ArticlePDF Available

Engraftment in Nonobese Diabetic Severe Combined Immunodeficient Mice of Human CD34 1 Cord Blood Cells After Ex Vivo Expansion: Evidence for the Amplification and Self-Renewal of Repopulating Stem Cells

Authors:

Abstract and Figures

Understanding the repopulating characteristics of human hematopoietic stem/progenitor cells is crucial for predicting their performance after transplant into patients receiving high-dose radiochemotherapy. We have previously reported that CD34(+) cord blood (CB) cells can be expanded in vitro for several months in serum containing culture conditions. The use of combinations of recombinant early acting growth factors and the absence of stroma was essential in determining this phenomenon. However, the effect of these manipulations on in vivo repopulating hematopoietic cells is not known. Recently, a new approach has been developed to establish an in vivo model for human primitive hematopoietic precursors by transplanting human hematopoietic cells into sublethally irradiated nonobese diabetic severe combined immunodeficient (NOD/SCID) mice. We have examined here the expansion of cells, CD34(+) and CD34(+)38(-) subpopulations, colony-forming cells (CFC), long-term culture initiating cells (LTC-IC) and the maintenance or the expansion of SCID-repopulating cells (SRC) during stroma-free suspension cultures of human CD34(+) CB cells for up to 12 weeks. Groups of sublethally irradiated NOD/SCID mice were injected with either 35,000, 20,000, and 10,000 unmanipulated CD34(+) CB cells, which were cryopreserved at the start of cultures, or the cryopreserved cells expanded from 35,000, 20,000, or 10,000 CD34(+) cells for 4, 8, and 12 weeks in the presence of a combination of early acting recombinant growth factors (flt 3/flk2 ligand [FL] + megakaryocyte growth and development factor [MGDF] +/- stem cell factor [SCF] +/- interleukin-6 [IL-6]). Mice that had been injected with >/=20,000 fresh or cryopreserved uncultured CD34(+) cells did not show any sign or showed little engraftment in a limited number of animals. Conversely, cells that had been generated by the same number of initial CD34(+) CB cells in 4 to 10 weeks of expansion cultures engrafted the vast majority of NOD/SCID mice. The level of engraftment, well above that usually observed when the same numbers of uncultured cells were injected in the same recipients (even in the presence of irradiated CD34(-) cells) suggested that primitive hematopoietic cells were maintained for up to 10 weeks of cultures. In addition, dilution experiments suggest that SRC are expanded more than 70-fold after 9 to 10 weeks of expansion. These results support and extend our previous findings that CD34(+) CB stem cells (identified as LTC-IC) could indeed be grown and expanded in vitro for an extremely long period of time. Such information may be essential to design efficient stem cell expansion procedures for clinical use.
Content may be subject to copyright.
1999 93: 3736-3749
Fagioli, Eliana Perissinotto, Giuliana Cavalloni, Orit Kollet, Tsvee Lapidot and Massimo Aglietta
Wanda Piacibello, Fiorella Sanavio, Antonella Severino, Alessandra Danè, Loretta Gammaitoni, Franca
the Amplification and Self-Renewal of Repopulating Stem Cells
of Human CD34+ Cord Blood Cells After Ex Vivo Expansion: Evidence for
Engraftment in Nonobese Diabetic Severe Combined Immunodeficient Mice
http://bloodjournal.hematologylibrary.org/content/93/11/3736.full.html
Updated information and services can be found at:
(3132 articles)Hematopoiesis and Stem 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.
20036.
the American Society of Hematology, 2021 L St, NW, Suite 900, Washington DC
Blood (print ISSN 0006-4971, online ISSN 1528-0020), is published weekly by
For personal use only. by guest on June 7, 2013. bloodjournal.hematologylibrary.orgFrom
HEMATOPOIESIS
Engraftment in Nonobese Diabetic Severe Combined Immunodeficient
Mice of Human CD34Cord Blood Cells After Ex Vivo Expansion:
Evidence for the Amplification and Self-Renewal of Repopulating Stem Cells
By Wanda Piacibello, Fiorella Sanavio, Antonella Severino, Alessandra Dane`, Loretta Gammaitoni,
Franca Fagioli, Eliana Perissinotto, Giuliana Cavalloni, Orit Kollet, Tsvee Lapidot, and Massimo Aglietta
Understanding the repopulating characteristics of human
hematopoietic stem/progenitor cells is crucial for predicting
their performance after transplant into patients receiving
high-dose radiochemotherapy. We have previously reported
that CD34cord blood (CB) cells can be expanded in vitro for
several months in serum containing culture conditions. The
use of combinations of recombinant early acting growth
factors and the absence of stroma was essential in determin-
ing this phenomenon. However, the effect of these manipula-
tions on in vivo repopulating hematopoietic cells is not
known. Recently, a new approach has been developed to
establish an in vivo model for human primitive hematopoi-
etic precursors by transplanting human hematopoietic cells
into sublethally irradiated nonobese diabetic severe com-
bined immunodeficient (NOD/SCID) mice. We have exam-
ined here the expansion of cells, CD34and CD3438
subpopulations, colony-forming cells (CFC), long-term cul-
ture initiating cells (LTC-IC) and the maintenance or the
expansion of SCID-repopulating cells (SRC) during stroma-
free suspension cultures of human CD34CB cells for up to
12 weeks. Groups of sublethally irradiated NOD/SCID mice
were injected with either 35,000, 20,000, and 10,000 unma-
nipulated CD34CB cells, which were cryopreserved at the
start of cultures, or the cryopreserved cells expanded from
35,000, 20,000, or 10,000 CD34cells for 4, 8, and 12 weeks in
the presence of a combination of early acting recombinant
growthfactors (flt 3/flk2ligand[FL] megakaryocyte growth
and development factor [MGDF] stem cell factor [SCF]
interleukin-6[IL-6]). Mice thathadbeen injected with H20,000
fresh or cryopreserved uncultured CD34cells did not show
any sign or showed little engraftment in a limited number of
animals. Conversely, cells that had been generated by the
same number of initial CD34CB cells in 4 to 10 weeks of
expansion cultures engrafted the vast majority of NOD/SCID
mice. The level of engraftment, well above that usually
observed when the same numbers of uncultured cells were
injected in the same recipients (even in the presence of
irradiated CD34cells) suggested that primitive hematopoi-
etic cells were maintained for up to 10 weeks of cultures. In
addition, dilution experiments suggest that SRC are ex-
panded more than 70-fold after 9 to 10 weeks of expansion.
These results support and extend our previous findings that
CD34CB stem cells (identified as LTC-IC) could indeed be
grown and expanded in vitro for an extremely long period of
time. Such information may be essential to design efficient
stem cell expansion procedures for clinical use.
r
1999 by The American Society of Hematology.
