+ All Categories
Home > Documents > Chimerism and cure: hematologic and pathologic correction of murine sickle cell disease

Chimerism and cure: hematologic and pathologic correction of murine sickle cell disease

Date post: 29-Nov-2023
Category:
Upload: independent
View: 0 times
Download: 0 times
Share this document with a friend
13
doi:10.1182/blood-2003-03-0712 Prepublished online August 21, 2003; 2003 102: 4582-4593 Andrew B. Adams, Christian P. Larsen, Lewis L. Hsu and David R. Archer Leslie S. Kean, Elizabeth A. Manci, Jennifer Perry, Can Balkan, Shana Coley, David Holtzclaw, sickle cell disease Chimerism and cure: hematologic and pathologic correction of murine http://bloodjournal.hematologylibrary.org/content/102/13/4582.full.html Updated information and services can be found at: (1880 articles) Transplantation (1174 articles) Red Cells Articles on similar topics can be found in the following Blood collections http://bloodjournal.hematologylibrary.org/site/misc/rights.xhtml#repub_requests Information about reproducing this article in parts or in its entirety may be found online at: http://bloodjournal.hematologylibrary.org/site/misc/rights.xhtml#reprints Information about ordering reprints may be found online at: http://bloodjournal.hematologylibrary.org/site/subscriptions/index.xhtml Information about subscriptions and ASH membership may be found online at: Copyright 2011 by The American Society of Hematology; all rights reserved. Washington DC 20036. by the American Society of Hematology, 2021 L St, NW, Suite 900, Blood (print ISSN 0006-4971, online ISSN 1528-0020), is published weekly For personal use only. by guest on June 3, 2013. bloodjournal.hematologylibrary.org From
Transcript

doi:10.1182/blood-2003-03-0712Prepublished online August 21, 2003;2003 102: 4582-4593   

 Andrew B. Adams, Christian P. Larsen, Lewis L. Hsu and David R. ArcherLeslie S. Kean, Elizabeth A. Manci, Jennifer Perry, Can Balkan, Shana Coley, David Holtzclaw, sickle cell diseaseChimerism and cure: hematologic and pathologic correction of murine

http://bloodjournal.hematologylibrary.org/content/102/13/4582.full.htmlUpdated information and services can be found at:

(1880 articles)Transplantation   � (1174 articles)Red Cells   �

Articles on similar topics can be found in the following Blood collections

http://bloodjournal.hematologylibrary.org/site/misc/rights.xhtml#repub_requestsInformation about reproducing this article in parts or in its entirety may be found online at:

http://bloodjournal.hematologylibrary.org/site/misc/rights.xhtml#reprintsInformation about ordering reprints may be found online at:

http://bloodjournal.hematologylibrary.org/site/subscriptions/index.xhtmlInformation about subscriptions and ASH membership may be found online at:

Copyright 2011 by The American Society of Hematology; all rights reserved.Washington DC 20036.by the American Society of Hematology, 2021 L St, NW, Suite 900, Blood (print ISSN 0006-4971, online ISSN 1528-0020), is published weekly    

For personal use only. by guest on June 3, 2013. bloodjournal.hematologylibrary.orgFrom

RED CELLS

Chimerism and cure: hematologic and pathologic correction of murinesickle cell diseaseLeslie S. Kean, Elizabeth A. Manci, Jennifer Perry, Can Balkan, Shana Coley, David Holtzclaw, Andrew B. Adams,Christian P. Larsen, Lewis L. Hsu, and David R. Archer

Bone marrow transplantation (BMT) is theonly curative therapy for sickle cell dis-ease (SCD). However, the morbidity andmortality related to pretransplantation my-eloablative chemotherapy often out-weighs the morbidity of SCD itself, thusseverely limiting the number of patientseligible for transplantation. Although non-myeloablative transplantation is expectedto reduce the risk of BMT, it will likelyresult in mixed-chimerism rather thancomplete replacement with donor stemcells. Clinical application of nonmyeloab-lative transplantation thus requires knowl-

edge of the effect of mixed chimerism onSCD pathophysiology. We have, there-fore, created a panel of transplanted SCDmice that received transplants displayingan array of red blood cell (RBC) and whiteblood cell (WBC) chimerism. A significantenrichment of RBC over WBC chimerismoccurred in these mice, because of thedramatic survival advantage of donor oversickle RBCs in the peripheral blood. In-creasing levels of RBC chimerism pro-vided progressive correction of hemato-logic and pathologic abnormalities.However, sickle bone marrow and splenic

hematopoiesis was not corrected untilperipheral blood sickle RBCs were fullyreplaced with donor RBCs. These resultshave important and unexpected implica-tions for nonmyeloablative BMT for SCD.As the critical hematopoietic organs werenot corrected without full RBC replace-ment, 100% peripheral blood RBC chimer-ism becomes the most important bench-mark for cure after nonmyeloablative BMT.(Blood. 2003;102:4582-4593)

© 2003 by The American Society of Hematology

Introduction

Although bone marrow transplantation (BMT) remains the onlycure for sickle cell disease (SCD), the morbidity and mortalityassociated with conventional myeloablative BMT has limited itsuse to those patients who suffer the most devastating diseasecomplications.1 Furthermore, the limitations of posttransplantationimmunosuppression make BMT available only to those patientswith immunologically matched donors. Together, these 2 limita-tions severely curtail the number of patients offered BMT, with 1study estimating that of all the potential transplant recipients, onlyapproximately 1% met strict criteria for eligibility and also had anavailable HLA-matched donor.1 Given these serious limits toconventional BMT, there is growing interest in developing nonmy-eloablative protocols designed to induce mixed hematopoieticchimerism rather than complete replacement with donor marrow.2

The vast clinical experience with transient mixed red blood cell(RBC) chimerism after simple or exchange transfusion,3 as well asa small number of patients who received conventional transplantswho serendipitously developed mixed chimerism,2,4 and had im-provement in SCD complications,2,4 suggests that this approachmay be a clinically feasible method of treating many of thecomplications of SCD. A key question then becomes: What level ofmixed RBC chimerism is sufficient to provide long-term correctionof the complications of SCD?

We have previously shown that mixed white blood cell (WBC)chimerism and transplantation tolerance across allogeneic barrierscan be achieved in mice when a nonmyeloablative conditioningdose of busulfan is used in conjunction with peritransplantationT-cell costimulation blockade of the B7 and CD40 costimulationpathways.5 That work demonstrated that, although chimerismacross major histocompatibility complex (MHC) barriers failed todevelop in the absence of busulfan pretreatment, in the presence ofbusulfan and costimulation blockade stable chimerism was titrat-able by increasing the dose of transplanted bone marrow. In acongenic transplantation model, low levels of chimerism devel-oped even in the absence of busulfan pretreatment when largenumbers of donor bone marrow cells were infused.5

We next showed that in a murine model of SCD, the combina-tion of busulfan, bone marrow, and peritransplantation costimula-tion blockade resulted in mixed WBC chimerism, and full replace-ment of the peripheral RBC compartment in 8 of 11 mice thatreceived transplants and that these mice demonstrated cure of SCDpathophysiology.6 Interestingly, in that study, a small proportion ofmice that did not receive busulfan pretreatment developed signifi-cant peripheral blood RBC chimerism even in the setting ofextremely low levels of WBC chimerism.6 In agreement with ourand others’ previous work showing that sickle RBCs are cleared

From the Division of Hematology/Oncology/BMT, Department of Pediatrics,Emory University School of Medicine, Atlanta, GA; Centralized Pathology Unitfor Sickle Cell Disease at University of South Alabama College of Medicine,Mobile; Department of Pediatrics, Division of Hematology, Ege UniversitySchool of Medicine, Izmir, Turkey; and the Department of Surgery, EmoryUniversity School of Medicine, Atlanta, GA.

Submitted March 6, 2003; accepted August 10, 2003. Prepublished online asBlood First Edition Paper, August 21, 2003; DOI 10.1182/blood-2003-03-0712.

Supported in part by research grants DK/AI40519 (C.P.L.), CA74364-03(C.P.L.), AI44644 (C.P.L.), R29HL60127 (D.R.A.), CURE Childhood Cancer

(D.R.A.), The Jill Andrews Beat Leukemia Celebrity Classic (D.R.A.), theCarlos and Marguerite Mason Trust (C.P.L.), and a grant from the NationalHeart, Lung, and Blood Institute (N01-HB-07086) (E.M.).

Reprints: David R. Archer, Division of Hem/Onc/BMT, Department ofPediatrics, Emory University School of Medicine, 2040 Ridgewood Dr NE,Atlanta, GA 30322; e-mail: [email protected].

The publication costs of this article were defrayed in part by page chargepayment. Therefore, and solely to indicate this fact, this article is herebymarked ‘‘advertisement’’ in accordance with 18 U.S.C. section 1734.

