Banfi 2000 Experimental-Hematology

  • Upload
    jivkos

  • View
    216

  • Download
    0

Embed Size (px)

Citation preview

  • 8/9/2019 Banfi 2000 Experimental-Hematology

    1/9

    Experimental Hematology 28 (2000) 707715

    0301-472X/00 $see front matter. Copyright 2000 International Society for Experimental Hematology. Published by Elsevier Science Inc.

    PII S0301-472X(00)00160-0

    Proliferation kinetics and differentiationpotential of ex vivo expanded human bone marrow

    stromal cells: Implications for their use in cell therapy

    Andrea Banfia,b,c, Anita Muragliaa, Beatrice Dozina,b,Maddalena Mastrogiacomoa,c, Ranieri Canceddaa,b,c, and Rodolfo Quartoa,b

    aCentro di Biotecnologie Avanzate, Genova, Italy;bIstituto Nazionale per la Ricerca sul Cancro, Genova, Italy; cDipartimento di Oncologia, Biologiae Genetica, Universita di Genova, Italy

    (Received 4 January 2000; revised 12 February 2000; accepted 22 February 2000)

    Objective. Bone marrow stromal cells (BMSC) are an attractive target for novel strategies in

    the gene/cell therapy of hematologic and skeletal pathologies, involving BMSC in vitro expan-

    sion/transfection and reinfusion. We investigated the effects of in vitro expansion on BMSCpluripotentiality, proliferative ability, and bone-forming efficiency in vivo.

    Materials and Methods. BMSC from three marrow donors were cultured to determine their

    growth kinetics. At each passage, their differentiation potential was verified by culture in in-

    ductive media and staining with alizarin red, alcian blue, or Sudan black, and by immu-

    nostaining for osteocalcin or collagen II. First passage cells were compared to fresh marrow

    for their bone-forming efficiency in vivo. Stromal cell clones were isolated from five donors

    and characterized for their multidifferentiation ability. The lifespan and differentiation kinet-

    ics of five of these clones were determined.

    Results. After the first passage, BMSC had a markedly diminish proliferation rate and gradu-

    ally lost their multiple differentiation potential. Their bone-forming efficiency in vivo was re-

    duced by about 36 times at first confluence as compared to fresh bone marrow. Experiments

    on the clones yielded comparable results.

    Conclusions. Culture expansion causes BMSC to gradually lose their early progenitor proper-ties. Both the duration and the conditions of culture could be crucial to successful clinical use

    of these cells and must be considered when designing novel therapeutic strategies involving

    stromal mesenchymal progenitor manipulation and reinfusion. 2000 International Society

    for Experimental Hematology. Published by Elsevier Science Inc.

    Keywords: ChondrogenesisOsteogenesisAdipogenesisBone marrow stromal cellsCell

    therapy

    Introduction

    The bone marrow stromal system is defined as the connec-

    tive tissue elements providing structural and functional sup-

    port for hemopoiesis [1,2] and includes various cell types:bone marrow fibroblastsreticular cells, adipocytes, osteo-

    blasts, macrophages, and endothelial cells. It has long been

    recognized that the fibroblastoid component of the stromal

    system contains a rich reserve of osteogenic progenitors that

    also are capable of transferring the hemopoietic microenvi-

    ronment [35] both in vitro [6] and in vivo [7,8].

    A mesenchymal stem cell (MSC) is defined as a pluripo-

    tent cell capable of replicating extensively and self-main-taining and whose progeny eventually gives rise to skeletal

    tissues: cartilage, bone, tendon, ligament, and marrow

    stroma [5]. By definition, such cells exist during embryo-

    genesis, and it is the object of current research to determine

    whether these cells persist during adult life.

    Bone marrow stromal cells (BMSC) can be cultivated in

    vitro and contain progenitors capable of pluridifferentiating

    into the mesenchymal lineages of bone, cartilage, fat, mus-

    cle, and other connective tissues [3,5,911]; therefore, they

    are an interesting target for use in cell and gene therapy.

    Offprint requests to: Rodolfo Quarto, M.D., Laboratorio di Differenzia-

    mento Cellulare, Centro di Biotecnologie Avanzate/Istituto Nazionale per

    la Ricerca sul Cancro, L.go R. Benzi n. 10, 16132 Genova, Italy; E-mail:

    [email protected]

  • 8/9/2019 Banfi 2000 Experimental-Hematology

    2/9

    708 A. Banfi et al. /Experimental Hematology 28 (2000) 707715

    BMSC attractiveness is based on the ease with which they

    can be isolated from the patients marrow, expanded many-

    fold in vitro, manipulated, and reinfused to the same patient.

    In this setting, BMSC can have many hematologically rele-

    vant applications. They are being investigated as vehicles

    for gene therapy of hemophilia B [12]. Several authors have

    shown that the bone marrow microenvironment is heavily

    and permanently damaged by bone marrow transplantation

    (BMT) conditioning regimens [1321]. A preliminary re-

    port has claimed that autologous ex vivo expanded BMSC

    accelerated hemopoietic recovery after high-dose chemo-

    therapy with peripheral blood progenitor cells (PBPC) res-

    cue in breast cancer patients [22]. BMSC can be used in

    BMT as a therapy for skeletal pathologies [23,24] and has

    been applied to cases of osteogenesis imperfecta [25,26].

    The use of BMSC in such therapeutic strategies relies on

    the ability of BMSC to proliferate readily and give rise to a

    differentiated progeny that can substitute for the diseased

    counterpart. It is of interest to determine whether, after the

    ex vivo expansion necessary before their therapeutic manip-ulation and reinfusion, these cells possess stem features. In

    this study, we investigated the stem cell characteristics of ex

    vivo expanded human BMSC by asking the following ques-

    tions: (1) Do BMSC maintain a constant doubling time dur-

    ing their in vitro culture? (2) Do BMSC maintain their own

    multilineage differentiation potential after a number of mi-

    totic divisions? (3) Do BMSC maintain a constant effi-

    ciency in their bone-forming ability in vivo? (4) Do clones

    derived from individual colony-forming unit-fibroblasts

    (CFU-F) display a multilineage differentiation potential and

    do they maintain it throughout their lifespan?