AN ESSENTIAL PROPERTY of hematopoietic stem cells
is their ability to divide without significant alteration of
their proliferative potential or differentiation state. Characteriza-
tion and quantitation of these progenitor cells is fundamental to
our understanding of the developmental sequence, and it is of
great importance for human hematopoietic cell transplantation,
ex vivo expansion, and gene therapy.1-4 To date, quantitative
analysis of human primitive hematopoietic cells has been
limited to in vitro studies using colony assays (colony-forming
cells [CFC]) or long-term cultures (LTC). CFC assays detect
only committed and multipotent progenitors. LTC assays detect
more primitive cells (LTC-initiating cells [IC]), capable of
generating myeloid colonies for at least 5 weeks of culture on
competent feeder layers.5,6
The transplantation assay available in the mouse system has
been instrumental in defining and characterizing the most
primitive elements of the hematopoietic system.7Recently, a
similar in vivo approach, derived from the work in the mouse,
has become available for humans. As a result, several groups
have transplanted human hematopoietic precursors into differ-
ent mouse mutants in an attempt to develop a reproducible
transplantation assay.7-9 In particular, the intravenous injection
of human hematopoietic precursors in sublethally irradiated
severe combined immunodeficient (SCID) and nonobese dia-
betic/SCID (NOD/SCID) mice has resulted in the engraftment
of primitive human cells that proliferate and differentiate to
multiple lineages in the murine bone marrow (BM) and
spleen.10-13 The transplanted human cells home to and engraft
the murine BM, where they proliferate and differentiate to
produce large numbers of LTC-IC, CFC, immature and mature
myeloid, erythroid, and lymphoid cells without the influence of
exogenously supplied human growth factors.14 The engrafted
human cells have been defined SCID-repopulating cells (SRC)15
or, in a similar quantitative assay, competitive repopulating
units (CRU).16 Although the SRC (or CRU) represents a very
primitive hematopoietic cell, the exact place in the stem cell
From the Department of Biomedical Sciences and Human Oncology,
University of Torino Medical School, Torino; the Hematology/Oncology
Section, Mauriziano Hospital, Torino; the Pediatric Department; the
Institute for Cancer Research and Treatment (IRCC), Candiolo, Torino,
Italy; and the Department of Immunology, the Weizmann Institute of
Science, Rehovot, Israel.
Submitted May 11, 1998; accepted January 29, 1999.
Supported by grants from Associazione Italiana per la Ricerca sul
Cancro (AIRC; Milan, Italy) and from the Ministero dell’Universita`e
della Ricerca Scientifica e Tecnologica (MURST) (to W.P. and M.A.).
A.D. is a recipient of the FIRC grant; A.S. and L.G. are both recipients
of the ‘G. Ghirotti Foundation,’’sez. Piemonte grants.
Address reprint requests to Wanda Piacibello, MD, Department of
Biomedical Sciences and Human Oncology, Clinical Section, Via
Genova 3, 10126 Torino, Italy; e-mail: w.piacibello@mail.ircc.unito.it.
The publication costs of this article were defrayed in part by page
charge payment. This article must therefore be hereby marked ‘adver-
tisement’’ in accordance with 18 U.S.C. section 1734 solely to indicate
this fact.
r1999 by The American Society of Hematology.
0006-4971/99/9311-0001$3.00/0
3736
Blood,
Vol 93, No 11 (June 1), 1999: pp 3736-3749
For personal use only. by guest on June 7, 2013. bloodjournal.hematologylibrary.orgFrom
hierarchy and its relation to early in vitro hematopoietic
progenitors, such as LTC-IC, is not fully understood. SRC have
been reported to be biologically distinct from and more
primitive than most CFC and LTC-IC: SRC are exclusively
CD34CD38in contrast to CFC and LTC-IC, which are found
also in the CD34CD38fraction.17-19 By contrast, according to
Eaves’s data, even though the cells identified by the LTC-IC
and the CRU assay may not necessarily represent identical
cell populations, they belong to both CD34CD38and
CD34CD38subpopulations and seem to increase in response
to the same culture conditions in vitro.16
We conclude that this experimental transplantation assay that
measures the repopulating potential of various human progeni-
tor fractions is most valuable in increasing our understanding of
several biological properties of hematopoietic progenitors for
both experimental and clinical hematology.
A second very important issue, to date, is the identification of
culture conditions that support the self-renewal and the expan-
sion of human hematopoietic stem cells. In particular, cord
blood (CB) has recently attracted attention as a source of
hematopoietic stem cells for both transplantation and gene
therapy applications.20-24 However, concern that a single CB
collection may not be sufficient to guarantee engraftment of
adult allogeneic recipients has also stimulated considerable
interest in expanding CB stem cell number in vitro. Experi-
ments conducted in our laboratories as well as data from others
have recently shown that CD34(or CD34CD38) CB cells
can indeed be expanded in fairly well-defined culture condi-
tions.25-29 The common denominator of the expansion systems
was the absence of stroma layers and hence the use of
combinations of recombinant early acting hematopoietic growth
factors. A net expansion of LTC-IC was observed in all
cases.25-29 In addition, it was shown that 4-day or 5- to 8-day
stroma-free liquid cultures of CB cells in the presence of
flt3/flk2 ligand (FL), stem cell factor (SCF), interleukin-3
(IL-3), IL-6 without29 or with granulocyte colony-stimulating
factor (G-CSF)16 supported LTC-IC expansion and also a
modest, albeit significant expansion, of CRU in NOD/SCID
mouse recipients. Conversely, it has been reported that cocul-
tures of human BM and CB cells with allogeneic human stroma
resulted in a reduced repopulating capacity of cocultured cells,
which, by contrast, contained an equal or even higher number of
CFC and LTC-IC as compared with uncultured cells.30 On the
other hand, very recently Xu et al31 reported that in vivo
long-term repopulating hematopoietic stem cells from CB could
be maintained for at least 4 weeks when cocultured on a stroma
cell line derived from the aorta-gonad-mesonephros region of
mouse embryo.
The aim of our studies was to investigate whether the
combinations of early acting growth factors, which in our
previous studies proved capable of inducing a massive and
prolonged expansion of hematopoietic progenitors and more
primitive LTC-IC from CB cells,27,28 could also maintain the in
vivo repopulating ability of human stem cells, or even amplify
their number.
We show that CD34CB cells can be expanded for up to 10
weeks in stroma-free cultures in the presence of FL, megakaryo-
cyte growth and development factor (MGDF), SCF, and IL-6
without losing their in vivo repopulating potential. Furthermore,
our studies show that considerable expansion of SRC is
obtainable in vitro, as sublethally irradiated NOD/ SCID mice
are consistently and reproducibly repopulated by the equivalent
of 1,250, 625, and 312 initial CD34CB cells, which have been
expanded in vitro for up to 9 to 10 weeks.
MATERIALS AND METHODS
Human cells. Human BM was obtained by aspiration from the
posterior iliac crest of fully informed hematologically normal donors.
Umbilical CB was obtained at the end of full-term deliveries, after
clamping and cutting of the cord, by drainage of blood into sterile
collection tubes containing the anticoagulant citrate-phosphate dex-
trose.
CD34cell purification. Mononuclear cells (MNC) were isolated
from CB using Ficoll Hypaque (density, 1077; Nyegaard, Oslo,
Norway) density centrifugation. Cells were subjected to two cycles of
plastic adherence (60 minutes each); they were then washed with
Hanks’ Balanced Salt Solution (HBSS, GIBCO BRL, Grand Island,
NY). The CD34MNC fraction was isolated with superparamagnetic
microbead selection using high-gradient magnetic field and miniMACS
column (Miltenyi Biotech, Gladbach, Germany). The efficiency of the
purification was verified by flow cytometry counterstaining with a
CD34-phycoerythrin (PE; HPCA-2; Becton Dickinson, San Jose, CA)
antibody. In the cell fraction containing purified cells, the percentage of
CD34cells ranged from 90% to 98%.
Recombinant human cytokines. The following recombinant puri-
fied human cytokines were used in these studies: recombinant human
(rh) stem cell factor (rhSCF) and rh megakaryocyte growth and
development factor (MGDF) were a generous gift from Amgen
(Thousand Oaks, CA); recombinant human granulocyte colony-
stimulating factor (rhG-CSF) was from Genzyme (Cambridge, MA);
recombinant human granulocyte-macrophage colony-stimulating factor
(rhGM-CSF), recombinant human interleukin-6 (rhIL-6) and recombi-
nant human interleukin-3 (rhIL-3) were from Sandoz (Basel, Switzer-
land); recombinant human erythropoietin (rhEPO; EPREX) was from
Cilag (Milan, Italy); recombinant human FLT3-ligand (rhFL) was
kindly provided by S.D. Lyman (Immunex Corp, Seattle, WA).