© 2003 by The American Society of Hematology

4582 BLOOD, 15 DECEMBER 2003 � VOLUME 102, NUMBER 13

For personal use only. by guest on June 3, 2013. bloodjournal.hematologylibrary.orgFrom

extremely rapidly from peripheral blood,7,8 these results suggestedsignificant enrichment of RBC versus WBC chimerism and furtherthat the RBC chimerism may have a substantial ameliorative effecton SCD pathophysiology.6 However, that previous work did notallow us to determine the threshold of WBC and RBC chimerismnecessary to cure SCD.

To begin to answer the question of the threshold of chimerismnecessary to cure SCD, Iannone et al9 showed that when murinesickle bone marrow was transplanted into healthy recipients, adose-dependent increase in sickle pathology occurred. However,because that study focused on “reverse chimeras,” in which healthymice were used as transplant recipients, the effect of mixedchimerism on established sickle pathology could not be addressed.Although our previous work produced a small number of mice withmixed RBC chimerism, the vast majority showed full replacementof the peripheral RBC compartment with donor RBCs, therebypreventing assessment of mixed RBC chimerism on the sicklephenotype.6 Given the importance of determining the threshold ofRBC chimerism for cure of SCD, we have now created a panel oftransplanted sickle mice that received transplants with increasinglevels of RBC chimerism. This study has allowed us to addressmechanistic questions that are of paramount importance to nonmy-eloablative transplantation for SCD. First, we determined themechanism of enrichment of donor over sickle RBCs that occurredin sickle mice receiving transplants. Second, we analyzed the effectof increasing amounts of donor RBCs on hematologic, physiologic,and pathologic parameters to determine the minimal level of RBCchimerism that consistently gave a total-body cure. Our resultsprovide the first chimerism targets for nonmyeloablative BMT forSCD and suggest that the most rigorous benchmark for cure ofSCD is full replacement of the peripheral blood with donor RBCs.

Materials and methods

Animals

Sickle mice were originally supplied by Dr Paszty (then at the LawrenceBerkley National Laboratory10). They express exclusively human �- andsickle �-globin and were originally bred by selective mating and exist on amixed genetic background (strains FVB/N, 129, DBA/2, C57BL/6, andBlack Swiss10). Expression of exclusively sickle �-globin in sickle micewas confirmed by differential hemoglobin electrophoresis as described in“Analysis of RBC chimerism.” Enhanced green fluorescent protein (eGFP)mice (B6-TgN(�-act-EGFP)osbY01) were generously supplied by DrOkabe.11 Both the eGFP and sickle strains are now maintained at EmoryUniversity according to recommendations of the institutional animal careand use committee (IACUC). Sickle transplant recipients were both maleand female, aged 7 to 12 weeks. Transplant recipients and donors werealways both age- and sex-matched prior to BMT. Although the donor eGFPmice and recipient sickle mice share homology at the MHC locus (both areH2-Kb), they originate from different strain backgrounds10,11 and areexpected to be mismatched at a variety of minor histocompatibility loci.

BMT protocol

Bone marrow was obtained from donor eGFP mice by flushing femurs andtibiae with normal saline through a 23-guage needle. Nucleated cells werecounted, and the bone marrow was suspended at the following concentra-tions: 20 � 107/mL, 15 � 107/mL, 10 � 107/mL, 5 � 107/mL, and 1 � 107/mL. A volume of 100 �L of each of these suspensions was injectedintravenously (through the retroorbital venus plexus) into groups of 4 to 8sickle mice so that the final dose of bone marrow transplanted was20 � 106, 15 � 106, 10 � 106, 5 � 106, and 1 � 106 cells, respectively. Inaddition, given that donor and recipient animals were expected to havemultiple minor histocompatibility mismatches, all mice that received

transplants also received 500 �g each of hamster antimouse-CD40Lmonoclonal antibody (mAb; MR1; BioExpress, Lebanon, NH) and humanCTLA4-immunoglobulin (Ig; Bristol-Myers Squibb, Princeton, NJ) (forcostimulation blockade) intraperitoneally on days 0, 2, 4, and 7 relative toBMT. Control mice received preconditioning with busulfan (Busulfex; 20mg/kg intraperitoneally; Orphan Medical, Minnetonka, MN) on day �1,transplant of 20 � 106 eGFP bone marrow cells on day 0, and 500�g eachof anti-CD40L and CTLA4-Ig on days 0, 2, 4, and 7.

Analysis of RBC chimerism

RBC chimerism was determined by differential hemoglobin electrophoresisof donor murine “single” �-globin and recipient human sickle �-globin.6,10

Electrophoresis was performed with the Helena Titan III electrophoresissystem (Helena Laboratories, Beaumont, TX). The percentage of donor orrecipient hemoglobin was determined by densitometry with the use ofKodak 1-D Image Analysis software (Kodak, Rochester, NY).

Flow cytometry

Peripheral blood was analyzed by staining with fluorochrome-conjugatedantibodies followed by RBC lysis and washing with a whole blood lysis kit(R&D Systems, Minneapolis, MN). At 6 to 10 months after transplantation,bone marrow was obtained by flushing the femurs and tibiae of mice thatreceived transplants with Hanks balanced saline solution (HBSS; Sigma, StLouis, MO), and splenocytes were obtained by passing spleens through asterile screen into HBSS. Cell suspensions were then counted with ahemocytometer and stained with fluorescent antibodies. Antibodies (Pharm-ingen, San Diego, CA) included anti–Ter-119, anti–pan-CD45, anti-CD45.1, anti–Sca-1, anti–c-kit, anti-B220, anti-CD3, anti–Mac-1, anti–GR-1, anti–H2-Kb, and Annexin-V. Dead cells were identified by stainingwith propidium iodide (PI; Sigma) just prior to flow cytometry, and PI�

cells were excluded from subsequent analysis. Both directly fluorochrome-conjugated and biotinylated antibodies were used. When biotinylatedantibodies were used, a secondary incubation with Streptavidin conjugatedto allophycocyanin (APC), Cychrome, or APC-Cy7 (Pharmingen) wasused. Flow cytometry was performed on FACScan (for 3-color analysis),FACScalibur (for 4-color analysis), or FACSVantage (for 5-color analysis)flow cytometers (Becton Dickinson, Mountain View, CA) and analyzed byusing WinList analysis software (Verity Software House, Topsham, ME).

Analysis of bone marrow and spleen stem cells

Stem cells were defined as Sca-1�/c-kit�/Ter-119�/B220�/CD3�/Mac-1�/GR-1� (Sca-1�/c-kit�/Lineage�)12 cells in the bone marrow and spleen. Toidentify these cells, bone marrow cells and splenocytes were stained withdirectly conjugated antibodies to Sca-1 and c-kit and with biotinylatedantibodies to the lineage markers. After incubation with Strepdavidin-APC-Cy7 and staining with PI to exclude dead cells, flow cytometry wasperformed by using a FACSVantage flow cytometer. The percentage of totalSca-1�/c-kit�/Lineage� cells that were either donor (eGFP�) or recipient(eGFP�) was identified by using WinList analysis software.

Analysis of RBC ontogeny

The ontogeny of RBC progenitors was determined as previously de-scribed.13 Briefly, bone marrow cells and splenocytes were triple labeledwith Ter-119, CD71, and PI (to exclude dead cells). Four populations wereidentified for further analysis: region I (proerythroblasts, Ter-119med/CD71high), region II (basophilic erythroblasts, Ter-119high/CD71high), regionIII (late basophilic and chromatophilic erythroblasts, Ter-119high/CD71med),and region IV (orthochromatophilic erythroblasts, Ter-119high/CD71low).13

The percentage of each population in relation to total Ter-119�/PI� cellswas determined by using WinList flow cytometry analysis software.

Analysis of RBC progenitor engraftment

As others have previously shown, there is a subpopulation of early RBCprogenitors in the bone marrow and spleen that express CD45 in addition toTer-119 and CD71.14 These cells also express eGFP and thus could be easily

MIXED CHIMERISM AND CURE OF MURINE SCD 4583BLOOD, 15 DECEMBER 2003 � VOLUME 102, NUMBER 13

For personal use only. by guest on June 3, 2013. bloodjournal.hematologylibrary.orgFrom

distinguished as donor (Ter-119�/CD71�/pan-CD45�/eGFP�/CD45.1�) orrecipient (Ter-119�/CD71�/pan-CD45�/eGFP�/CD45.1�). Bone marrowcells and splenocytes were thus labeled with Ter-119, CD71, CD45, andCD45.1 and analyzed flow cytometrically with the use of a FACSVantageflow cytometer. The percentage of total Ter-119�/CD71�/CD45� cells thatwere either donor (eGFP�/CD45.1�) or recipient (eGFP�/CD45.1�) wasdetermined with WinList analysis software to directly measure the percent-age of engraftment of donor RBC progenitors.

Determining hematopoietic balance (lymphomyeloid/erythroidratio) in the hematopoietic organs

Hematopoietic balance was determined as the percentage of total bonemarrow cells or splenocytes that were either lymphomyeloid (LM; CD45�/PI�) or erythroid (E; Ter-119�/PI�).