    To this purpose, we investigated the in vitro growth ki-netics of BMSC from three different donors in the primary

    culture and up to the fourth passage. We analyzed their dif-

    ferentiation potential at each passage to monitor the mainte-

    nance of pluripotentiality during mitotic expansion. To fur-

    ther test this point, we compared the in vivo bone-forming

    efficiency of first passage BMSC to that of fresh bone mar-

    row. In both experiments we found that, as a population, ex-

    panded BMSC progressively lost their differentiation poten-

    tial.

    The nearest approximation to the putative MSC that can

    be studied is the CFU-F. We isolated, expanded, and char-

    acterized 80 nonimmortalized clones originating from sin-

    gle CFU-F of human bone marrow. Their osteo-chondro-adipogenic potential was assessed by in vitro assays. All

    BMSC clones analyzed were able to differentiate in vitro

    into the osteogenic lineage and about one-third displayed

    osteo-chondro-adipogenic potential. The lifespan was deter-

    mined and the differentiation potential was assessed at in-

    creasing cell doublings in a number of clones. The results

    paralleled those of BMSC cultures.

    This study provides evidence that, with extended culture

    in the conditions currently in use, human BMSC display a

    tendency to lose their multipotentiality, proliferation poten-

    tial, and in vivo bone-forming efficiency. The question

    whether the graduality of this process could make a limited

    expansion compatible with successful clinical use awaits in

    vivo studies. Nevertheless, the effect of in vitro expansion

    on the stem cell features of BMSC cultures need to be con-

    sidered when designing strategies for use of BMSC in gene

    and cell therapy.

    Materials and methods

    Cell culture and BMSC growth kinetics determination

    Stromal cells were obtained from iliac crest marrow aspirates from

    healthy donors for BMT procedures carried out in S. Martino Hos-

    pital, Genova, Italy, and G. Gaslini Pediatric Hospital, Genova, It-

    aly. Donor ages ranged between 5 months and 30 years; all donors

    were white Caucasians. Informed consent was obtained from all

    donors, and all procedures were approved by the institutional ethi-

    cal committee. BMSC cultures were performed essentially as pre-

    viously described [13], except that they were always carried out

    with medium supplemented with 1 ng/ML human recombinant fi-broblast growth factor 2 (FGF-2) (Austral Biologicals, San Ra-

    mon, CA). After 2 weeks, CFU-F number was determined as de-

    scribed [13]. Confluent cultures were detached with 0.05%

    trypsin-0.01% EDTA (Sigma, St. Louis, MO). Cells were replated

    for the in vitro differentiation experiments and for determination

    of BMSC growth kinetics. At the first passage, BMSC were re-

    plated at a concentration of 5 105 cells in one 10-cm Petri dish.

    When subconfluent, this plate was successively split 1:2 unit eight

    dishes were obtained. At this point, cells again were detached,

    pooled, counted, and analyzed for differentiation potential. The

    same procedure was repeated for five passages, and the cumulative

    cell doublings of the populations were plotted against time in cul-

    ture to determine the growth kinetics of BMSC expansion.

    Cell cloning

    Marrow samples were used to obtain clones by limiting dilution.

    Briefly, 2 105 nucleated marrow cells were plated in each of four

    to five 96-well plates for each marrow sample. Cultures were per-

    formed in standard medium in the presence of FGF-2 and exam-

    ined daily for the appearance of stromal colonies. Wells containing

    more than one colony were not considered. All clones that reached

    confluence after passaging into one well of a 24-well plate were

    trypsinized and each of them replated in four wells (replicas). Of

    the four replicas of each clone, three were stimulated for the differ-

    entiation experiments when cells reached confluence and one was

    frozen to be kept as stock.

    In vitro differentiation of BMSC cultures and clones

    BMSC potential to differentiate into the chondrogenic, osteogenic

    and adipogenic lineages was verified at the level of primary cul-

    tures and at each passage for four passages. Clones were analyzed

    at the stage of replicas and at the following passages during their

    lifespan. In all cases, cells were enzymatically detached from cul-

    ture dishes on reaching 90% confluence and plated at 5 104

    cells/cm2 in 24-well culture plates. Some dishes were not replated

    in the wells, but instead were stimulated under the same conditions

    and then used for RNA extraction and reverse transcriptase-poly-

    merase chain reaction (RT PCR) analysis. Each well or dish was

    kept in complete medium until confluence was reached. Cultures

  • 8/9/2019 Banfi 2000 Experimental-Hematology

    3/9

    A. Banfi et al. /Experimental Hematology 28 (2000) 707715 709

    were stimulated with the appropriate differentiating medium ac-

    cording to the following conditions:

    Chondrogenic differentiation: cultures were stimulated for 1

    week in standard medium (F-12 with 10% fetal calf serum

    [FCS] and without FGF-2) supplemented with 50 g/mL

    ascorbic acid and 1 ng/mL human recombinant transforming

    growth factor 1 (TGF-1; Austral Biologicals, San Ramon,

    CA). Type II collagen expression was revealed by immu-nostaining with the monoclonal antibody CIICI (Develop-

    mental Studies Hybridoma Bank) and the deposition of pro-

    teoglycans was revealed by alcian blue staining.

    Osteogenic differentiation: cultures were stimulated for 2

    weeks in standard medium supplemented with 50 g/mL

    ascorbic acid, 1.5 mg/mL -glycerophosphate, and 108M

    dexamethasone. Osteocalcin expression was revealed by im-

    munostaining with an antiserum against bovine osteocalcin

    kindly provided by Dr. Simon Robbins, and the presence of

    calcium deposits was revealed by von Kossa and alizarin S

    staining.