Animals. NOD/LtSz scid/scid (NOD/SCID) mice were obtained
from Charles River Italia (Calco, Italy) and maintained in the animal
facilities at Antoine Marxer-RBM (Colleretto Giacosa, Italy). In part of
the studies, mice were bred and maintained at the animal facilities of the
Weizman Institute of Science (Rehovot, Israel).
All animals were handled under sterile conditions and maintained in
cage microisolators. Mice to be transplanted were irradiated at 6 to 8
weeks of age with 350 to 375 cGy of total body irradiation from a 137Cs
source and then within 24 hours were given a single intravenous
injection of: (1) human CD34CB cells, which had been previously
separated from several CB samples, then pooled, and cryopreserved
(control cells). After thawing, 97% to 99% CD34cells were viable by
trypan blue dye exclusion; (2) cells were harvested from expansion
cultures as described. Also the latter cells were cryopreserved and
injected at the same time as the control cells. When low numbers of
unmanipulated CD34cells were to be transplanted, at least 2 105
irradiated CD34cells were coinjected as carrier cells.
Mice were killed 6 to 8 weeks posttransplant for assessment of the
number and types of human cells detectable in both femurs, tibias, and
spleen. As a control for the frequency of SRC in cryopreserved CD34
CB cells, the same numbers of fresh CD34cells from pooled CB
samples were injected in similarly pretreated NOD/SCID recipients; the
frequency of SRC was not significantly different from that found in
cryopreserved CD34cells.
Flow cytometric detection of human cells in murine tissues. BM
cells were flushed from the femurs and tibias of each mouse to be
assessed using a syringe and a 26-gauge needle. To prepare cells for
EXPANSION OF IN VIVO REPOPULATING STEM CELLS 3737
For personal use only. by guest on June 7, 2013. bloodjournal.hematologylibrary.orgFrom
flow cytometry, contaminating red blood cells were lysed with 8.3%
ammonium chloride and the remaining cells were then washed in HBSS
with 0.1% bovine serum albumin (BSA; Sigma Chemical Co, Milan,
Italy), 0.01% sodium azide (HFN). The cells were then resuspended at 1
to2106cells/mL and incubated with mouse IgG (Fluka Chemika
Biochemika, Buchs, Switzerland) and with 5% human serum (HS), to
block nonspecific binding to Fc receptor. Cells were then incubated with
monoclonal antibodies (MoAb) specific for human CD45, CD71, and
glycophorin-A (GpA), directly labeled with fluorescein isothiocyanate
(FITC) or PE (all from Dako A/S, Glostrup, Denmark) for 30 minutes at
4°C to assess the total population of human hematopoietic cells. Cells
stained with an anti-CD45 conjugated to an R-phycoerythrin-Cy5
tandem conjugate were simultaneously stained with an anti-human-
CD34-PE (Becton Dickinson) and CD19-PE (Dako) for quantitation of
the total human CD34and CD19cell populations. In some mice,
additional aliquots were stained with anti-human-CD33-PE (Dako),
CD41-FITC in combination with anti-human-CD45-Cy5 RPE to allow
discrimination of subpopulations within the CD45 gate and with
CD71-FITC plus -GpA-PE.30 Some cells from each suspension were
similarly incubated with irrelevant (control) MoAbs labeled with FITC
and PE. Cells from an unmanipulated NOD/SCID mouse were also
stained with each of the MoAbs used for detecting positively stained
human cells. Only levels of fluorescence which excluded 99.9% of all
of these negative controls were considered as specific. After staining, all
cells were washed once in sodium azide (HFN) containing 2 µg/mL
propidium iodide (PI) to allow dead (PI) cells to be excluded from
analyses. Flow cytometric analysis was performed using a FACScan
cytometer (Becton Dickinson). At least 5,000 events were acquired for
each analysis. When fluorescent cells represented only a minority of the
total population (0.1%) many more events (at least 20,000) were
analyzed.
Hematopoietic cell cultures. Assays for granulopoietic, erythroid,
megakaryocytic, and multilineage (granulocyte-erythroid-macrophage-
megakaryocyte) colony-forming units (CFU-GM, BFU-E, CFU-Mk,
and CFU-GEMM, respectively) were usually performed as follows. For
CFU-GM, 1 103CD34CB cells of the initial cell suspension or
suitable aliquots of the stroma-free long-term cultures were cultured at 4
plates per point in 3% agar, 15% fetal calf serum (FCS) (HyClone,
Logan, UT) in Iscove’s Modified Dulbecco Medium (IMDM). For
CFU-Mk, the same number of cells was cultured in plasma-clot assay
(four dishes per point) as previously described.27 For BFU-E and
CFU-GEMM, the same number of cells was cultured in 1.3% methylcel-
lulose (Fluka) and IMDM containing 30% FCS at 37°C in a humidified
atmosphere at 5% CO2in air.27 Colony scoring was performed on day
12 for CFU-Mk (at the immunofluorescent microscope after staining
with an FITC-conjugated MoAb recognizing human GPIIbIIIa) and on
day 14 for CFU-GM, BFU-E, and CFU-GEMM.27,28 Several growth
factors were added at optimum concentrations to sustain the formation
of BFU-E and CFU-GEMM: rhuIL-3 (20 ng/mL), rhuGM-CSF (10
ng/mL), rhuEpo (3 U/mL), and rhuSCF (50 ng/mL). For CFU-GM,
rhuGM-CSF (20 ng/mL), rhuIL-3 (20 ng/mL) and rhuSCF (50 ng/mL)
were added. For CFU-Mk, rhuIL-3 (5 ng/mL) was used as a single
growth factor. When transplanted NOD/SCID mouse BM cells were to
be evaluated for their human hematopoietic progenitor content, the FCS
in the methylcellulose medium was replaced with an equivalent volume
of a pretested pool of equivalently supportive normal human serum and
bovine plasma in the plasma clot assay was replaced with an equivalent
volume of human plasma. Plasma clot assays were adopted not only to
detect CFU-Mk colonies (with the addition of rhuIL-3), but for
CFU-GM, BFU-E, and CFU-GEMM as well (with the addition of
rhuIL-3, rhuGM-CSF, rhuSCF, and rhuEPO). G-CSF was omitted to
minimize the stimulation of murine clonogenic cells. These culture
conditions have been reported to be selective for colony formation by
human progenitors and do not support coexisting murine progeni-
tors.8,13 In addition, colonies grown in plasma-clot and colonies plucked
from methylcellulose cultures were stained with FITC-conjugated
anti-human GPIIbIIIa, CD45, CD13, and GpA and scored at the
immunofluorescence microscope. The presence of fluorescent colonies
was the index of their human origin. As a control, BM cells from
untreated NOD/SCID mice were plated at identical cell concentrations
in the same culture assays (plasma clot and methylcellulose) containing
only human serum and/or human plasma and the above-reported
human-specific growth factors. Dishes were scored from day 12 up to
day 21: in these culture conditions no colonies could be detected.