Analysis of hematologic characteristics

Complete blood counts were performed on a Hemavet 1500 blood analyzer(CDC Technologies, Oxford, CT). Reticulocytes were analyzed flowcytometrically and were defined as the percentage of peripheral bloodcells that were Ter-119� and Thiazole-Orange� (a nucleic acid stainingdye; Sigma).

Urine osmolarity

Urine (10 �L) was collected from both mice that received transplants andcontrol mice that did not receive transplants. For consistency, this collectionwas always performed in the early morning, although for mice (in contrastto humans), early morning urine does not represent the most concentratedsample. Urine osmolarity was then determined by using a Vapro osmometer(Wescor, Logan UT) according to manufacturer’s instructions.

Soluble vascular cell adhesion molecule measurement

Soluble vascular cell adhesion molecule (sVCAM) concentration in serumwas determined by enzyme-linked immunosorbent assay (ELISA) with theuse of a commercially available kit (R&D Systems).

RBC half-life

RBC population half-life was determined by a pulsed biotinylationexperiment.6,15 N-hydroxysuccidimide biotin (50 mg/kg; Calbiochem, SanDiego, CA) was injected into animals that received transplants or controlanimals. The percentage of biotinylated peripheral RBCs was determinedby staining for RBCs with Ter-119 and for biotin with fluorescent-labeledStreptavidin (Ter-119�/Strepdavidin-Cychrome�). The percentage of Ter-119�/Strepdavidin-Cychrome� cells remaining over time was analyzedflow cytometrically to determine the half-life of the RBC population. Giventhe chimerism present in the mice that received transplants, there was abiphasic nature to the RBC half-life curves, indicating the presence of 2separate RBC populations (short-lived sickle RBCs and long-lived donorRBCs) with dramatically different RBC life spans. Although these 2populations clearly have separate RBC life spans, thus precluding aninterpolation of a single RBC half-life in the chimeric mice, the time atwhich half the biotinylation signal had disappeared (referred to as thebiotinylation50 time) could be directly interpolated from the biotinylationdecay curves.

Analysis of pathology

Dissections, weights, and microscopic studies were performed by 1pathologist (E.A.M.) to provide uniformity of diagnoses. Tissue was fixedin neutral-buffered formalin at room temperature for at least a week,dehydrated in graded alcohols, embedded in paraffin, cut at 5 to 7 �m, andstained with hematoxylin and eosin. Three levels of each block were cut.Special stains, including Masson trichrome and Gomori-modified ironstains, were accompanied by appropriate controls. Although sickling of theerythrocytes made blinding of the pathologist to presence or absence ofsickle hemoglobin impossible, blinding to the percentage of correction was

maintained throughout the microscopic studies. Tissues surveyed includedbrain, heart, lungs, thymus, liver, pancreas, kidneys, gonads, adrenals, bonemarrow, eyes, skeletal muscle, adipose, mesentery, penis, intestines, lymphnodes, thyroid, spinal cord, and bone. Microscopic sections were studied forchanges detectable by routine light microscopy, and the changes weresubjectively graded on a scale of 0 (absence) to 4� (abundant/severe). Thefindings that were present most consistently included cardiac vascularectasia (dilatation of the medium-sized arteries), increased thickness of themedia of the pulmonary arteries (measured by ocular micrometer), ectasiaof medium-sized pulmonary arteries, hepatic fibrosis consistent withremote infarct (focal increase in fibrous tissue by trichrome stain), recenthepatic infarcts (coagulative necrosis with/without inflammatory infiltratesand pigmented histiocytes), hepatic iron deposition (increased staining onGomori-modified iron stain in both hepatocytes and Kupffer cells), renalglomerular enlargement (increased diameter of appropriately orientedglomerular tufts measured by ocular micrometer), renal mesangial glomer-ular changes (focal increase in mesangial tissue), renal tubular irondeposition, splenic red pulp/white pulp architectural integrity, splenichematopoiesis (increased amount of hematopoietic cells within the redpulp), and estimation of ongoing injury in heart, lungs liver, kidneys, orspleen (sickle-related changes of chronic ischemia).

Results

Titratable chimerism after nonmyeloablative transplantationin murine SCD

We used the transgenic eGFP strain of mice as bone marrow donorsfor transplantation into sickle mice and produced a panel of animalswith a wide range of WBC and RBC chimerism (Figure 1A). WBCchimerism was easily detected by eGFP fluorescence without theneed for antibody labeling. RBC chimerism was detected by thedifferential electrophoretic mobilities of donor and recipient hemo-globin isoforms (Figure 1B). The donor eGFP mice and therecipient sickle mice were MHC-matched at MHC class I, bothbeing H-2Kb.10,11 Thus, as we have previously shown,5 we antici-pated that acquisition of transplantation tolerance and donorchimerism with this transplant pair would occur readily, withoutthe need for pretransplantation myelosuppression with busulfan.However, the eGFP donor mice and the sickle recipients werederived from different strain backgrounds,5,10,11 thus, despite beingMHC-matched, these 2 strains were likely to have multiple minorhistocompatibility mismatches. We, therefore, included our well-established method of inducing transplantation tolerance throughT-cell costimulation blockade as part of the transplantation regi-men.5,6 We transplanted sickle mice with either 1 � 106 (n � 5),5 � 106 (n � 5), 10 � 106 (n � 5), 15 � 106 (n � 5), or 20 � 106

(n � 8) whole eGFP bone marrow cells on day 0 and gavecostimulation blockade (with anti-CD40L and CTLA4-Ig) on days0, 2, 4, and 7. As a positive control for engraftment an additional 4mice were pretreated with busulfan (20 mg/kg) before transplanta-tion of 20 � 106 whole bone marrow cells and treatment withanti-CD40L and CTLA4-Ig.6 Of the 32 mice that received trans-plants, 5 died during procedures (anesthesia- or phlebotomy-related deaths) prior to any analysis, and an additional 4 wereanalyzed partially (up to 3 months after transplantation) but diedprior to terminal experiments. The remaining 23 mice wereanalyzed for 6 to 10 months prior to terminal experiments.Dose-dependent chimerism developed, with the highest levelsoccurring after pretreatment with busulfan (Figure 1A-C). Micethat received very low doses of bone marrow (1 � 106 or 5 � 106

cells) developed 0.1% to 16% WBC chimerism and 5% to 79%RBC chimerism (Figure 1A,C). In all but 1 mouse that receivedmore than 10 � 106 bone marrow cells, RBC chimerism was more

4584 KEAN et al BLOOD, 15 DECEMBER 2003 � VOLUME 102, NUMBER 13

For personal use only. by guest on June 3, 2013. bloodjournal.hematologylibrary.orgFrom

than 80% (84%-95%), whereas WBC chimerism was much lower(3%-18%). Multilineage WBC chimerism was present, includingdonor B cells, T cells, macrophages, and granulocytes (data notshown). Busulfan-pretreated mice developed significantly higherWBC chimerism (53%-81%) and complete replacement of theperipheral blood with donor RBCS (Figure 1A,C).

WBC chimerism (defined as the percentage of total CD45�/PI�

cells that were also eGFP�) was similar in both the peripheralblood and the hematopoietic organs and was significantly lowerthan peripheral blood RBC chimerism (Figure 2A-B). Likewise,

stem cell chimerism (defined as the percentage of all Sca-1�/c-kit�/PI�/lineage� cells in either the bone marrow or the spleen that werealso eGFP�)12 showed a similar pattern to peripheral blood WBCchimerism (Figure 2C). Thus, donor stem cell chimerism in thebone marrow and spleen was significantly lower than donor RBCchimerism in the peripheral blood (Figure 2C). Although bonemarrow and splenic stem cell chimerism were both significantlylower than peripheral blood RBC chimerism, the percentage ofstem cells in the spleen that were donor in origin (ie, the percentageof total splenic c-kit�/Sca-1�/PI�/lineage� cells that were eGFP�)

Figure 1. Durable, titratable WBC and RBC chimerism was produced in the sickle mice receiving transplants. (A) Increasing amounts of donor bone marrow yieldedtitratable WBC and RBC chimerism. Unmanipulated bone marrow cells from donor eGFP mice were transplanted into sickle recipients, and chimerism was determined at theterminal time point (6-10 months after transplantation). E � percentage of donor WBCs, Œ � percentage of donor Hb in the peripheral blood, respectively, for individual mice.(B) A wide range of RBC chimerism developed in the mice that received transplants. Shown is a representative hemoglobin electrophoresis with blood from animals thatreceived transplants that demonstrates increasing amounts of donor Hb: From left to right, 0% (sickle control), 17% (no. 1582), 43% (no. 1550), 57.5% (no. 1636), 79% (no.1630), 90% (no. 70), 100% (no. 62), and 100% (eGFP control). (C) Chimerism is stable over time. WBC chimerism was determined as the percentage of eGFP�/CD45� cells inthe peripheral blood. The average WBC chimerism over time for each of the 6 different treatment conditions is shown: � � 1 � 106 donor bone marrow cells, Œ � 5 � 106,‚ � 10 � 106, f � 15 � 106, � � 20 � 106, F � 20 � 106 after pretreatment with 20 mg/kg busulfan (inset). For each bone marrow dose, n � 3-7. Error bars show SEM.