    Adipogenic differentiation: cultures were stimulated for 3

    weeks in F12 medium supplemented with 1% FCS, 107

    Mdexamethasone, and 6 ng/mL insulin. Lipid droplets were re-

    vealed by staining with Sudan black IV.

    As controls, the following culture conditions were used: (a)

    proliferating unstimulated cells expanded with FGF-2 up to con-

    fluence (basal phenotype); and (b) confluent cultures kept in stan-

    dard medium for the same period of time as the corresponding

    differentiation protocols, but without the stimulating factors (sponta-

    neous over-time phenotype).

    The cultures were scored as positive, borderline, or negative.

    For adipogenesis, scores were defined as follows: negative no

    more than a single adipocyte in from a total of three random mi-

    croscopic fields; borderline one or two adipocytes in each of at

    least two of three random microscopic fields; positive at leastthree adipocytes in each of at least two of three random micro-

    scopic fields. For osteogenesis and chondrogenesis, we quantified

    alizarin S and alcian blue staining, respectively. Images of the

    stained wells were acquired with a Cohu black-and-white camera

    and analyzed with the NIH-Image 1.60 public domain software.

    We scored the average of duplicate wells for each condition. Back-

    ground was calculated from empty wells and subtracted from all

    experimental readings. Differentiation units were calculated as the

    fold increase in staining induced by the differentiation schedule as

    compared with the negative control. Cultures were scored as fol-

    lows: negative less than 1.2 differentiation units; borderline

    between 1.2 and 2 differentiation units; positive more than 2 dif-

    ferentiation units.

    In vivo bone-forming efficiency

    In vivo ectopic bone formation was assayed according to a proto-

    col previously optimized in our laboratory [27]. Briefly, on reach-

    ing their first confluence, 2.5 105 BMSC were loaded on a

    highly porous hydroxyapatite support and implanted subcutane-

    ously on the back of nude CD-1 nu/nu mice purchased form

    Charles River Italia (Calco, Italy).

    When fresh bone marrow was used, nucleated cells were iso-

    lated by Ficoll gradient centrifugation and counted, and each cube

    was loaded with the appropriate number of cells. Preliminary ex-

    periments were carried out to determine the number of fresh mar-

    row nucleated cells, of which the majority are hemopoietic progen-

    itors, that produce a bone formation equivalent to the standard

    2.5 105 expanded BMSC.

    The fresh marrow content of CFU-F was determined as de-

    scribed earlier. For BMSC, and additional colony-forming effi-

    ciency experiment was performed on reaching the first confluence,

    in parallel with the in vivo implants, by plating 103 BMSC per 10-

    cm Petri dish. After 2 weeks of culture, the plates were fixed andstained, and the colonies were counted as for the fresh marrow.

    Animals were sacrificed 8 weeks after implantation, and the grafts

    removed and processed for histologic analysis as previously de-

    scribed [27].

    Clone lifespan

    Five clones were selected after characterization at the stage of rep-

    licas for determination of their lifespan and maintenance of differ-

    entiation ability: three were tripontential (OCA [osteo-chondro-

    adipogenic]) and two were bipotential (OC [osteo-chondrogenic])

    at this stage. The frozen replica was thawed and plated in a 6-cm

    Petri dish. When it reached confluence, cells were trypsinized,

    counted for determination of cell doublings, and split into four

    equal parts: three were plated each in a well of a 24-well plate andstimulated for differentiation to the three lineages as described ear-

    lier, and the fourth was replated in a 6-cm Petri dish to continue

    expansion. For these experiments, osteogenesis and chondrogene-

    sis were assayed as osteocalcin and collagen II expression as deter-

    mined by immunohistochemistry. The results could not be quanti-

    fied and were only scored as positive or negative as compared to

    the negative control.

    Semiquantitative RT-PCR measurements

    Total RNA was extracted from BMSC and control tissues (human

    articular cartilage, bone, and adipose tissue) with the guanidinium

    isothiocyanate procedure by Chomczynski and Sacchi [28]. Semi-

    quantitative RT-PCR was performed using the GeneAmp RNAPCR kit from Perkin Elmer. For each RNA sample, a master RT

    reaction was performed with 2 g total RNA in a 40 L mixture

    containing 1 PCR buffer from the provider, 5 mM MgCl2, 1 mM

    each of dCTP, dATP, dGTP, and dTTP, 1 U/

    L of RNAse inhibi-

    tor, 250 pmol of random examer primers, and 2.5 U/L of MuLV

    RT. Reaction time was 1 hour at 42C. Each master cDNA product

    was divided into two equal parts that were used for PCR amplifica-

    tion either of the housekeeping gene glyceraldehyde phosphate de-

    hydrogenase (GAPDH) or of one of the following specific genes:

    1(II) collagen, osteocalcin, and lipoprotein lipase (LPL). The

    PCR reactions were performed in a 100-L mixture that contained

    1 PCR buffer, 2 mM MgCl2, 1 mM each of dCTP, dATP, dGTP,

    and dTTP, 30 pmol of 5 and 3 specific primers, and 2.5 U of Am-

    pliTaq DNA polymerase. Primer sequences for all the aforemen-

    tioned genes were as follows: GAPDH: 5-ACCACAGTCCAT-

    GCCATCAC-3 and 5-TCCACCACCCTGTTGCTGTA-3 [29];

    1(II) collagen: 5-CTGCTCGTCGCCGCTGTCCTT-3 and 5-

    AAGGGTCCCAGGTTCTCCATC-3 [11]; OC: 5-CATGAG-

    AGCCCTCACA-3 and 5-AGAGCGACACCCTAGAC-3 [30];