LTC-IC. The LTC-IC content of cell suspension was determined by
limiting dilution assays as previously described.27 Briefly, 10 to 1,000
purified CD34CB cells at the start of cultures or suitable aliquots of
cultured cells were washed and seeded onto preestablished irradiated
human BM stromal layers (derived by culturing 107fresh BM MNC in a
T25 flask for at least 2 weeks in 5 mL stromal medium [12.5% horse
serum, 12.5% FCS, IMDM, 2-mercaptoethanol, 106mol/L hydrocorti-
sone, and penicillin/streptomycin] and by plating the irradiated [15 Gy]
and trypsinized stroma at 7 103/cm2in 24-well plates) and maintained
at 37°C for 5 to 6 weeks with weekly half media changes, at the end of
which all cells were harvested and plated for CFC determination in
methylcellulose medium. These cultures were incubated at 37°C for 2
weeks in the presence of 1.3% methylcellulose, 30% FCS, EPO (3
U/mL), IL-3 (20 ng/mL), G-CSF (20 ng/mL), GM-CSF (20 ng/mL), and
SCF (50 ng/mL); LTC-IC enumeration was based on the number of
CFU-C scored in the limiting dilution assay (LDA).
Stroma-free liquid cultures. Stroma-free expansion cultures were
performed as follows. (1) Atotal of 10 to 20,000 CD34CB cells were
cultured in quadruplicate flat-bottomed 24-well plates in 1 mL of
IMDM supplemented with 10% pooled normal human serum with the
following growth factors: FL (50 ng/mL) MGDF (20 ng/mL); SCF
(50 ng/mL) FL (50 ng/mL) MGDF (20 ng/mL); SCF (50 ng/mL)
FL (50 ng/mL) MGDF (20 ng/mL) IL-6 (10 ng/mL), which were
added to each series of microwells twice a week. The wells were grown
at 37°C. At initiation of the cultures, the number of CFC and CFU-Mk
present in 1 mL of a single well was determined by triplicate plasma clot
assays. Every week all of the wells were demidepopulated by removal
of one half the culture volume (and cells), which was replaced with
fresh medium and growth factors. Cells of the harvested media were
counted and suitable aliquots of the cell suspensions were assayed for
CFC and CFU-Mk content, for immunophenotype analysis (CD34,
CD34CD38) and for LTC-IC determination every 2 to 3 weeks by
LDA. The total number of CFC or of LTC-IC generated CFC was
calculated as previously reported.27,28
(2) In a second series of expansion cultures, the same concentrations
of CD34CB cells were cultured in identical culture conditions (same
growth factors and serum). Cultures were set up in quadruplicate. The
only difference with the series (1) was that every week, instead of being
split in two, the cell suspension of each well was resuspended in twice
its volume, split in two, and plated in new 24-well plates. This way
every week additional new 24-wells were set up (eg, derived from the 1
mL prepared at start of cultures, 2, 4, and 8 wells were set up at weeks 2,
3, and 4, respectively).
(3) In this series of experiments, 1 to 5 104CD34cells/mL were
deposited on the bottom of tissue culture T25 or T75 flasks in
quadruplicate. Every week the culture volume was doubled. Cell counts
were performed every week. At weeks 4, 6, 8, and 12, the immunophe-
notype of the cells harvested from the different sets of expansion was
performed, and the CFC content of each expansion set was determined
by seeding suitable aliquots of the pooled wells or flasks in triplicate
plasma clot cultures. For LTC-IC assay, limiting dilutions of the cell
suspensions deriving from each series of expansion wells or flasks were
seeded onto preirradiated stroma layers in 96-well plates for 5 weeks
and then the number of CFC generated by methylcellulose cultures was
enumerated as described above.
To limit the volume of the expansion cultures, in most experiments, at
3738 PIACIBELLO ET AL
For personal use only. by guest on June 7, 2013. bloodjournal.hematologylibrary.orgFrom
week 6, 7/8 of the total culture volume was cryopreserved and
expansion studies were performed with the remaining 1/8. When
inocula were to be prepared, 7/8 of week 6 cryopreserved cells were
thawed, washed, counted, and grown for an additional 6 to 10 days in
the presence of the same media and growth factors previously used;
hence, these inocula represented weeks 7 to 8 expanded cells. When
weeks 9 to 10 and week 12 expanded cells were to be injected, the 1/8
expanded cells (which had been cryopreserved at weeks 8 and 10,
respectively) were thawed, grown for an additional 6 to 10 days and
mixed with 7/8 of week 6 cryopreserved cells that had been thawed and
grown as previously described. The two samples were mixed, washed,
and resuspended in a small aliquot and injected in mice.
For limiting dilution studies, the 1/8 cells expanded for 6 and 8 weeks
(and then cryopreserved) were thawed and grown for an additional 6 to
10 days before being injected into mice. In these experiments, the cells
were not mixed with 7/8 of week 6 cryopreserved cells; hence they
represented the first dilution (1/8 of initial CD34cells). The next
dilutions were prepared from the 1/8 cells (1/16, 1/32, etc).
DNA extraction and analysis of human cell engraftment. High
molecular weight DNA was extracted from the BM of transplanted mice
by phenol-chloroform extraction using standard protocols. DNA was
digested with EcoRI and separated by agarose gel electrophoresis,
transferred onto a positively charged nylon membrane, and probed with
a labeled human chromosome 17-specific -satellite probe (p17H8)
(limit of detection, approximately 0.05% human DNA). To quantify the
level of human cell engraftment, the intensity of the characteristic
2.7-kb band in samples was compared with those of human: mouse
DNA control mixtures (0%, 0.1%, 1%, 10% human DNA).
Statistical analysis. For purposes of LDAs (LTC-IC and SRC),
Poisson statistics for the single-hit model were applied. The frequency
of LTC-IC and SRC in cell suspensions was calculated using maximum
likelihood estimator.32
RESULTS
Groups of sublethally irradiated NOD/SCID mice were
injected with decreasing concentrations of CD34CB cells,
which had been previously separated from several CB samples,
then pooled, and stored frozen until the time of transplant. Six to
8 weeks after inoculation, BM cells of the sacrificed animals
were obtained from both femurs and both tibias and assessed for
the presence of human hematopoietic cells. Table 1 presents a
summary of these data. While 2 105and 1 105CD34cells
Fig 1. Representative DNA analysis of human cell engraftment in
the BM of NOD/SCID mice transplanted with 1 104,2104, and
3.5 104unmanipulated CD34CB cells. Cells were injected together
with preirradiated 2 105CD34CB cells as carrier cells. Human DNA
was assessed by Southern analysis using a human chromosome
17-specific -satellite probe. Human:mouse controls are given as
percent human DNA.
Table 1. Main Characteristics of Injected CB CD34Cells and Their Ability to Engraft the BM of NOD/SCID Recipients
Injected/Mouse No. of
Positive Mice Engraftment (%) Human CFC (103)*CD34Cells CFC LTC-IC
210549,330 17,800 3/3† 12.5, 21, 7‡ 51.2, 98.4, 27.6
110524,665 8,900 4/4 1.3, 3, 11, 25 17.7, 14.2, 41.5, 77.6
510412,332 4,450 4/5 1.4, 16, 3, 8, 0 ND, 74.9, 10.2, 36.4, 0
3.5 1048,631 3,115 4/6 1.2, 2.3, 0, 0, 1.6, 3 4.9, 9.8, 0, 0, 8.08, 9.5
21044,933 1,780 3/7 0.18, 0.5, 0.3, 0, 0, 0, 0 0.76, 2.1, 1.9, 0, 0, 0, 0
11042,466 890 0/5 0, 0, 0, 0, 0 0, 0, 0, 0, 0
CD34cells deriving from 6 different CB samples were separated as described, pooled, and then stored frozen. At the time of injection, the
samples were thawed, washed, counted, resuspended in 300 µL of IMDM containing 10% FCS and then injected into 3 to 7 animals as described.
BM of NOD/SCID recipients was recovered from both the femurs and the tibias at week 8 posttransplant.
Abbreviation: ND, not done.