Figure 2. Enrichment of peripheral blood RBC chimerism compared with WBC chimerism in the blood, hematopoietic organs, and stem cells. (A) In the individualmice, a striking enrichment of peripheral blood RBC versus WBC chimerism occurred. The RBC chimerism (expressed as percentage of donor Hb, x-axis) versus WBCchimerism (y-axis) for each of the mice that received transplants is shown (E). Control sickle (0% donor Hb) or eGFP (100% donor Hb) are shown (F). (B) Enrichment ofperipheral RBC chimerism (x-axis) compared with WBC chimerism (y-axis) in the bone marrow (Œ) and spleen (E) occurred in mice that received transplants. Control sicklemice that did not receive transplants (0% donor Hb) or eGFP mice (100% donor Hb) are shown as ‚ (bone marrow) and F (spleen). (C) Enrichment of peripheral RBCchimerism (x-axis) compared with stem cell chimerism (y-axis) in the bone marrow (Œ) and spleen (E) also occurred in the mice that received transplants. The stem cellcompartment was defined as those bone marrow cells or splenocytes that were Sca-1�/c-kit�/ PI�/lineage�. The percentage of these stem cells that were either donor derived(eGFP�/CD45.1�) or recipient derived (eGFP�/CD45.1�) was then determined. Control sickle mice that did not receive transplants (0% donor Hb) or eGFP mice (100% donorHb) are shown as ‚ (bone marrow) and F (spleen). (D) A higher percentage of splenic stem cells than bone marrow stem cells were donor derived in the chimeric mice. Stemcells were defined as described in panel C. The percentage of stem cells that were donor in the bone marrow (x-axis) versus spleen (y-axis) was plotted (F). The line depicts thetheoretical 1:1 ratio of bone marrow to spleen stem cells.

MIXED CHIMERISM AND CURE OF MURINE SCD 4585BLOOD, 15 DECEMBER 2003 � VOLUME 102, NUMBER 13

For personal use only. by guest on June 3, 2013. bloodjournal.hematologylibrary.orgFrom

was significantly higher than the percentage of stem cells in thebone marrow that were donor in origin (Figure 2D). This differencewas not attributable to different organ sizes, as each measurementwas a percentage compared with total numbers of stem cells forthat organ (either bone marrow or spleen). These higher levels ofengraftment may reflect preferential homing or expansion of stemcells in the spleen over bone marrow in the setting of hyperactivesplenic hematopoiesis7 in the sickle mice.

These data show that in murine SCD, even without anypreconditioning with busulfan, significant WBC and RBC chimer-ism could be produced and was stable for more than 6 months afteran MHC-matched BMT. Furthermore, we observed dramaticenrichment of peripheral blood RBC chimerism over WBC chimer-ism in the mice that received transplants.

Enrichment of RBC chimerism is caused by the survivaladvantage of donor RBCs in the peripheral blood

Two models can explain the enrichment in peripheral blood RBCchimerism compared with WBC chimerism in the animals thatreceived transplants: (1) Enhanced survival of healthy RBCs in theperipheral blood compared with the rapidly cleared sickle RBC; (2)enrichment in the hematopoietic organs of donor erythroid progeni-tors, because of increased survival of healthy progenitors or,possibly, enhanced erythroid progenitor engraftment. Although weand others have previously shown that donor RBCs have enhancedsurvival compared with sickle RBCs in the periphery,7,8 noprevious study has investigated the possibility of enrichment at thelevel of an erythroid progenitor. To differentiate these possibilities,we looked at the following 3 parameters:

First, we directly determined the source of early RBC progeni-tors in the hematopoietic organs to investigate whether enhancedengraftment of RBCs compared with WBC progenitors hadoccurred. We studied the small population of early RBC progeni-tors in the bone marrow and spleen that express both Ter-119high

and CD71high and that still express the pan-WBC marker CD45 ontheir cell surface,14 as these cells also express eGFP and thus can beeasily identified as donor or recipient. Donor Ter-119high/CD71high/CD45� cells are eGFP� and CD45.1�, allowing identification ofthe Ter-119high/CD71high/pan-CD45� cells as either donor (eGFP�/CD45.1�) or recipient (eGFP�/CD45.1�) (Figure 3A-B). Thisanalysis showed that the percentage of donor RBC progenitors inmice with less than 100% peripheral RBC chimerism was ex-tremely low and did not increase until full replacement of theperipheral RBC compartment with donor cells had occurred(Figure 3A-B). Thus, progressively increasing engraftment ofdonor RBC progenitors did not account for the enrichment inperipheral blood RBCs over WBC chimerism observed in the micethat received transplants. Furthermore, a comparison of Figure2A-C (which shows the enrichment of peripheral blood RBCchimerism compared with WBC chimerism) with Figure 3A-B(which shows the enrichment of peripheral blood RBC chimerismcompared with bone marrow [Figure 3A] or spleen [Figure 3B]early RBC progenitor chimerism) reveals strikingly similar pat-terns. This finding strengthens the argument that peripheral bloodWBC chimerism, which can be accessed routinely, serves as areliable surrogate for both WBC chimerism in the bone marrowand spleen and RBC progenitor chimerism in these same hemato-poietic organs.

To further explore the relationship between peripheral bloodRBC chimerism and the regulation of hematopoiesis, we examinedthe ontogeny of RBCs from proerythroblasts through orthochro-matophilic erythroblasts in animals that received transplants with

increasing levels of RBC chimerism. Socolovsky et al13 havepreviously shown that labeling with Ter-119 and CD71 allows flowcytometric differentiation of erythroid progenitors. After double-labeling, proerythroblasts (region I) are Ter119med/CD71high. Baso-philic erythroblasts (region II) are Ter119high/CD71high. Late baso-philic and chromatophilic erythroblasts (region III) are Ter119high/CD71med and orthochromatophilic erythroblasts (region IV) areTer119high/CD71low13 (Figure 4). We noted a marked difference inthe balance of erythroid progenitors in the hematopoietic organswhen recipient sickle mice and donor eGFP mice were compared(Figure 4A-B). Thus, although eGFP mice have approximatelyequivalent numbers of early (regions I � II � III) and mature(region IV) erythrocytes in the bone marrow and predominantlymature (region IV) erythrocytes in the spleen (Figure 4A-D), sicklemice have a predominance of early erythroid progenitors (Figure4A-D). In the chimeric mice, the balance of erythroid progenitorsin the hematopoietic organs closely resembled that found in sicklemice that did not receive transplants and did not normalize untilcomplete replacement of the peripheral blood with donor RBCsoccurred (Figure 4B-D). For example, even when more than 80%of the peripheral RBCs were donor in origin, the hematopoieticorgans still produced RBC progenitors in a balance resemblingsickle mice that did not receive transplants, and this production didnot change until fully 100% peripheral RBC chimerism waspresent. These data argued against enhanced survival of donorerythroid progenitors in the hematopoietic organs of chimeric mice,as a predicted shift of erythroid progenitor balance toward donortype did not occur until full replacement with donor RBC existed.Of importance, both the bone marrow and spleen were perfusedwith peripheral blood in the live animals prior to this analysis, and

Figure 3. Enrichment of peripheral blood RBC chimerism compared with RBCprogenitor chimerism in the bone marrow and spleen. Early RBC progenitors(defined as Ter-119high/CD71high/pan-CD45�/PI�) were quantified by flow cytometry,and their origin (donor, eGFP�/CD45.1�) or host (eGFP�/CD45.1�) was determined.In both bone marrow (A) and spleen (B), sickle RBC progenitors predominateddespite near complete replacement of the peripheral blood with donor RBCs. Thex-axis shows the percentage of donor Hb in the peripheral blood. The y-axis depictsthe percentage of eGFP�/CD45.1� donor (E) or eGFP�/CD45.1� sickle (Œ) RBCprogenitors in individual mice receiving transplants. In sickle (0% donor Hb) or eGFP(100% donor Hb) control animals the percentage of donor (F) or host (‚) RBCprogenitors is also shown.

4586 KEAN et al BLOOD, 15 DECEMBER 2003 � VOLUME 102, NUMBER 13

For personal use only. by guest on June 3, 2013. bloodjournal.hematologylibrary.orgFrom

no manipulations were made to remove this blood from thehematopoietic organs prior to flow cytometry. Thus, it is possiblethat some contribution to the erythroid progenitor balance observedin these organs came from the peripheral blood. However, acomparison of the reticulocyte percentage in the peripheral blood(Figure 6C) and the progenitor balance in the bone marrow andspleen (Figure 4) shows this contribution to be minimal. Thus, in arepresentative animal with 90% donor chimerism, the peripheralblood reticulocyte count was only 4% (Figure 6C, closely resem-bling donor mice), but the percentage of early RBC progenitors(regions I � II � III) in the bone marrow and spleen were 84% and44%, respectively, closely resembling sickle mice that did notreceive transplants (Figure 4C-D). The balance of RBC progenitorsin the bone marrow and spleen was, therefore, significantlydifferent than the balance in the peripheral blood. This experimentthus indicates that enhanced survival of erythroid progenitors in thehematopoietic organs does not explain the enrichment of RBCversus WBC chimerism present in mice that received transplants.