    LPL: 5-GAGATTTCTCTGTATGGCACC-3 and 5-CTGCAA-

    ATGAGACACTTTCTC-3 [30]. The expected product sizes were

    GAPDH 450 bp, 1(isoform IIB) collagen 225 bp, OC 310 bp, and

    LPL 276 bp. The number of cycles used was in the linear range of

    amplification for the specific gene product, as preliminary experi-

    ments were carried out by the authors originally describing the

  • 8/9/2019 Banfi 2000 Experimental-Hematology

    4/9

    710 A. Banfi et al. /Experimental Hematology 28 (2000) 707715

    primer sets. The reaction profiles used were as follows, with all

    preceded by a first denaturation step at 95

    C for 4 min: GAPDH:

    95C for 1 minute, 55C for 1 minute, and 72C for 1 minute, for

    24 cycles; OC: 95C for 1 minute, 55C for 1 minute, and 72C for

    1 minute, for 30 cycles; LPL: 95C for 1 minute, 60C for 1

    minute, and 72C for 1 minute, for 35 cycles; 1(II) collagen:95C for 3 minutes, 58C for 2 minutes, and 72C for 1.5 minutes,

    for one cycle and then 95C for 1 minute, 58C for 1 minute, and

    72C for 1.5 minute, for 30 cycles. All PCR reactions ended with a

    7-minute incubation at 72C. RT-PCR products were analyzed by

    electrophoresis of 20-L aliquots in 1% agarose gels and visual-

    ized by ethidium bromide, except for the 1(II) fragment obtained

    by the BMSC RNAs that were ethanol precipitated to load the en-

    tirety of the products. The intensity of individual bands was quan-

    tified by image analysis using the public domain software NIH

    1.60. The amount of PCR product for each single gene was nor-

    malized according to the corresponding GADPH PCR product.

    Results

    Growth kinetics and maintenance

    of differentiation potential of primary BMSC cultures

    The growth kinetics of three different BMSC cultures was

    investigated from the primary culture through the fifth pas-

    sage, corresponding to about 24 doublings. Expanded

    BMSC were tested for their ability to undergo osteogenic,

    chondrogenic, and adipogenic differentiation after appropri-

    ate induction until the fourth passage.

    Primary cultures reached the first confluence in about 3

    weeks and 1215 doubling (Fig. 1). On replating, BMSC

    slowed their proliferation rate considerably and by the sec-

    ond passage stabilized their growth rate at about 1014 daysper population doubling (Fig. 1 and Table 1). The average

    doubling time increased from 1.3 days at the primary cul-

    ture stage to 7.7 at the first passage and 14.7 at the third pas-

    sage (Table 1). Concurrent with this slowing of growth rate,

    BMSC showed a change in appearance, from the initial

    spindle shape to a more flattened square morphology (data

    not shown).

    Differentiation in the three lineages was induced as de-

    scribed in the Materials and methods section. Figure 2

    shows the results of a representative experiment with

    BMSC at their first confluence. As revealed by immu-

    nostaining with specific antibodies, OC expression was evi-

    dent after the second week in osteogenic culture medium

    (Fig. 2A), type II collagen was observed earlier, after 1

    week in chondrogenic medium (Fig. 2C), and adipogenic

    differentiation was always the last to be observed. Although

    sporadic adipocytes already were evident after several days

    in adipogenic medium, only after 3 weeks of stimulation did

    cultures display several large adipocytic colonies, as re-

    Figure 1. BMSC culture growth kinetics and differentiation potentialmaintenance. Growth kinetics of three BMSC cultures was determined.

    Primary cultures reached the first confluence in about 3 weeks and 1215

    doublings. On replating, BMSC slowed their proliferation rate consider-

    ably. The curves represent the cumulative doubling number of the three

    individual cultures versus time in culture.

    Table 1.

    Growth kinetics and differentiation of one BMSC primary culture

    Differentiation

    Passage Total doublings Average doubling time (days) Osteogenesis Chondrogenesis Adipogenesis

    0 14.6 1.3

    1 17.2 7.7

    2 18.7 10.0

    3 20.2 14.7

    4 22.2 9.5

    5 24.1 15.8 ND ND ND

    Population doublings and differentiation potential of one BMSC culture were determined at the first confluence (passage 0) and at each passage thereafter.

    Average doubling time at each passage was determined by dividing the number of doublings undergone by the culture since the previous passage by the time

    (in days) taken to reach confluence. Differentiation was assayed by staining with alizarin S (osteogenesis), alcian blue (chondrogenesis), and Sudan black (ad-

    ipogenesis). Scoring was as follows: positive; borderline; negative, as defined in the Materials and methods section. ND not done.

  • 8/9/2019 Banfi 2000 Experimental-Hematology

    5/9

    A. Banfi et al. /Experimental Hematology 28 (2000) 707715 711

    vealed by phase contrast microscopy and Sudan black stain-

    ing (Fig. 2E). von Kossa, alizarin S, and alcian blue staining

    were positive on similar cultures maintained in osteogenic

    and chodrogenic medium, respectively (data not shown),

    Control unstimulated cultures yielded barely detectable lev-

    els of expression of the differentiation markers (Fig. 2B, D,

    and F).Differentiation in the three lineages induced by culture

    with inductive media was verified by semiquantitative RT-

    PCR analysis using primers for type II collagen, OC, and

    LPL (Fig. 3 and Table 2). As shown in Figure 3 (lane 5), un-

    stimulated cultures already show some, although variable,

    basal expression of the specific makers studied:

    1(II)

    (panel B) and LPL (panel C) were clearly detectable,

    whereas the OC level (panel A) was barely above the back-

    ground. With appropriate stimulation (Fig. 3, lane 6), the

    expression of the three markers was significantly induced.