*A total of 1 105to5105BM cells of the transplanted mice were seeded in triplicate dishes for plasma clot assays. Recombinant human IL-3,
GM-CSF, SCF, and EPO were added at start of cultures. Dishes were scored after 14 days of incubation, after staining with MoAb as described in
Materials and Methods. Only fluorescent colonies were considered of human origin. Values are calculated taking into account that the BM of both
femurs and tibias represent 25% of the total BM.
†Positive mouse ⫽⬎0.1% of human CD45, CD71, and -GpA Acells in the whole BM on FACSscan analysis.
‡Percent of human CD45, CD71, and GpAcells in the whole BM on FACSscan analysis.
EXPANSION OF IN VIVO REPOPULATING STEM CELLS 3739
For personal use only. by guest on June 7, 2013. bloodjournal.hematologylibrary.orgFrom
Fig 2. (A) Representative fluorescence-activated cell sorting (FACS) profiles of marrow cells from NOD/SCID mice transplanted 8 weeks
previously with unmanipulated CD34CB cells. (a) Negative control: a nonengrafted mouse (transplanted with 2 105irradiated CD34CB
carrier cells). (Middle) A mouse transplanted with 1 105CD34CB cells. (Bottom) A mouse transplanted with 2 104CD34cells. CD45/CD34
and CD45/CD19 analysis was performed on total BM cells. (B) Multilineage engraftment in the BM of a representative mouse transplanted with
1105unmanipulated CD34CB cells. Analysis of lineage markers (CD45/CD34, CD19, CD41, and CD13/CD33) was performed on cells comprised
within the CD45 gate. Analysis of GpA/CD71-positive cells was performed on total BM cells.
Table 2. Comparison Between Three Different Expansion Protocols
Growth
Factors
Input 4wk 6wk 8wk
Demide-
population
Well Exp. Wells Flasks
Demide-
population
Well Exp. Wells Flasks
Demide-
population
Well Exp. Wells Flasks
FL MGDF
Cells 1060.02 3.7 1.09 2.97 0.87 4.8 1.8 15.2 7 14.2 6.4 14.02 7.9 42.4 24.4 39.8 25.6 37.4 19.9
CD34%918.5 4.08 1.0 3.76 0.98 3.6 0.99 1.83 0.6 1.90 0.8 2.57 1.1 2.14 0.9 2.35 0.87 2.61 0.77
CFC 1032.8 1.4 115.2 48.8 85.2 41.6 144.1 77.9 196.7 135.4 148.9 110.6 213 106.7 1,008 69.5 943.6 372.2 863.3 295.8
LTC-IC 1033.6 0.9 27.5 17.5 20.6 11.2 24.7 11.5 113.2 14.8 126.8 36.1 131.6 51.2 929.6 659.5 873.2 312 899.6 236
SCF FL
MGDF
Cells 1060.02 9.09 3.2 8.89 2.7 10.3 1.9 31.28 8 29.6 9.6 27.9 6.9 103.7 32.9 91 36.7 106 40.6
CD34%918.5 3.28 0.29 3.61 1.03 3.02 0.7 1.67 0.9 1.58 0.12 1.7 0.9 2.97 0.7 2.06 1.3 2.06 0.85
CFC 1032.8 1.4 194.2 60.2 177.7 55.7 255.3 41 314.7 99.1 527.1 85.8 298.7 106.2 1,252.6 508.1 1,196.6 676.4 1,066.5 737.4
LTC-IC 1033.6 0.9 48.2 24.8 46.4 19.9 57.4 22 172.8 122.4 232.4 150.5 204.6 79 1,012.9 795.6 985 345 1,003 604
IL6 SCF
FL MGDF
Cells 1060.02 10.01 3.2 11.74 2.7 6.96 2.91 55.6 21.7 49.5 19.6 57.1 23.6 113.4 49.8 100.4 38 249 73.1
CD34%918.5 4.32 0.95 3.77 1.4 3.73 0.7 2.47 0.8 1.98 0.6 2.55 0.8 2.87 0.85 1.94 0.7 2.03 0.9
CFC 1032.8 1.4 180.6 46.8 153.5 57.1 223.2 58.8 473 16 416.2 91 389.9 76.1 1,898.6 1,785 1,679.9 2,696 863.4
LTC-IC 1033.6 0.9 52.2 21.9 41.2 23.4 55.2 41 290.6 78.5 215.7 48 256.4 91.3 1,100.5 665 903.6 114.1 1,264 592
Twenty thousand CB CD34cells were resuspended in 1 mL IMDM containing 10% pooled human serum and cultured for up to 8 weeks in 24-well plates. In the first
series(demidepopulationwells)cellsin1mLofasinglewellweredemidepopulatedeveryweekbyremovalofone half the culture medium (and cells), which was replaced
with the same volume of fresh medium and growth factors. In the second series (expansion wells), 2 104CD34cells were prepared as described above in 24-well plates
in 1 mL. Every week the cells were resuspended in twice the volume of culture medium, so that every week additional new 24-wells were set up. In the third series (flasks)
2104CD34cells/mL were layered on the bottom of T flasks and every week the culture volume was doubled. At the indicated time points, the cells were harvested,
counted, and aliquots analyzed for CD34antigen expression. Suitable aliquots were assayed in semisolid assays for CFC and on stroma cocultures for LTC-1C
enumeration. Mean SEM of 4 to 6 separate experiments performed in quadruplicate.
3740 PIACIBELLO ET AL
For personal use only. by guest on June 7, 2013. bloodjournal.hematologylibrary.orgFrom
engrafted the totality of mice, 35,000 and 20,000 CD34cells
engrafted 4 of 6 and 3 of 7 mice, respectively. No mouse was
found to be engrafted by 10,000 CD34cells. The level of
engraftment was variable and dependent on the number of
injected CD34cells (human CD45,CD71
, and -GpA
cells ranged from 0.18% to 25%). The level of human engraft-
ment was also evaluated by DNA analysis (as shown in Fig 1).
The frequency of SRC in the unmanipulated cryopreserved
CD34CB cells was found to be 1 in 30,900 CD34cells (95%
confidence interval 1:53,600 to 1:17,800). It was similar to that
of fresh CD34CB cells (1 in 29,800; 95% confidence interval
1:51,700 to 1:17,300). BM cells of engrafted mice were further
analyzed for evidence of multilineage development from input
CD34cells. Cells within the huCD45 gate or costained with
CD45 were quantified for myeloid and lymphoid surface
markers as well as for the expression of the CD34 antigen.
Erythroid and megakaryocyte surface marker expression was
also investigated. Figure 2A and B shows representative
analysis of BM cells from engrafted and nonengrafted mice. As
an additional proof of human myeloid engraftment, BM cells of
sacrificed mice were cultured in semisolid assays in culture
conditions that have been reported to allow human and not
mouse colony growth.8,13 In addition, colonies were stained
with MoAbs recognizing human total leukocytes, granulocyte-
macrophages, erythroid cells, and megakaryocytes and only
fluorescent colonies (consisting of human cells) were counted
(Table 1).
In the second part of our study, CB CD34cells were
cultured in vitro in stroma-free liquid cultures as described.27,28
Cell counts, CD34, CD3438subpopulations, CFC output,
and LTC-IC expansion obtained in the different settings were
compared. At weeks 4, 6, and 8 of expansion cultures, all cells
were harvested from quadruplicate cultures, counted, and
subjected to the same screening (immunophenotype analysis,
CFC output, and LTC-IC enumeration) as described before. All
of the remaining cells (in 7.5 to 127 mL) were washed,
cryopreserved, and stored frozen for further injection into
sublethally irradiated NOD/SCID recipients (one mouse for
each expansion).