Having ruled out enhanced engraftment or survival of donorerythroid progenitors to explain the enrichment of RBC over WBCchimerism in mice that received transplants, we next studied theeffect of differential survival of peripheral blood RBCs on RBCchimerism. We have previously shown that the half-life of RBCs inthe peripheral blood of healthy animals is approximately 20-foldlonger than for sickle RBCs.6,7 Furthermore, we and others havedemonstrated that the mechanism of anemia in sickle mice is therapid clearance of sickle RBCs and reticulocytes from the periph-eral blood and not because of ineffective erythropoiesis in thehematopoietic organs.7,8 To confirm these previous observations,

the peripheral blood of the animals that received transplants wasbiotinylated, and the amount of biotin that remained over time wasmeasured.15 This experiment (Figure 5A-B) showed that, inkeeping with the chimeric nature of the animals that receivedtransplants, there is a biphasic shape to the biotinylation survivalcurves, having both a short component (from the sickle RBCs) anda long component (from the donor RBCs). Given this mixture of 2separate RBC populations, a single RBC half-life could not bedirectly interpolated from the chimeric biotinylation curves. How-ever, the measurement of the time required for half the originalbiotin signal to disappear (referred to here as the biotinylation50

time) did give important information about the correction ofperipheral blood hemolytic anemia in the chimeric animals. Thisanalysis showed that mice with low levels of donor hemoglobin(Hb) had composite biotinylation50 times resembling sickle micethat did not receive transplants, whereas a progressive increase inthis measurement occurred with increasing levels of donor Hb(Figure 5A-B). This correlation between peripheral RBC chimer-ism and RBC biotinylation50 time gives direct support for thehypothesis that enhanced survival of healthy RBCs in the periph-eral blood is the cause of the enrichment in peripheral RBC overWBC chimerism. Furthermore, most of the mice that receivedtransplants had peripheral RBC/WBC chimerism ratios of approxi-mately 20 or less (the ratio of the differential survival of healthyand sickle RBCs as shown in Figure 5 and in our previous work7),indicating that the differential peripheral blood survival of donorover sickle RBCs is sufficient to explain the enrichment in RBCversus WBC chimerism observed in the chimeric mice.

Figure 4. The erythroid progenitor balance in mice receiving transplants closely resembled the sickle mice not receiving transplants until complete replacement ofthe peripheral blood with donor RBCs occurred. (A) Representative flow cytometric analysis of RBC ontogeny in sickle and eGFP spleens. Red cell maturation was mappedby determining the relative proportions of Termed/CD71high (region I), Terhigh/CD71high (region II), Terhigh/CD71med (region III), and Terhigh/CD71low (region IV) cells.13

Representative density-contour maps show the splenic RBC progenitor balance from eGFP and sickle mice. (B) Comparison of regions I to IV from spleens of control sickle (f)and eGFP (u) mice and representative mice with either 90% (z) or 100% (�) peripheral RBC chimerism. This analysis gives a vivid illustration of the similarity of mice with 90%RBC engraftment to sickle mice that did not receive transplants and demonstrates that correction of RBC progenitor balance occurred only after 100% replacement with donorRBCs. (C-D) In both bone marrow (C) and the spleen (D), the RBC progenitor balance did not resemble donor eGFP mice until complete replacement of the peripheral bloodwith donor RBC occurred. For clarity, the percentage of early RBC progenitors (regions I, II, and III) were combined (Œ,‚) and compared with the percentage of mature RBCprogenitors (region IV; F,E). For individual mice that received transplants, the percentage of RBC progenitors in the bone marrow (C) or spleen (D) that resided in region I � II �III (Œ) or IV (E) was determined (y-axis) and plotted against the percentage of donor Hb in the peripheral blood (x-axis). Early RBC progenitors (regions I � II � III) in controlsickle (0% donor Hb) or eGFP (100% donor Hb) mice are shown as [‚]. Mature RBC progenitors (region IV) in control sickle (0% donor Hb) or eGFP (100% donor Hb) mice areshown as F.

MIXED CHIMERISM AND CURE OF MURINE SCD 4587BLOOD, 15 DECEMBER 2003 � VOLUME 102, NUMBER 13

For personal use only. by guest on June 3, 2013. bloodjournal.hematologylibrary.orgFrom

The survival advantage of donor versus sickle RBCs alsoexplains the differential effect of busulfan on WBC versus RBCchimerism in the animals that received transplants. In mice nottreated with busulfan, increasing bone marrow dose led to increas-ing WBC chimerism, ranging from 0.1% to 18% (Figure 1A).Given the survival advantage of healthy versus sickle RBCs, theconcomitant RBC chimerism measured in the peripheral blood wasmuch higher, ranging from 5% to 95%. When mice were pretreatedwith busulfan, chimerism further increased. Peripheral blood WBCchimerism increased from an average of 6.4% without busulfan toan average of 57% with busulfan pretreatment. RBC chimerismalso increased, from an average of 86% when 20 � 106 bonemarrow cells were transplanted without busulfan to 100% withbusulfan pretreatment. Although there appears to be a more subtleeffect of busulfan on RBC chimerism than WBC chimerism, thiseffect is due to reaching the ceiling of 100% chimerism that exists.Because one cannot measure more than 100% donor cells in thechimeric animals, the potential to measure an effect of busulfan onWBC chimerism (from an initial measurement of 6.4% chimerism)was much greater than the potential to measure an effect ofbusulfan on RBC chimerism (from an initial measurement of 86%).However, a comparison of peripheral blood, bone marrow, orspleen WBC chimerism after busulfan pretreatment (aver-age � 57%, 81%, 75%, respectively; Figures 1A and 2A-B) withbone marrow and spleen RBC progenitor chimerism after busulfanpretreatment (average � 63% and 64%, respectively; Figure 3A-B)showed a similar effect of busulfan on both myelolymphoid anderythroid progenitor populations.

Chimeric mice demonstrate normalization of hematologicand physiologic parameters

We next determined the level of RBC chimerism necessary forcorrection of hematologic abnormalities and sickle pathophysiol-ogy. We examined a wide range of parameters in the chimeric mice,including hematocrit (Hct), WBC, reticulocyte percentage, RBCphosphatidylserine (PS) exposure, urine osmolarity, and sVCAMexpression. The Hct and reticulocyte percentage in mice with lowRBC chimerism resembled sickle mice that did not receivetransplants but demonstrated progressive normalization, especiallyin animals with more than 70% to 80% donor Hb (Figure 6A,C).Likewise, the WBC count was reduced toward normal levels withincreasing amounts of RBC chimerism, such that mice with morethan 60% donor Hb had normal WBC counts (Figure 6B). We andothers have shown that sickle RBCs display increased PS on theircell surface that correlates with clinical disease severity and withendothelial adhesion.7,16-20 In a similar manner to the Hct andreticulocyte count, mice with more than 70% to 80% normal levelsof Hb showed minimal RBC PS exposure, whereas those with lowRBC chimerism closely resemble sickle mice that did not receivetransplants (Figure 6D). Sickle mice demonstrate kidney pathol-ogy6,10 and hyposthenuria. Chimeric mice progressively improvedtheir urine-concentrating ability (Figure 6E). This finding provideda key physiologic correlation between correction of hematologicparameters and functional correction of end-organ damage in theanimals that received transplants. Sickle mice and patients withsickle cell disease also demonstrate overexpression of endothelialadhesion molecules that is reflected in increased levels ofsVCAM.21-23 Chimeric mice demonstrated progressive decrease inthe level of sVCAM (Figure 6F). This normalization may havegreat importance, as many of the clinical complications in patientswith sickle cell disease arise from abnormalities in adhesion andinflammatory pathways.

Analysis of pathology reveals progressive correctionwith increasing RBC chimerism

An exhaustive analysis of pathology in the mice that receivedtransplants identified 12 areas of characteristic sickle pathologythat improved after transplantation, including (1) cardiac vascularectasia, (2) pulmonary artery medial thickness, (3) pulmonaryvascular ectasia, (4) remote hepatic infarct, (5) recent hepaticinfarct, (6) hepatocyte iron deposition, (7) hepatic Kupffer cell irondeposition, (8) renal glomerular hypertrophy, (9) renal mesangialhypercellularity, (10) renal tubular iron deposition (11) splenic lossof architectural integrity and vascular congestion, and (12) evi-dence of ongoing multiorgan injury. Evidence of ongoing multior-gan injury included primarily small-vessel changes (ectasia, perivas-cular fibrosis, and congestion), parenchymal chronic ischemicchanges, pericentral vein sclerosis and hepatocytic ischemic changesin the liver, and evidence of ongoing hemolysis (especially in theliver and kidneys). A pathology score of 0 (no pathology) to 4(sickle pathology) was given to each of these 12 characteristics(Table 1). Representative histopathology is shown (Figure 7A-T).The sum of the pathology scores for each mouse was also plottedagainst the percentage of donor Hb (Figure 8). This analysisshowed a progressive correction of organ pathology with increas-ing amounts of donor peripheral RBCs.