    With respect to the basal level, the OC,

    1(II), LPL levels

    were increased by a factor of 16, 3.8 and 4.8, respectively(Table 2). In long-term unstimulated cultures, the levels of

    1(II) and LPL were similar to or lower than the basal level,

    showing absence of spontaneous differentiation (Table 2

    and Fig. 3 B and C lane 7). In contrast, the level of OC

    expression in long-term unstimulated cultures was about

    three times higher than the basal level (Table 2 and Fig. 3A,

    lane 7).

    For the three markers considered, the expected sizes of

    the PCR products were consistent with those obtained with

    Figure 2. Differentiation of human BMSC. Confluent human BMSC cul-

    tures were maintained in differentiation medium (A, C, E) and in standard

    control medium (B, D, F) for different time intervals. Osteocalcin deposi-

    tion in the extracellular matrix, as revealed by immunostaining, was posi-

    tive after the second week in osteogenic medium (A). After 1 week in

    chondrogenic medium, discrete matrix noduli were positive for type II col-

    lagen (C). Several large adipocytic colonies were observed with Sudan

    black staining after 3 weeks of stimulation (E). Without the stimulating

    factors, cultures kept in the same conditions for the same time period dis-

    played only basal levels of differentiation (B, D, F). Immunostaining for

    osteocalcin (A, B), immunostaining for type II collagen (C, D), and Sudan

    black staining (E, F). Bar 1 mm.

    Figure 3. RT-PCR analysis for expression of osteoblast, chondrocyte, and

    adipocyte-related genes in BMSC, bone, cartilage, and adipose tissue.

    RNA was extracted from unstimulated proliferating BMSC ( lanes 2 and 5),

    from confluent cells stimulated for the individual lineage (lanes 3 and 6),

    and from parallel cultures of confluent unstimulated cells (lanes 4 and 7).

    RNA also was extracted from native tissues as control for PCR products

    (lanes 8 and 9): bone (A), cartilage (B), and adipose tissue (C). Reverse

    transcribed cDNA was amplified by using specific oligonucleotide primers

    (described in the Materials and methods section). PCR products were visu-

    alized in ethidium bromide stained gels. Lanes 2, 3, 4, and 8 correspond to

    GADPH product, lanes 5, 6, 7, and 9 (A) to OC product, lanes 5, 6, 7, and 9

    (B) to 1(II) product, and lanes 5, 6, 7, and 9 (C) to LPL product. A 123-bp

    ladder was used as standard (lane 1: from top to bottom 492, 369, 246,

    123 bp).

    Table 2. Quantification of PCR products in differentiating

    BMSC cultures

    Condition OC 1(II) LPL

    Basal expression 1 1 1

    Induced differentiation 16 3.8 4.8

    Spontaneous differentiation 2.9 0.7 0.6

    RT-PCR reaction products shown in Figure 2 were quantified by image

    analysis. For each set of reactions, product intensity was corrected accord-

    ing to the amount of the corresponding GAPDH PCR product and normal-

    ized to the relative basal level.

  • 8/9/2019 Banfi 2000 Experimental-Hematology

    6/9

    712 A. Banfi et al. /Experimental Hematology 28 (2000) 707715

    RNA extracted from the corresponding native tissues (Fig.

    3AC, lane 9).

    Parallel with the loss of proliferation velocity was a loss

    of differentiation potential (Table 1). By the 19th doubling,

    BMSC greatly reduced their response to adipogenic stimu-

    lation to a borderline level and completely lost it by the

    22nd passage. Chondrogenesis, as revealed by alcian blue

    staining, and osteogenesis, as assessed by alizarin S stain-

    ing, were maintained longer, but dropped to borderline liv-

    els by the fourth passage, corresponding to about 22 dou-

    blings.

    In vivo bone-forming efficiency

    Osteogenic potential of freshly harvested bone marrow

    cells, which include both osteogenic and hemopoietic cells,

    was investigated and compared to that of expanded primary

    BMSC, which reached confluence in about three weeks, un-

    dergoing 17.5 population doublings. Samples of fresh bone

    marrow were adsorbed onto HA ceramic cubes and subcuta-

    neously implanted in nude mice as described in the Materi-als and methods section. We first determined that when 5

    million nucleated cells from fresh bone marrow after sepa-

    ration on Ficoll were implanted, the amount of bone tissue

    formed was roughly comparable to that observed in the ex-

    periments where the standard 250,000 expanded BMSC

    were used. Hemopoietic marrow was detected in both speci-

    mens (data not shown).

    Several independent experiments were performed, and

    the data from one representative experiment are given in

    Table 3. It should be noted that over the 3-week culture pe-

    riod, only about 14% of the expanded BMSC remained clo-

    nogenic (data not shown). When the capacity to promotebone formation in the nude mice assay was evaluated, we

    observed that the osteogenic potential of these expanded

    CFU-F decreased about 36 times with respect to CFU-F in

    fresh bone marrow. We found that, with fresh marrow, 1

    mm3 of bone could be formed for every 161 CFU-F im-

    planted, whereas, with expanded BMSC, 1 mm3 could be

    produced for every 5,781 CFU-F implanted, on average.

    Lifespan and maintenance

    of differentiation potential of BMSC clones

    One hundred twenty BMSC clones were obtained from five

    different marrow samples, of which 80 were able to reach

    confluence after passing from the cloning 96 well into one

    well of a 24-well plate. At this stage, they were divided into

    four replicas for induction of differentiation. Only three of

    the seven theoretically possible phenotypes (O, C, A, OC,

    OA, CA, OCA) were observed: 34% of the clones were os-

    teo-chondro-adipogenic (tripontential or OCA clones), 61%

    were osteo-chondrogenic (bipotential or OC clones), and

    5% were purely osteogenic (O clones).