Evident from these experiments is the finding that only
marginal differences could be detected in the qualitative and
quantitative content of the three different expansion procedures
(Table 2). After a 4-week expansion in the presence of
FLMGDF, cell counts ranged from 148-fold to 240-fold the
initial number; with SCFFLMGDF, the cell count was
444-fold to 515-fold the input cells; the four-factor combination
Table 3. Antigenic Composition of CD34CB Cells at Initiation of Liquid Cultures and at Various Time Intervals During Ex Vivo Expansion
Culture
Conditions CD34 CD38 CD3438CD33 CD13 CD3313CD14 CD2 CD19 CD41 GpA CD71
Start 91 8.5 94 3.1 0.95 0.05 49 6.4 0 0 0.3 0.03 2.0 1.6 1.8 1.1 0.4 0.2 4.1 1.6 0
wk 4 3.9 0.4 76 4.8 0.18 0.04 96 13 94.7 7.8 91 11.6 59 7.1 0.25 0.04 0.9 0.2 3 1.4 5 222
wk 8 2.1 0.17 27 70.60.15 32 9.6 1.72 16.4 1.7 0.3 25 1.4 0.12 0.01 0.07 0.01 0.5 0.01 0.3 0.01 2.63 1.6
wk 12 2.3 13411 0.46 0.9 85.4 2.4 7.2 4.3 7.1 2.7 36.3 2.1 0.1 0 0.19 0.03 0.18 0.6 0.56 0.04 13.1 2.9
Positive cells (%) of each subpopulation. Mean standard deviation (SD) from four to seven separate experiments performed in replicate wells for each growth factor
combination at different time points.
Table 4. Comparison Between Cell Number, CFC, and LTC-IC Injected and Ability to Engraft the BM and the Spleen of NOD/SCID Mice
by Human CB Cells Cultured in Stroma-Free Cultures for 4 Weeks
CB Cells Growth Factors Added
Injected/Mouse Engraftment
Cells 106CFC 103LTC-IC 103% Human
Cells† Human CFC/Mouse
103
EXP. 1 Start of cultures 0.02 2.2 0.7 4.62 0.36 0.18, 0 0.86, 0
4 weeks of cultures
FL MGDF 9.9 2.8 (495)* 116.2 18.6 (52.8)* 23.42 3.3 (5.06)* 10.4, 12 20.48 0.4, 86.74 1.3
SCF FL MGDF 11.7 3.1 (585)* 79.3 10.2 (36)* 29.97 5.1 (6.48)* 11, 15 ND, 54.71 2.7
IL6 SCF FL MGDF 7.4 1.2 (370)* 111.6 17.8 (50.7)* 23.58 4.2 (5.12)* 21, 6 25.7 4.6, 46.72 3.7
EXP. 2 Start of cultures 0.02 2.46 0.6 2.55 0.4 0, 0.3 0, 1.68
4 weeks of cultures
FL MGDF 4.22 0.77 (211)* 48.48 7.1 (19.7)* 26.55 4.7 (11.8)* 9.8, 0 74.26 6.7, 0
SCF FL MGDF 5.38 0.98 (269)* 59.89 8.9 (24.3)* 25.82 6.1 (11.5)* ND, 21 ND, 59.64 6.4
IL6 SCF FL MGDF 5.6 1.2 (280)* 70.72 10.6 (28.7)* 30.41 4.6 (13.5)* 22.8, 16 ND, 114.71 11.6
EXP. 3 Start of cultures 0.01 0.52 0.5 1.7 0.6 0, 0 0, 0
4 weeks of cultures
FL MGDF 4.16 0.89 (416)* 34.24 7.9 (68.5)* 38.65 5.1 (22.7)* 10.5, 13 ND, 39.42 5.2
SCF FL MGDF 6.9 1.5 (690)* 43.65 6.9 (83.9)* ND 16, 12 43.74 5, 110.71 12.6
IL6 SCF FL MGDF 7.2 2.2 (720)* 42.49 6.3 (81.7)* 41.69 8.8 (24.5)* 14, 0 165.71 13, 0
CD34cells were cultured at 10 103/mLor20103/mL in 24-well plates as described. Cells at the start of cultures and cells harvested after 4
weeks of suspension cultures were cryopreserved and kept frozen until the time of injection in NOD/SCID mice. The expansion cultures were
performed in quadruplicate. Two mice were injected each with 20,000 or 10,000 uncultured cells and two mice each with the content of a single
expansion culture.
*Fold increase (compared with the input value).
†Each value represents the percentage of human CD45, CD71, and -GpAcells detected in the BM of each individual mouse.
‡Mean SEM of the number of human CFU-GM BFU-E CFU-GEMM CFU-Mk generated by plating 1 105to5105BM cells of engrafted
NOD/SCID mice (at least three dishes per point), taking into account that BM cells of femurs and tibias represent 25% of the total BM.
EXPANSION OF IN VIVO REPOPULATING STEM CELLS 3741
For personal use only. by guest on June 7, 2013. bloodjournal.hematologylibrary.orgFrom
appeared also most effective in inducing and maintaining a
sustained cell production (348-fold to 587-fold) and CFC output
(55-fold to 80-fold). Expansion of the LTC-IC population was
induced to a very similar degree by all three growth factor
combinations and in all three culture settings (Table 2).Also, at
weeks 6 and 8 of expansion cultures, cell number, percent of
CD34cells, as well as CFC production and LTC-IC expansion,
were not much different if demidepopulated and expanded wells
or flasks were compared (Table 2). Table 3 shows the proportion
of different hematopoietic subpopulations at start of cultures
and at different time points of expansion. No differences could
be detected in the cellular composition of the cells expanded
with the three growth factor combinations (not shown).
To determine whether the expanded cells retained their
ability of full hematopoietic reconstitution (ie, sustained and
multilineage engraftment) cohorts of 6- to 8-week-old NOD/
SCID mice were irradiated with 350 to 375 cGy from a 137Cs
source and, within 24 hours, injected with either 35,000,
20,000, or 10,000 CD34CB cells, which were cryopreserved
at the start of cultures, or the progeny of identical numbers of
CD34cells that were grown for 4 weeks with FLMGDF,
SCFFLMGDF, IL-6SCFFLMGDF (one mouse for
each expansion). Six to 8 weeks later, BM cells of all mice were
prepared, counted, and subjected to DNA and phenotype
analysis to determine whether or not human cells were present;
which subpopulation was represented and whether human
hematopoietic progenitors (BFU-E, CFU-GM, CFU-GEMM)
could be detected. Table 4 shows the results of three separate
experiments performed in quadruplicate.
In keeping with the results reported in Table 1, only 2 of 6
mice that had been injected with 20,000 or 10,000 cryopre-
served uncultured CD34CB cells were engrafted and the level
of human engraftment was very low. These data were confirmed
by subsequent DNA analysis: the results obtained by injecting
an additional 13 mice with 35,000 and 20,000 unmanipulated
CD34cells are shown in Fig 3. The same figure, Fig 4 and
Table 4 show that, at the opposite side, mice injected with the
cells that had been generated by the same number of initial
CD34CB cells in 4-week expansion cultures supported by
FLMGDF, SCFFLMGDF, and IL6SCFFLMGDF
engrafted the vast majority (15 of 18) of NOD/SCID mice,
although at a variable degree. Human CD45,CD71
, and
GpAcells constituted 6% to 23% of the entire BM. Further
analysis of the engrafted BM cells showed the consistent
presence of human CD45CD19and CD45CD34cells.