Figure 5. Effect of increasing RBC chimerism on the decay kinetics ofbiotinylated peripheral RBCs. (A) Biotinylation curves for representative mice thatreceived transplants and control mice. y-axis � percentage of biotinylated RBCsremaining. x-axis � time (days) after biotinylation. Average survival curves for micewith 0% (sickle mice that did not receive transplants, F), 17% to 43% (E), 61% to 76%(Œ), 87% to 91% (‚), and 100% RBC chimerism (f) are shown. (B) The relationshipbetween the time at which 50% of the biotinylation signal had disappeared(biotinylation50 time, y-axis) and the percentage of donor Hb in the peripheral blood(x-axis) is shown for sickle (0% donor Hb) and eGFP (100% donor Hb) controls (E)and for the mice that received transplants (F).

4588 KEAN et al BLOOD, 15 DECEMBER 2003 � VOLUME 102, NUMBER 13

For personal use only. by guest on June 3, 2013. bloodjournal.hematologylibrary.orgFrom

Hematopoietic cure occurred only with 100% replacementwith donor hemoglobin

Although progressive correction of multiple areas of sickle patho-physiology occurred in the chimeric mice, a notable exception wasthe lack of progressive correction of important hematopoieticabnormalities in the bone marrow and spleen. Given the impor-tance of splenic hematopoiesis in the sickle mice, a detailedexamination of many aspects of splenic function was undertaken.The spleen is greatly enlarged in murine SCD, with sickle spleenstypically representing approximately 5% total body weight com-pared with less than 1% in healthy mice.6,7,10 Given that the spleenis a normal site of hematopoiesis in mice, this enlargement likelyoccurs because of the hemolysis-driven high erythropoietic rate inthese animals in the setting of size-constrained limits on bonemarrow hematopoiesis. Therefore, murine splenic hematopoiesislikely serves as a model of human bone marrow hematopoiesis.7,10

Although the spleen displayed progressive correction in selected

physiologic and pathologic parameters, in others, full RBC replace-ment was required before correction occurred. Thus, althoughspleen architecture (Table 1; Figure 7), size (Figure 6G), and RBCAnnexin-V binding and apoptosis (Figure 6H) all progressivelynormalized with increasing RBC chimerism, other critical indica-tors of hematopoietic regulation remained abnormal without 100%RBC chimerism. The spleens (and bone marrow) of the animalsthat received transplants displayed uncorrected RBC progenitorbalance despite significant (� 80%-90%) RBC chimerism and didnot show correction until 100% RBC chimerism was created(Figure 4A-D). Furthermore, splenic lymphomyeloid-erythroid(LM/E) ratio (highly skewed toward erythropoiesis in sicklemice7,10) resembled sickle mice that did not receive transplantseven with more than 90% RBC chimerism (Figure 6I) and did notnormalize without 100% RBC chimerism. Pathologic analysis ofthe spleen corroborated these results, in that even in animals withsignificant RBC chimerism, splenic hyperactive erythropoiesis

Figure 6. Effect of increasing RBC chimerism on sickle physiology and hematopoiesis. The various physiologic parameters (y-axis) are plotted against peripheral bloodpercentage of donor Hb (x-axis). For panels A-G, sickle (0% donor Hb) and eGFP (100% donor Hb) controls are shown as E. Mice that received transplants are shown as F. (A)Hematocrit versus percentage of donor Hb in the peripheral blood. (B) Peripheral WBC versus percentage of donor Hb in the peripheral blood. (C) Reticulocyte percentageversus percentage of donor Hb in the peripheral blood. (D) Annexin-V binding versus percentage of donor Hb in the peripheral blood. (E) Urine osmolarity versus percentage ofdonor Hb in the peripheral blood. (F) sVCAM concentration versus percentage of donor Hb in the peripheral blood. (G) Spleen weight (as percentage of total body weight)versus percentage of donor Hb in the peripheral blood. (H) Spleen apoptosis versus percentage of donor Hb in the peripheral blood. Apoptosis was determined either byAnnexin-V binding (F,E) or terminal deoxyribonucleotide transferase (TdT)–mediated dUTP nick end labeling (TUNEL) assay (Œ,‚). Sickle (0% donor Hb) and eGFP (100%donor Hb) controls are shown as E and Œ for the Annexin-V binding and TUNEL assays, respectively. Animals that received transplants are shown as F and ‚ for the Annexin-Vbinding and TUNEL assays, respectively. (I) Normalization of lymphomyeloid-erythroid (LM/E) ratio occurred only in mice with 100% donor Hb in the peripheral blood. SplenicLM cells (F,E) were defined as CD45�/PI�. Splenic E cells (Œ,‚) were defined as Ter119�/PI�. The percentage of total splenocytes that were LM or E (y-axis) is plotted againstperipheral blood percentage of donor Hb (x-axis). Sickle (0% donor Hb) and eGFP (100% donor Hb) controls are shown as F (LM) and ‚ (E). Mice that received transplants areshown as E (LM) and Œ (E).

MIXED CHIMERISM AND CURE OF MURINE SCD 4589BLOOD, 15 DECEMBER 2003 � VOLUME 102, NUMBER 13

For personal use only. by guest on June 3, 2013. bloodjournal.hematologylibrary.orgFrom

Tab

le1.

Pat

ho

log

ican

alys

iso

fch

imer

icm

ice

Mo

use

IDD

on

or

Hb

,%

Car

dia

cva

scu

lar

ecta

sia

Pu

lmo

nar

yar

tery

med

ial

thic

knes

s,�

Pu

lmo

nar

yva

scu

lar

ecta

sia

Rem

ote

hep

atic

infa

rct

Rec

ent

hep

atic

infa

rct

Hep

ato

cyte

iro

n

Hep

atic

Ku

pff

erce

llir

on

Glo

mer

ula

rh

yper

tro

ph

y

Ren

alm

esan

gia

lh

yper

-ce

llula

rity

Ren

altu

bu

lar

iro

n

Sp

len

icar

chit

ectu

ral

dis

rup

tio

n

On

go

ing

mu

ltio

rgan

inju

ryS

ple

ence

llula

rity

eGF

P1

100

00.

30

00

00

00

11

00

eGF

P2

100

00.

30

00

00

00

10

01

eGF

P3

100

00.

10

00

00

00

10

00

eGF

P4

100

00.

10

00

00

00

10

00

Sic

kle1

04

1.1

43

42

32

33

44

4

Sic

kle2

04

2.7

44

42

42

34

44

4

Sic

kle3

03

44

32

13

33

44

24

Sic

kle4

04

2.7

43

32

42

34

44

4

6210

02

1.6

10

00

02

32

02

0

6310

00

0.8

00

00

02

12

00

3

6510

00

2.7

30

00

14

22

00

0

6610

01

2.1

20

21

12

32

13

2

6876

32.

40

11

12

33

22

23

7090

32.

73

00

13

23

21

04

7285

01.

30

42

01

13

11

34

1550

434

2.7

21

42

43

44

43

4

1582

174

42

44

24

44

41

34

1604

613

1.3

03

42

32

24

42

4

1630

792

1.3

03

31

20

14

22

4

1636

57.5

33.

20

32

23

23

44

34

1661

882

2.7

30

21

21

13

13

4

1664

950

2.1

00

11

23

33

10

4

1710

890

NA

NA

01

22

11

23

14

1718

881

2.7

40

11

22

23

10

4

1725

853

1.9

30

21

21

23

13

4

1748

920

2.7

00

01

22

13

10

4

1749

842

2.7

20

21

33

14

13

3

1780

910

1.6

02

32

21

13

22

3

1790

870

1.3

04

10

13

42

10

3

1798

883

1.9

04

11

21

23

12

3

Mic

ew

ere

imm

ersi

onfix

edaf

ter

deat

h,se

ctio

ned,

stai

ned,

and

anal

yzed

ina

blin

ded

man

ner

bya

sing

lepa

thol

ogis

t.A

path

olog

icsc

ore

of0

(no

sick

lepa

thol

ogy)

to4

(cla

ssic

sick

lepa

thol

ogy)

was

assi

gned

toth

ein

dica

ted

char

acte

ristic

s.M

ouse

ID�

inte

rnal

iden

tifier

.eG

FP

1–4

refe

rto

cont

rolm

ice;

Sic

kle

1–4,

untr

ansp

lant

edsi

ckle

mic

e;an

d62

–179

8,tr

ansp

lant

edsi

ckle

mic

e.N

Ain

dica

tes

nota

sses

sed.

4590 KEAN et al BLOOD, 15 DECEMBER 2003 � VOLUME 102, NUMBER 13

For personal use only. by guest on June 3, 2013. bloodjournal.hematologylibrary.orgFrom

remained (Figure 7; Table 1). In those animals with 100% RBCchimerism, both spleen size and hematopoiesis normalized, and itis expected that these mice reverted to the use of the bone marrowas the predominant hematopoietic organ. These data thus distin-guish hematopoiesis from other sickle abnormalities and support athreshold of 100% peripheral blood RBC replacement for completehematopoietic correction.