    Lifespan and the ability to maintain differentiation po-

    tential with time in culture were determined for three OCA

    and two OC clones (Table 4). OCA clones were able to

    reach 2223 doublings after about 80 days of culture (Fig.4). All had lost adipogenic potential when tested at passage

    3 (equivalent to 22 doublings), but maintained their osteo-

    chondrogenic potential. The two OC clones had a slightly

    shorter lifespan, reaching 19 and 22 doublings, respectively.

    At passage 3, one clone lost its chondrogenic poten-

    tial, whereas the other clone maintained its bipotentiality

    (Table 4).

    Discussion

    The data presented in this study indicate that, after in vitro

    culture, the population of expanded BMSC progressively

    lost their ability to proliferate, differentiate in multiple mes-

    enchymal lineages, and efficiency in generating mature

    bone tissue. This effect is clearly evident after the first pas-

    Table 3. Osteogenic efficiency of fresh bone marrow versus primary

    expanded BMSC

    Fresh marrow* Expanded BMSC

    Population doublings 0 17.5

    Total bone formed (mm3)

    by 103 CFU-F 6.19 0.17

    Osteogenic unit 161 5,781

    In vivo implants were performed with 2.5 105 expanded BMSC or 5

    106 nucleated cells from fresh marrow and the CFU-F content of the cul-

    tures was determined as described in the Materials and methods section.

    From the results of these experiments, the expected yield of 103 fresh or ex-

    panded CFU-F and the sizes of the respective osteogenic units were calcu-

    lated.

    * Average of triplicate samples of one representative experiment. Average of duplicate samples of one representative experiment. Number of clonogenic CFU-F necessary to induce the formation of 1

    mm3 of bone when implanted as described in the Materials and methods

    sections.

    Table 4. Lifespan and differentiation potential of tripotential and

    bipotential clones

    Clone

    Doubling

    no. O C A

    Doubling

    no. O C A

    1 19 22

    2 19 23

    3 19 23

    4 18 19

    5 19 22

    Clone lifespan ranged between 19 and 23 cell doublings. OCA clones

    reached 2223 doublings, whereas OC clones had a slightly shorter

    lifespan, reaching 19 and 22 doublings. The three OCA clones lost adipo-

    genic potential by 22 doublings, although they maintained osteo-chondro-

    genic potential. Of the two OC clones tested, one lost its chondrogenic poten-

    tial, whereas the other clone maintained both osteogenic and chondrogenic

    potential.

    positive; negative.

    A adipogenic phenotype; C chondrogenic phenotype; O osteo-

    genic phenotype.

  • 8/9/2019 Banfi 2000 Experimental-Hematology

    7/9

    A. Banfi et al. /Experimental Hematology 28 (2000) 707715 713

    sage and is possibly already occurring during the primary

    culture, as indicated by the bone-forming efficiency assay.

    This behavior is consistent with a population undergoing

    commitment and differentiation, although this appears to be

    a gradual process.

    The presence in the BMSC population of progenitors for

    both the mesenchymal family of tissues and for hemopoi-

    esis-regulating stroma, together with the fact that these cells

    can be easily isolated from bone marrow aspirates and can

    be conveniently expanded and transfected in vitro, has

    prompted a wave of interest in their exploitation as vehicles

    for cell and gene therapy of both hematologic and musculo-skeletal pathologies.

    In the various therapeutic settings currently under inves-

    tigation, the need for BMSC to display stem cell features

    varies considerable. When used for hemophilia B gene ther-

    apy, BMSC are required to engraft and survive in a site with

    access to the general circulation and secrete factor IX with-

    out necessarily proliferating and differentiating; therefore,

    the presence of stem cells is not the main issue. On the other

    hand, if the reinfused BMSC are to reconstitute the he-

    mopoiesis-supporting stroma or, even more so, are to sus-

    tain healthy osteogenesis for the whole life of the recipient,

    possibly a young child, clearly what will determine the last-

    ing success of the procedure is the number of stem cells or

    very early progenitors present in the reinfused population.

    We sought, therefore, to study BMSC stem characteristics

    during culture expansion.

    Concepts currently accepted as defining a stem cell are

    the self-renewing capability, the high capacity for cell divi-

    sion (lifespan) maintained throughout the lifetime of the or-

    ganism, and the multipotential differentiation capability

    [31]. These criteria might not apply to cells of postnatal

    mesenchymal tissues, where the apparent cell plasticity

    might play the same role without the need for a real stem

    cell compartment, given the low tissue turnover in adult-

    hood [4,9]. In other words, the existence of the MSC in the

    postnatal organism is still debatable.

    To test the self-renewing ability of expanding BMSC, we

    investigated the growth kinetics of primary cultures and

    monitored their ability to differentiate into the three lin-

    eages considered at each passage.Gene expression for lineage-specific markers was ana-

    lyzed by RT-PCR and confirmed the immunochemical data.

    Interestingly, low but detectable levels of expression for the

    three markers considered were present before stimulation,

    indicating a possible commitment of some the these cells to

    differentiate into the three lineages tested already at the pri-

    mary culture stage. Noteworthy is that, in long-term unstim-

    ulated cultures, only the level of osteocalcin expression was

    increased spontaneously with respect to the basal value.

    This may indicate that the osteogenic pathway is the pre-

    ferred lineage through which these cells progress, possibly

    because of an intrinsic commitment or because the in vitroculture conditions represent a microenvironment favoring

    osteogenesis.

    The kinetics of the differentiation to the three lineages

    during mitotic expansion show that adipogenesis starts fad-

    ing by 18 doublings and is totally lost by 22 passages, when

    chondrogenesis and osteogenesis start weakening. In agree-

    ment with Bruder et al. [32], we found a slowing of cell pro-

    liferation as a function of increasing passages. Although

    these authors report a faster proliferation rate at every pas-

    sage considered, this may reflect different culture condi-

    tions, whereas the trend toward decreasing replication poten-

    tial with increasing cumulative doublings is in concordance

    with our data.This orderly loss of differentiation potential, paralleling

    that of proliferation velocity, strongly points to a progessive

    commitment of the population as a whole and to the inabil-

    ity, in these culture conditions, of the putative stem cells in

    the expanding population to self-renew and maintain a

    pluripotent differentiative capability.

