vynckier, J. Monbaliu,L. Silverman, and X. Navarro. 2010. Neurophysiological, histological and im-

munohistochemical characterization of bortezomib-induced neuropathy in mice.Exp. Neurol. 223: 599–608.

41. Kane, R. C., P. F. Bross, A. T. Farrell, and R. Pazdur. 2003. Velcade: U.S. FDAapproval for the treatment of multiple myeloma progressing on prior therapy.Oncologist 8: 508–513.

42. Engel, A. G., H. Sakakibara, K. Sahashi, J. M. Lindstrom, E. H. Lambert, andV. A. Lennon. 1979. Passively transferred experimental autoimmune myastheniagravis: sequential and quantitative study of the motor end-plate fine structure andultrastructural localization of immune complexes (IgG and C3), and of theacetylcholine receptor. Neurology 29: 179–188.

43. Maseda, D., S. Meister, K. Neubert, M. Herrmann, and R. E. Voll. 2008. Pro-teasome inhibition drastically but reversibly impairs murine lymphocyte de-velopment. Cell Death Differ. 15: 600–612.

44. Souroujon, M. C., T. Brenner, and S. Fuchs. 2010. Development of novel ther-apies for MG: studies in animal models. Autoimmunity 43: 446–460.

45. Pestronk, A., D. B. Drachman, R. Teoh, and R. N. Adams. 1983. Combinedshort-term immunotherapy for experimental autoimmune myasthenia gravis.Ann. Neurol. 14: 235–241.

46. Ubiali, F., S. Nava, V. Nessi, R. Longhi, G. Pezzoni, R. Capobianco,R. Mantegazza, C. Antozzi, and F. Baggi. 2008. Pixantrone (BBR2778) reducesthe severity of experimental autoimmune myasthenia gravis in Lewis rats. J.Immunol. 180: 2696–2703.

47. Karussis, D. M., D. Lehmann, T. Brenner, I. Wirguin, R. Mizrachi-Koll,C. Sicsic, and O. Abramsky. 1994. Immunomodulation of experimental auto-immune myasthenia gravis with linomide. J. Neuroimmunol. 55: 187–193.

48. Abramsky, O., R. Tarrab-Hazdai, A. Aharonov, and S. Fuchs. 1976. Immuno-suppression of experimental autoimmune myasthenia gravis by hydrocortisoneand azathioprine. J. Immunol. 117: 225–228.

The Journal of Immunology 2513

on October 4, 2011

ww

w.jim

munol.org

Dow

nloaded from


Recommended