Discussion

The growing interest in nonmyeloablative BMT for hematologicdisease is based on the assumption that partial chimerism may besufficient to produce a clinical cure. Indeed, there are clinical datasupporting this hypothesis. Chronic transfusion leads to fewercomplications, although patients are not disease free.3 Further, thefew patients with SCD that serendipitously developed mixedchimerism after conventional BMT experienced fewer complica-

tions in short-term follow-up.2 However, important clinical andmechanistic questions remain. Although hematologic indices mayimprove with increasing amounts of healthy peripheral RBCs, theeffect of a persistent state of mixed RBC chimerism on total-bodySCD pathophysiology had not been previously determined. Indeed,for SCD, with its many downstream inflammatory and thromboticcomplications,21,22,24-26 the persistence of significant amounts ofabnormal RBCs may have deleterious clinicopathologic effects.With ever-improving technologies for producing transplantationtolerance and mixed chimerism,5,6,27-30 it may be possible one dayto titrate pretransplantation conditioning to produce a desiredamount of chimerism while minimizing conditioning-related mor-bidity. Thus, it is crucial to determine the effects of mixedchimerism on hematologic and physiologic SCD abnormalities, todevelop meaningful targets for transplantation.

We and others2,6,9 have consistently seen an enrichment of RBCover WBC chimerism when transplanting patients or murinemodels of SCD. This study provided the first evidence suggesting

Figure 7. Analysis of pathology revealed correctionof characteristic sickle pathology in mice that re-ceived transplants. The 4 columns (from left to right)show microscopic findings in the sickle controls (panelsA,E,I,M,Q), a representative mouse that received a trans-plant with less than 50% RBC chimerism (panels B,F,N,R[mouse no. 1582], J [mouse no. 1550]), a representativemouse that received a transplant with more than 70% to80% RBC chimerism (panels C,G,K,O,S [mouse no. 70])and a healthy eGFP control (panels D,H,L,P,T). The 5rows (top to bottom) show sections of cardiac medium-sized arteries (panels A-D; hematoxylin and eosin [H&E]original magnification � 100), sections of a wall of apulmonary artery (panels E-H: H&E stain; original magni-fication � 400), representative hepatic section (panelsI-L: H&E stain; original magnification � 400) with ironstain inset (original magnification, � 400), representativerenal section (panels M-P: trichrome stain; original magni-fication � 100) with iron stain inset (original magnifica-tion, � 400), and representative spleen section (panelsQ-T: H&E stain; original magnification � 100). For thesickle controls and mice with less than 50% RBC chimer-ism, the figure illustrates ectasia of cardiac medium-sizedarteries; increased thickness of the media of the pulmo-nary artery; hepatic infarcts; severe iron deposition inhepatocytes, hepatic Kupffer cells, and hepatic histiocyticcells; cortical renal infarcts with chronic inflammatoryinfiltrates; severe deposition of iron in renal tubularepithelium; and loss of splenic architecture with in-creased hematopoietic cells, ectasia of medium-sizedarteries, and sinusoidal congestion. Mice that receivedtransplants with more than 70% to 80% RBC chimerismshowed changes intermediate between the healthy eGFPcontrol and sickle control in the cardiac vascular ectasia,pulmonary artery wall thickness, and hepatic and renaltubular iron deposition. They showed no recent hepatic orrenal infarcts, they demonstrated fibrosis consistent withremote infarcts, and they showed some return of thenormal nodular architecture of the splenic white pulp.

MIXED CHIMERISM AND CURE OF MURINE SCD 4591BLOOD, 15 DECEMBER 2003 � VOLUME 102, NUMBER 13

For personal use only. by guest on June 3, 2013. bloodjournal.hematologylibrary.orgFrom

that this enrichment occurs entirely on the basis of the survivaladvantage of donor RBCs over the rapidly cleared sickle RBCs inthe peripheral blood and not because of enhanced engraftment orsurvival of healthy erythroid progenitors (Figures 3-5). Thisperipheral enrichment of RBC chimerism compared with stem cellchimerism creates a scenario in which donor RBC and WBCprogenitors can exist as a small percentage of total hematopoieticprogenitors despite complete or near-complete replacement of theperipheral blood with donor RBCs. The implications are 2-fold: (1)Gentle transplantation regimens could produce significant levels ofperipheral RBC chimerism, especially when combined with atolerance-induction protocol. In this study, donors and recipientswere MHC matched but, because of strain background differences,were expected to have multiple minor histocompatibility mis-matches.10,11 We, therefore, used a transplantation regimen thatincluded agents known to induce tolerance through blockade ofT-cell costimulation pathways.5,6,30 Even in the absence of pretrans-plantation chemotherapy, mice that received transplants with thisregimen developed stable WBC chimerism and significant RBCchimerism and, with the exception of their hematopoietic organs,significant improvement in SCD pathophysiology. This improve-ment may be very important for adults with SCD, who areexquisitely sensitive to transplantation preconditioning,1 and forwhom conventional transplantation techniques are currently thoughtto be too risky. (2) The lack of enrichment of donor RBCprogenitors in the hematopoietic organs also implies that at lowlevels of stem cell engraftment, transplantation burnout maybecome a problem clinically. Thus, significant peripheral RBCchimerism may develop in the setting of low levels of donor stemcells, and these transplantations, over the lifetime of a youngpatient, may not be stable. Our studies were carried out for 6 to 10months (one quarter to one half the average murine life span) andindicate that WBC chimerism levels between 0.1% and 82% maybe stable in the long term, but the longevity of low levels of stemcell chimerism (even in the face of high peripheral RBC chimer-ism) in patients cannot be predicted. The addition of robusttolerance-induction protocols to nonmyeloablative transplantationsmay be crucial to produce stable, mixed chimerism after BMT.

Although these protocols are now well established in murinemodels,5,27-30 success has not yet been achieved in producingtransplantation tolerance in either patients or their closest preclini-cal model, the nonhuman primates. The establishment of tolerancein these immunologically complex, out-bred populations representsthe current vanguard of nonmyeloablative BMT research.

One of the critical issues in developing nonmyeloablative BMTfor SCD is determining the targets for RBC chimerism. Althoughthe data from patients with SCD who receive transplants is verylimited, there is a wealth of data from patients undergoing chronictransfusion. In this cohort, less than 30% sickle RBCs in theperipheral blood leads to improvement in acute hematologic andend-organ complications.3 However, the efficacy of transfusion atimproving long-term disease status remains unproven. Our cohortof engrafted mice allows the first direct examination of 3 areas ofsickle pathology relevant to the determination of transplantationbenchmarks: hematologic abnormalities, end-organ abnormalities,and hematopoietic abnormalities. We find that, although progres-sive improvement occurs in many hematologic and end-organabnormalities in mice with more than 70% to 80% normal levels ofHb (Figures 6-8; Table 1), correction of bone marrow and splenichematopoietic abnormalities does not occur despite high levels(� 90%) of healthy RBCs in the peripheral blood (Figures 4 and 6;Table 1). Thus, because the enrichment of RBC versus WBCchimerism occurred exclusively as a result of the peripheralsurvival advantage of healthy versus sickle RBCs, the hematopoi-etic organs in the mice that received transplants were stillpredominantly sickle, even in the face of significant (as high as90%) peripheral RBC chimerism. Thus, sickle splenic and bonemarrow hyperactive erythropoiesis7,8 continued to exist in theseanimals, despite high peripheral blood RBC chimerism. The highoutput of sickle RBCs into the peripheral blood was cleared withthe same rapid kinetics observed in sickle mice that did not receivetranplants,7,8 leading to a final mixture of donor and sickle RBCsthat greatly overestimated the amount of stem cell chimerismpresent in the hematopoietic organs of these animals. Two strikingconclusions are drawn from this observation: (1) Sickle-mediatedsolid-organ pathology is determined by the peripheral bloodchimerism. This determination is shown in Table 1 and Figures 6and 7 and indicates that, for much of the organ pathology thatplagues patients with SCD, significant improvement can be ex-pected as progressive increases in peripheral blood RBC chimerismoccur. (2) Sickle-mediated hematopoietic organ pathology reflectsstem cell chimerism rather than peripheral blood RBC chimerism.Thus, given low levels of stem cell chimerism, most of thehematopoietic precursors in the chimeric mice are still recipient(sickle) rather than donor, and “cure” of these organs does notoccur until stem cell chimerism reaches significant levels (� ap-proximately 50% in this study, as shown in Figure 2C). This hasimportant implications for nonmyeloablative transplantation in thatit suggests that partial peripheral blood RBC chimerism, even if itis significant (� 70%), will likely arise from a highly abnormalhematopoietic compartment. Indeed, on the basis of our cohort ofchimeric animals, a target of 50% or more stem cell chimerism(leading to 100% replacement of sickle with healthy RBCs in theperipheral blood) emerges as a benchmark at which all sickle-mediated abnormalities, including hematopoiesis, can be corrected.One important caveat to this target of more than 50% stem cellchimerism must be considered. This panel of mice did not includeany with stem cell chimerism intermediary between 18% (whichdid not produce full RBC replacement) and 50% (which did

Figure 8. Total pathology score showed progressive correction of sicklepathology. Total pathologic score (y-axis) versus percentage of normal Hb levels inthe peripheral blood (x-axis). The total pathology score includes the followingindividual parameters: (1) cardiac vascular ectasia, (2) pulmonary artery medialthickness, (3) pulmonary vascular ectasia, (4) remote hepatic infarct, (5) recenthepatic infarct, (6) hepatocyte iron deposition, (7) hepatic Kupffer cell iron deposition,(8) renal glomerular hypertrophy, (9) renal mesangial hypercellularity, (10) renaltubular iron deposition, (11) splenic loss of architectural integrity and vascularcongestion, and (12) evidence of ongoing multiorgan injury. This evidence includesprimarily small-vessel changes (ectasia, perivascular fibrosis, and congestion),parenchymal chronic ischemic changes, pericentral vein sclerosis and hepatocyticischemic changes in the liver, and evidence of ongoing hemolysis (especially in theliver and kidneys). Each of these parameters received a pathology score from 0 to 4(Table 1). The summed scores were plotted against the percentage of donor Hb.