CD33,CD41
,CD71
, and GpAcells were also represented
(not shown). Plasma clot assays performed by seeding BM cells
of transplanted NOD/SCID mice showed that human CFU-GM,
BFU-E, CFU-GEMM, and CFU-Mk were indeed present (Table
4). Colonies grown in plasma-clot assays were scored at the
immunofluorescent microscope after being stained with FITC-
conjugated anti-human CD41, CD45, CD13, and -GpA anti-
bodies. All of the colonies were positive, thus indicating their
human origin.
Expansion cultures were then extended for up to 12 weeks.
Flow cytometry analysis of the cells harvested at different time
points did not show substantial differences in the CD34
expression of the cells grown in the presence of the three growth
factor combinations. CD34cells still constituted a good
Fig 3. Engraftment of human CB CD34cells at start of cultures
and of their progeny after 4 to 12 weeks of expansion. The level of
human engraftment was evaluated by flow cytometry by determin-
ing the percent of human CD45, CD71, and GpAcells within the
total BM cells in individual NOD/SCID mice. Each animal was injected
either with unmanipulated CD34CB cells (2 104,;3.5104,)
either with the cells generated by initial 2 104or 3.5 104CD34
cells after 4, 7 to 8, 9 to 10 or 12 weeks of cultures as described in
Materials and Methods. Each circle represents an individual mouse.
Fig 4. Representative Southern blot analysis of individual NOD/
SCID mice transplanted with 1 104,2104, and 3.5 104
unmanipulated CD34CB cells and with week 4 expanded cells
(deriving from 1 104initial CD34cells). DNA was extracted from
the murine BM at week 8 after transplant and hybridized with a
human chromosome 17–specific -satellite probe.
3742 PIACIBELLO ET AL
For personal use only. by guest on June 7, 2013. bloodjournal.hematologylibrary.orgFrom
proportion of the total cell population ( Fig 5). CD34CD38
cells were also detected (Fig 6).
Cells cultured for 6 to 7 weeks engrafted the vast majority of
injected mice (Fig 3). Both DNA analysis and flow cytometry
performed on BM cells of the transplanted mice showed that
human cells were indeed detectable. CD45,CD71
, and
GpAcells accounted for up to 20% of the entire BM after 7 to
8 weeks of liquid cultures; human engraftment was detectable at
fairly good levels (up to 10%) in mice injected with cells
expanded for up to 12 weeks. Further flow cytometry analysis
of the BM cells of the engrafted mice showed that all of the
different hematopoietic lineages were represented (Fig 7A and
B). In addition, human colonies were detected in semisolid
cultures prepared with BM cells of mice transplanted with
expanded CB cells (Table 5). Expansion studies performed
beyond the week 6 cryopreservation comprised only one eighth
of the initial CD34cells; this is why we first inoculated the
expanded 1/8 together with the cryopreserved 7/8 (hence the
engraftment activity might be due completely to the latter).
Therefore, to better define whether weeks 7 to 8 and week 10
expanded cells contained SRC and to quantitate the extent of the
amplification of the in vivo repopulating cells in the expansion
cultures, the 1/8 cells expanded for 7 to 8 and 9 to 10 weeks
were injected into sublethally irradiated NOD/SCID recipients
Fig 5. Analysis of CD34 anti-
gen expression on CB CD34
cells at start of cultures (week 0)
and at weeks 4, 6, 8, and 12 of ex
vivo expansion in stroma-free
culturessupplemented with FL
MGDF and IL6SCFFLMGDF.
EXPANSION OF IN VIVO REPOPULATING STEM CELLS 3743
For personal use only. by guest on June 7, 2013. bloodjournal.hematologylibrary.orgFrom
without further mixing with the remaining 7/8 and were
considered to represent the first dilution. Taking into account
the number of the CD34cells at the start of cultures (50,000,
35,000, 20,000, or 10,000 CD34cells), the number of the
initial cells injected per mouse was calculated (eg, 1/8 of the
cells expanded by 20,000 initial CD34cells represents the
corresponding initial 2,500 CD34cells; 1/16 of initial 20,000
cells represents 1,250 cells, and so on). Shown in Table 5 are the
results of two separate experiments performed in quadruplicate
and injected in replicate mice (two per cell dose). BM cells of
the injected recipients contained human cells. The degree of
human engraftment in mice injected with cells expanded for 7 to
8 weeks ranged between 6.8% and 18%. In particular, BM cells
of engrafted mice when cultured in semisolid assays specific for
human progenitors, generated colonies (CFU-GM, CFU-Mk,
and BFU-E), which were positively stained by human specific
MoAbs. The experiments reported in Table 5 and additional
limiting dilution experiments reported in Fig 8 showed that the
cells generated after 7 to 8 weeks of expansion by initial 1,250,
625, and 312 CD34cells repopulated 9 of 10, 7 of 10, and 8 of
14 mice, respectively.Also, Figs 9 and 10 show that after 9 to 10
weeks of expansion, the cells obtained by initial 1,560 CD34
cells engrafted 7 of 7 mice, those obtained by 625 CD34cells
engrafted 5 of 7 mice, and those deriving from 312 CD34cells
engrafted 7 of 12 mice.
Poisson statistics allowed us to calculate that the frequency of
SRC after 7 to 8 weeks of expansion was 1 in 471 initial CD34
cells (95% confidence interval 1:571 to 1:296), while, after 9 to
10 weeks, it was 1 in 393 initial CD34cells (95% confidence
interval 1:678 to 1:228).
DISCUSSION
The development of SRC assay for primitive human hemato-
poietic cells capable of repopulating the BM of NOD/SCID
immune deficient mice with myeloid and lymphoid lineages
provides a new approach to investigate the organization of the
human hematopoietic system and to characterize primitive stem
cells. The experiments reported here show that CB CD34cells
after a 4- to 10-week expansion in stroma-free liquid cultures
containing the combinations of a few growth factors
(FLMGDFSCFIL-6) retained their capacity to com-
pletely engraft the BM of sublethally irradiated NOD/SCID
Fig 6. Analysis of CD34 and
CD38 antigen expression on
CD34CB cells at start of cultures
(week 0) and after 4 and 8 weeks
of expansion in stroma-free cul-
tures supplemented with FL
MGDF and IL6SCFFLMGDF.
3744 PIACIBELLO ET AL
For personal use only. by guest on June 7, 2013. bloodjournal.hematologylibrary.orgFrom
recipients. The level of engraftment, well above that usually
observed when the same number of uncultured cells were
injected in the same recipients, suggested that SRC were not
only maintained, but, rather, expanded.
This result supports and extends our previous findings that
CD34CB cells could be grown in vitro for an extremely long
period of time, during which a massive and continuously
increasing production of CD34,CD34
CD38cells, commit-
ted unipotent and multipotent progenitors occurred; also primi-
tive stem cells, identified in vitro as LTC-IC were shown to be
amplified more than 200,000-fold after 20 weeks. It was
concluded that the extremely prolonged and impressive expan-
sion of progenitors belonging to all of the hematopoietic
lineages was supported by a similar expansion of primitive
progenitors and that in our system, some degree of self-renewal,
beside differentiation, was taking place.27,28
The culture conditions used in the present study were similar
to those previously reported and yielded similar numbers of
CFC and LTC-IC, attesting that the small-scale demidepopula-
tion assay could be reproduced in a larger scale expansion
Fig 7. (A) Representative FACS profiles of marrow cells from individual NOD/SCID mice transplanted 8 weeks previously with cells deriving
from 3.5 104CD34CB cells after a 10-week expansion in stroma-free cultures containing IL-6, SCF, FL and MGDF. From top to bottom: isotype
control and (a) a nonengrafted mouse. Human CD45/CD34 and CD45/CD19 in the BM cells of a mouse transplanted with 1/8, 1/25, and 1/70 of the
cell progeny deriving from 3.5 104initial CD34cells. CD45/CD34 and CD45/CD19 analysis was performed on total BM cells. (B) Multilineage
engraftment in the BM of a representative mouse transplanted with week 10 expanded cells (deriving from 2 104initial CD34cells). Analysis of
lineage markers (CD45/CD34, CD19, CD41, and CD13/CD33) was performed on cells comprised within the CD45 gate. Analysis of GpA/CD71 cells
was performed on total BM cells.