    Interestingly, the largest drop in proliferation rate in all

    three primary cultures occurs on the first passage, whereas

    the differentiation potential is maintained for one passage

    longer. At the primary culture stage, CFU-F are plated to-

    Figure 4. Lifespan and differentiation potential of BMSC tripotential

    clones. The lifespan of three OCA clones was determined and the mainte-

    nance of the original differentiation potential at increasing cell doublings

    was verified. They reached 2223 doublings in about 80 days of culture.

    By then, they had lost any apparent capability of proliferating. The threee

    clones maintained the original differentiation potential up to 19 doublings,

    but they lost the adipogenic potential by the 22nd doubling still retaining

    the OC potential The curve represents the average cumulative doubling

    number of the three clones versus time in culture ( SE). The differen-

    tiation ability is represented by the underlying histograms. Arbitrary units:

    0 negative; 1 borderline; 2 positive, as defined in the Material and

    methods section.

  • 8/9/2019 Banfi 2000 Experimental-Hematology

    8/9

    714 A. Banfi et al. /Experimental Hematology 28 (2000) 707715

    gether with the whole marrow microenvironment: a part of

    this is made of nonadherent cells (mainly hemopoietic) and

    is removed with the medium changes, but a part is surely

    adherent to the plastic or the stromal cells themselves (mac-

    rophages, endothelial cells, and early hemopoietic progeni-

    tors at least) and can be responsible for a complex network

    of soluble and cell-contact signals to the BMSC. It is tempt-

    ing to speculate that dilution of this nonproliferating com-

    partment with the first passage can be related to BMSC loss

    of proliferation and multidifferentiative ability. Further ex-

    periments are needed to test this hypothesis.

    It is possible that stem cells are present in the initial cul-

    ture and that they undergo unequal division during expan-

    sion, producing a vast progeny of committed and differenti-

    ating daughter cells and becoming progressively diluted

    during the following passages. The nearest approximation

    to the putative MSC that can be studied is the CFU-F.

    Therefore, we compared the osteogenic efficiency of native

    CFU-F in fresh marrow to that of expanded CFU-F in first

    confluence BMSC. The result was that first confluence ex-panded CFU-F were 36 times less efficient than fresh mar-

    row samples at generating bone. The in vivo bone formation

    assay depends on the presence in the sample of a sufficient

    number of putative stem cells and/or early progenitors able

    to proliferate and give rise to a consistent differentiated

    progeny. Therefore, after in vitro expansion of about 17

    doublings, which is in the range necessary to obtain a thera-

    peutically useful numbers of cells, the clonogenic popula-

    tion in a BMSC primary culture is significantly depleted of

    osteogenic early progenitors/putative stem cells in compari-

    son with a fresh bone marrow sample.

    Results of experiments we performed on a number ofclones to determine their actual lifespan are consistent with

    the results obtained from primary cultures. Results indicate

    that these cells are able to undergo a limited number of mi-

    totic divisions, with characteristic kinetics, progressing

    faster at the primary culture level, gradually slowing down

    during the following passages, and reaching a complete stop

    after 2223 cell doublings. Futhermore, the three OCA

    clones analyzed displayed a loss of adipogenic potential by

    doubling 22, and one of two OC clones lost its chondro-

    genic potential by 22 cell doublings.

    Ex vivo expansion is necessary to obtain a sufficient

    number of cells for in vitro engineering and use in novel

    strategies of gene and cell therapy. When the reinfusedBMSC are used to sustain healthy histogenesis for the rest

    of the life of the patients, possibly young children, as in the

    case of osteogenesis imperfecta, the stem cell content of the

    population delivered clearly is of paramount importance

    and the effects of the in vitro expansion stage must be con-

    sidered. However, it is not known how many stem cells/

    early progenitors are needed in the reinfused population to

    obtain a therapeutic benefit, and it is possible that a limited

    expansion would be compatible with a successful proce-

    dure. A better understanding of the culture conditions re-

    sponsible for BMSC maintenance in an earlier progenitor

    state is desirable.

    AcknowledgmentsPartially supported by grants from Associazione Italiana Ricerca

    sul Cancro, Italian Space Agency (ASI) and European Space

    Agency (ESA). We wish to thank the Bone Marrow TransplantCentres at both S. Martino Hospital and G. Gaslini Pediatric Hos-

    pital for their factive collaboration and Prof. G. Corte for helpful

    discussion.

    References1. Bentley SA (1982) Bone marrow connective tissue and the haemopoi-

    etic microenvironment. Br J Haematol 50:1

    2. Dexter TM (1982) Stromal cell associated haemopoiesis. J Cell Phys-

    iol Suppl 1:87

    3. Friedenstein AJ (1990) Osteogenic stem cells in the bone marrow.

    Bone Miner Res 7:243

    4. Beresford J (1989) Osteogenic stem cells and the stromal system of

    bone and marrow. Clin Orthop Rel Res 240:2705. Caplan AI (1991) Mesenchymal stem cell. J Orthop Res 9:641