4592 KEAN et al BLOOD, 15 DECEMBER 2003 � VOLUME 102, NUMBER 13

For personal use only. by guest on June 3, 2013. bloodjournal.hematologylibrary.orgFrom

produce full RBC replacement). There may be a level of WBCchimerism between these 2 limits that also produces full RBCreplacement and cure of all SCD pathophysiology.

These results represent the first preclinical trial of nonmyeloab-lative transplantation for SCD designed to produce RBC as well asWBC mixed chimerism. They allowed us to investigate themechanism of donor RBC enrichment as well as to determine that,for a complete reversal of all categories of sickle pathology,complete replacement of the peripheral blood with donor RBCs isrequired. WBC chimerism is, therefore, a reasonable goal for

nonmyeloablative BMT for SCD, but anything less than 100%replacement of the diseased RBC compartment leaves importantsickle pathology uncorrected.

Acknowledgments

We thank Diane Hollenbaugh, Robert Peach, and Alejandro Aruffo(Bristol-Myers Squibb) for providing CTLA4-Ig. We further thankRobert Karaffa for assistance with the Vantage flow cytometer.

References

1. Walters MC, Patience M, Leisenring W, et al. Bar-riers to bone marrow transplantation for sickle cellanemia. Biol Blood Marrow Transplant. 1996;2:100-104.

2. Walters MC, Patience M, Leisenring W, et al.Stable mixed hematopoietic chimerism after bonemarrow transplantation for sickle cell anemia. BiolBlood Marrow Transplant. 2001;7:665-673.

3. Eckman JR. Techniques for blood administrationin sickle cell patients. Semin Hematol. 2001;38:23-29.

4. Walters MC, Storb R, Patience M, et al. Impact ofbone marrow transplantation for symptomaticsickle cell disease: an interim report. Multicenterinvestigation of bone marrow transplantation forsickle cell disease. Blood. 2000;95:1918-1924.

5. Adams AB, Durham MM, Kean L, et al. Costim-ulation blockade, busulfan, and bone marrowpromote titratable macrochimerism, induce trans-plantation tolerance, and correct genetic hemo-globinopathies with minimal myelosuppression.J Immunol. 2001;167:1103-1111.

6. Kean LS, Durham MM, Adams AB, et al. A curefor murine sickle cell disease through stablemixed chimerism and tolerance induction afternonmyeloablative conditioning and major histo-compatibility complex-mismatched bone marrowtransplantation. Blood. 2002;99:1840-1849.

7. Kean LS, Brown LE, Nichols JW, et al. Compari-son of mechanisms of anemia in mice with sicklecell disease and beta-thalassemia: peripheraldestruction, ineffective erythropoiesis, and phos-pholipid scramblase-mediated phosphatidylserineexposure. Exp Hematol. 2002;30:394-402.

8. de Jong K, Emerson RK, Butler J, Bastacky J,Mohandas N, Kuypers FA. Short survival of phos-phatidylserine-exposing red blood cells in murinesickle cell anemia. Blood. 2001;98:1577-1584.

9. Iannone R, Luznik L, Engstrom LW, et al. Effectsof mixed hematopoietic chimerism in a mousemodel of bone marrow transplantation for sicklecell anemia. Blood. 2001;97:3960-3965.

10. Paszty C, Brion CM, Manci E, et al. Transgenicknockout mice with exclusively human sickle he-moglobin and sickle cell disease. Science. 1997;278:876-878.

11. Okabe M, Ikawa M, Kominami K, Nakanishi T,Nishimune Y. ‘Green mice’ as a source of ubiqui-tous green cells. FEBS Lett. 1997;407:313-319.

12. Zhao Y, Lin Y, Zhan Y, et al. Murine hematopoieticstem cell characterization and its regulation in BMtransplantation. Blood. 2000;96:3016-3022.

13. Socolovsky M, Nam H, Fleming MD, Haase VH,Brugnara C, Lodish HF. Ineffective erythropoiesisin Stat5a(-/-)5b(-/-) mice due to decreased sur-vival of early erythroblasts. Blood. 2001;98:3261-3273.

14. Kina T, Ikuta K, Takayama E, et al. The monoclo-nal antibody TER-119 recognizes a molecule as-sociated with glycophorin A and specificallymarks the late stages of murine erythroid lineage.Br J Haematol. 2000;109:280-287.

15. Christian JA, Rebar AH, Boon GD, Low PS. Meth-odologic considerations for the use of canine invivo aged biotinylated erythrocytes to study RBCsenescence. Exp Hematol. 1996;24:82-88.

16. Tait JF, Gibson D. Measurement of membranephospholipid asymmetry in normal and sickle-cellerythrocytes by means of annexin V binding.J Lab Clin Med. 1994;123:741-748.

17. Wood BL, Gibson DF, Tait JF. Increased erythro-cyte phosphatidylserine exposure in sickle celldisease: flow-cytometric measurement and clini-cal associations. Blood. 1996;88:1873-1880.

18. de Jong K, Larkin SK, Styles LA, Bookchin RM,Kuypers FA. Characterization of the phosphati-dylserine-exposing subpopulation of sickle cells.Blood. 2001;98:860-867.

19. Setty BN, Kulkarni S, Stuart MJ. Role of erythro-cyte phosphatidylserine in sickle red cell-endo-thelial adhesion. Blood. 2002;99:1564-1571.

20. Setty BN, Rao AK, Stuart MJ. Thrombophilia insickle cell disease: the red cell connection. Blood.2001;98:3228-3233.

21. Setty BN, Stuart MJ. Vascular cell adhesion mole-cule-1 is involved in mediating hypoxia-inducedsickle red blood cell adherence to endothelium:potential role in sickle cell disease. Blood. 1996;88:2311-2320.

22. Duits AJ, Pieters RC, Saleh AW, et al. Enhancedlevels of soluble VCAM-1 in sickle cell patientsand their specific increment during vasoocclusivecrisis. Clin Immunol Immunopathol. 1996;81:96-98.

23. Hebbel RP. Blockade of adhesion of sickle cellsto endothelium by monoclonal antibodies. N EnglJ Med. 2000;342:1910-1912.

24. Makis AC, Hatzimichael EC, Bourantas KL. Therole of cytokines in sickle cell disease. Ann He-matol. 2000;79:407-413.

25. Platt OS. Sickle cell anemia as an inflammatorydisease. J Clin Invest. 2000;106:337-338.

26. Platt OS. The acute chest syndrome of sickle celldisease. N Engl J Med. 2000;342:1904-1907.

27. Wekerle T, Kurtz J, Ito H, et al. Allogeneic bonemarrow transplantation with co-stimulatory block-ade induces macrochimerism and tolerance with-out cytoreductive host treatment. Nat Med. 2000;6:464-469.

28. Sykes M, Szot GL, Swenson KA, Pearson DA.Induction of high levels of allogeneic hematopoi-etic reconstitution and donor-specific tolerancewithout myelosuppressive conditioning. Nat Med.1997;3:783-787.

29. Wekerle T, Sayegh MH, Hill J, et al. Extrathymic Tcell deletion and allogeneic stem cell engraftmentinduced with costimulatory blockade is followedby central T cell tolerance. J Exp Med. 1998;187:2037-2044.

30. Durham MM, Bingaman AW, Adams AB, et al.Cutting edge: administration of anti-CD40 ligandand donor bone marrow leads to hemopoietic chi-merism and donor-specific tolerance without cy-toreductive conditioning. J Immunol. 2000;165:1-4.

MIXED CHIMERISM AND CURE OF MURINE SCD 4593BLOOD, 15 DECEMBER 2003 � VOLUME 102, NUMBER 13

For personal use only. by guest on June 3, 2013. bloodjournal.hematologylibrary.orgFrom


Recommended