    6. Brockbank KG, de Jong JP, Piersma AH, Voerman JS (1986) He-

    mopoiesis on purified bone-marrow-derived reticular fibroblast in

    vitro. Exp Hematol 14:386

    7. Friedenstein AJ, Chailakhyan RK, Latsinik NV, Panasyuk AF, Keiliss-

    Borok IV (1974) Stromal cells responsible for transferring the mi-

    croenvironment of the hemopoietic tissues. Cloning in vitro and re-

    transplantation in vivo. Transplantation 17:331

    8. Tavassoli M, Friedenstein A (1983) Hemopoietic stromal microenvi-

    ronment. Am J Hematol 15:195

    9. Owen M (1988) Marrow stromal stem cells. J Cell Sci 10:63

    10. Beresford JN, Bennett JH, Devlin C, Leboy PS, Owen ME (1992) Evi-

    dence for an inverse relationship between the differentiation of adipo-

    cytic and osteogenic cells in rat marrow stromal cell cultures. J Cell

    Sci 102:34111. Johnstone B, Hering TM, Caplan AI, Goldberg VM, Yoo JU (1998) In

    vitro chondrogenesis of bone marrow-derived mesenchymal progeni-

    tor cells. Exp Cell Res 238:265

    12. Cherington V, Chiang GG, McGrath CA, et al. (1998) Retroviral vec-

    tor-modified bone marrow stromal cells secrete biologically active fac-

    tor IX in vitro and transiently deliver therapeutic levels of human fac-

    tor IX to the plasma of dogs after reinfusion. Hum Gene Ther 9:1397

    13. Galotto M, Berisso G, Delfino L, et al. (1999) Stromal damage as con-

    sequence of high-dose chemo/radiotherapy in bone marrow transplant

    recipients. Exp Hematol 27:1460

    14. Barbot C, Rice A, Vanes I, Mahon FX, Jazwiec B, Reiffers J (1994)

    Quality and functional capacity of the bone marrow microenvironment

    of autologous blood stem cell transplantation (ABSCT) recipients.

    Nouv Rev Fr Hematol 36:325

    15. Carlo-Stella C, Tabilio A, Regazzi E, et al. (1997) Effect of chemo-

    therapy for acute myelogenous leukemia on hematopoietic and fibro-

    blast marrow progenitors. Bone Marrow Transplant 20:465

    16. del Canizo C, Lopez N, Caballero D, et al. (1999) Haematopoietic

    damage persists 1 year after autologous peripheral blood stem cell

    transplantaion. Bone Marrow Transplant 23:901

    17. Domenech J, Gihana E, Dayan A, et al. (1994) Haemopoiesis of trans-

    planted patients with autologous marrows assessed by long-term mar-

    row culture. Br J Haematol 88:488

    18. Domenech J, Roingeard F, Herault O, et al. (1998) Changes in the

    functional capacity of marrow stromal cells after autologous bone mar-

    row transplantation. Leuk Lymphoma 29:533

    19. Radford JA, Testa NG, Crowther D (1990) The long-term effects of

  • 8/9/2019 Banfi 2000 Experimental-Hematology

    9/9

    A. Banfi et al. /Experimental Hematology 28 (2000) 707715 715

    MVPP chemotherapy for Hodgkins disease on bone marrow function.

    Br J Cancer 62:127

    20. Rice, A, Reiffers J, Bernard P, et al. (1992) Incomplete stroma forma-

    tion after allogeneic marrow or autologous blood stem cell transplanta-

    tion. Nouv Rev Fr Hematol 34:167

    21. Testa NG, Hendry JH, Molineux G (1985) Long-term bone marrow

    damage in experimental systems and in patients after radiation or che-

    motherapy. Anticancer Res 5:101

    22. Koc ON, Gerson SL, Cooper BW, et al. (1998) Rapid hematopoieticrecovery after coinfusion of autologous culture-expanded human mes-

    enchymal stem cells (hMSCs) and PBPCs in breast cancer patients re-

    ceiving high-dose chemotherapy. Blood 92:274a (abstr)

    23. Onyia JE, Clapp DW, Long H, Hock JM (1998) Osteoprogenitor cells

    as targets for ex vivo gene transfer. J Bone Miner Res 13:20

    24. Hou Z, Nguyen Q, Frenkel B, et al. (1999) Osteoblast-specific gene

    expression after transplantation of marrow cells: implications for skel-

    etal gene therapy. Proc Natl Acad Sci U S A 96:7294

    25. Pereira RF, OHara MD, Laptev AV, et al. (1998) Marrow stromal

    cells as a source of progenitor cells for nonhematopoietic tissues in

    transgenic mice with a phenotype of osteogenesis imperfecta. Proc

    Natl Acad Sci U S A 95:1142

    26. Horwitz EM, Prockop DJ, Fitzpatrick LA, et al. (1999) Transplantabil-

    ity and therapeutic effects of bone marrow-derived mesenchymal cells

    in children with osteogenesis imperfecta. Nat Med 5:309

    27. Martin I, Muraglia A, Campanile G, Cancedda R, Quarto R (1997) Fi-

    broblast growth factor-2 supports ex vivo expansion and maintenance

    of osteogenic precursors from human bone marrow. Endocrinology

    138:4456

    28. Chomczynski P, Sacchi N (1987) Single-step method of RNA isolation

    by acid guanidinium thiocyanate-phenol-chloroform extraction. Anal

    Biochem 162:15629. Hicok KC, Thomas T, Gori F, Rickard DJ, Spelsberg TC, Riggs BL

    (1998) Development and characterization of conditionally immortal-

    ized osteoblast precursor cell lines from human bone marrow stroma. J

    Bone Miner Res 13:205

    30. Rickard DJ, Kassem M, Hefferan TE, Sarkar G, Spelsberg TC, Riggs

    BL (1996) Isolation and characterization of osteoblast precursor cells

    from human bone marrow. J Bone Miner Res 11:312

    31. Morrison SJ, Shah NM, Anderson DJ (1997) Regulatory mechanisms

    in stem cell biology. Cell 88:287

    32. Bruder SP, Jaiswal N, Haynesworth SE (1997) Growth kinetics, self-

    renewal, and the osteogenic potential of purified human mesenchymal

    stem cells during extensive subcultivation and following cryopreserva-

    tion. J Cell Biochem 64:278