EXPANSION OF IN VIVO REPOPULATING STEM CELLS 3745
For personal use only. by guest on June 7, 2013. bloodjournal.hematologylibrary.orgFrom
Fig 8. Summary of the level of human cell engraftment in the BM
of 35 mice transplanted with cells deriving from ex vivo expansion
cultures at weeks 7 to 8 in the presence of IL-6, SCF, FL, and MGDF.
Individual NOD/SCID mice (each symbol represents a mouse) were
injected 8 weeks previously with fractions of the expanded cells,
corresponding to numbers of initial CD34 cells indicated on the
abscissa. The level of human engraftment in the mouse BM was
evaluated by both flow cytometry (as percent of human CD45, GpA,
and CD71 positive cells) and DNA analysis as described in Materials
and Methods.
Fig 9. Summary of the level of human cell engraftment in the BM
of 35 mice transplanted 8 weeks previously with cells deriving from
ex vivo expansion cultures at weeks 9 to 10 in the presence of IL-6,
SCF, FL, and MGDF. Individual NOD/SCID mice (each symbol repre-
sents a mouse) were injected with fractions of the expanded cells,
corresponding to numbers of initial CD34cells indicated on the
abscissa. The level of human engraftment in the mouse BM was
evaluated by both flow cytometry and DNA analysis as described in
Materials and Methods.
Table 5. Determination of the Cell Number, CFC, LTC-IC, and the Ability to Engraft the BM of NOD/SCID Recipients by Human CB Cells During
Stroma-Free Cultures
In Culture Injected/Mouse Engraftment
Source
of Cells Growth Factors
Added Cells
(fold increase) CFC
(fold increase) LTC-IC
(fold increase) Cells CFC LTC-IC % of Human
Cells* Human CFC/
mouse 103
Start 10,000 866.4 1,332 10,000 866.4 1,332 0, 0 0, 0
5,000 433 666 0, 0 0, 0
2,500 216 333 0, 0 0, 0
1,250 108 166 0, 0 0, 0
4 weeks IL-6 SCF
FL MGDF 3.16 106
(316) 37,184
(43) 11,248
(8.4) 3.160 106
0.395 10637,184
4,648 11,248
1,456 8.5, 9.9
2.5, 3 15.64, 18.4
7.2, 9.6
8 weeks IL-6 SCF
FL MGDF 28.99 106
(2,899) 129,464
(149) 56,304
(42) 28.99 106
3.60 106129,464
16,183 56,304
7,038 7.3, 18
2.8, 3.6 18.22, 36.72
ND, 34.32
1.80 1068,091 3,519 0.7, 1.9 1.34, 3.87
12 weeks IL-6 SCF
FL MGDF 109 106
(10,900) 2,909,061
(3,358) 382,284
(287) 109 106
13.6 106
363,632
47,785 ——
11, 16 34.98, 50.76
6.8 106181,816 23,892 8, 6.8 23.44, 44.66
3.4 10690,908 11,946 1.4, 0.9 12.6, 19.3
Start 20,000 2,248 3,632 20,000 3,248 3,632 0, 0 2.08, 0
5,000 812 908 0, 0 0, 0
2,500 406 454 0, 0 0, 0
1,250 203 227 0, 0 0, 0
7 weeks FL MGDF 22 106
(1,100) 372,736
(166) 90,112
(25) 22 106
2.75 106124,930
15,616 90,112
11,264 6.8, 15
4, 11 29.9, 63.84
8.3, 75.9
1.37 1067,808 5,632 0, 2.6 0, 13.4
SCF FL
MGDF 36.4 106
(1,820) 381,728
(170) 126,976
(35) 36.40 106
4.55 106381,728
47,716 126,976
15,872 7.8, 14.2
5.6, 9 41.87, 46.86
2.08, ND
2.27 10623,858 7,936 0.9, 3 2.54, 10.1
IL-6 SCF
FL MGDF 17.8 106
(890) 361,081
(160) 102,400
(28) 17.80 106
2.22 106361,081
45,135 102,400
12,800 18, 10.4
4, 6 108.9, 54.16
ND, 23.9
1.11 10622,567 6,400 1, 2.1 6.32, 15.6
12 weeks IL-6 SCF
FL MGDF 710 106
(35,500) 2,928,640
(1,303) 980,460
(270)
88.75 106
366,080
122,580 ——
ND, ND ——
ND, ND
22.20 10691,520 30,641 11, 6.5 50.16, 30.07
11.10 10645,760 15,322 0.6, 3.9 1.87, 6
5.50 10622,880 7,661 0.9, 0.5 5.56, 2.3
The value in parentheses represents the fold increase compared with input values.
*Each value represents the percent of human CD45, CD71, and -GPAcells in the BM of each individual mouse.
†As explained in Table 4. Expansion studies were performed in quadruplicate. Two mice were injected with each cell dose as indicated.
3746 PIACIBELLO ET AL
For personal use only. by guest on June 7, 2013. bloodjournal.hematologylibrary.orgFrom
setting without losing its initial quality. In addition, in the
cultures presented here, 10% human serum was substituting for
10% FCS, therefore a further step toward a clinical application
has been secured.
In the first part of our study when decreasing numbers of
uncultured CB CD34cells were injected into sublethally
irradiated NOD/SCID recipients, less than 35,000 CD34cells
did not engraft the totality of the recipients. Using Poisson
statistics,33 the frequency of SRC in the cryopreserved CD34
population was calculated to be 1 in 30,900, while in fresh
CD34cells, it was 1 in 29,800, which appears in keeping with
that reported by others.14,16,33 Four to 10 weeks of culturing
CD34CB cells in stroma-free cultures led to an increase in the
repopulating capacity of the cultured hematopoietic cells.
Injection of the cultured cells deriving from 35,000, 20,000, or
10,000 initial CD34cells resulted in a significant increase in
the level of human engraftment, similar to that observed by
injecting at least 100,000 uncultured CD34cells.
Limiting dilution experiments performed by injecting NOD/
SCID mice with decreasing concentrations of cells expanded
for up to 10 weeks allowed us to show that the frequency of
SRCs after 7 to 8 weeks of expansion was 1 in 471 and after 9 to
10 weeks, it was 1 in 391 input CD34cells, therefore the
expansion of SRC was 65-fold and 78-fold, respectively,
60-fold and 70-fold if compared with the frequency of SRC in
fresh CD34CB cells. Recently, it has been reported that
incubation of CD34CD38CB cells in serum-free medium
containing FL, SCF, IL-3, IL-6, and G-CSF for 5 to 8 days,
resulted in a 100-fold expansion of CFC, a 4-fold expansion of
LTC-IC, and a modest (2-fold), but significant, increase of
CRU. CRU were found, although at different frequencies, also
in the CD34CD38CB subpopulations and with a distribu-
tion between the two CB subsets as LTC-IC. It was concluded
that conditions, which were able to stimulate LTC-IC expan-
sion, might also stimulate increases in CRU.16 Similarly, Bhatia
et al29 showed a fourfold increase of SRC after a 4-day
incubation of CD34CD38CB cells in stroma-free condi-
tions; SRC, however, were lost after 9 days of culture. Gan et
al30 reported that cocultures of BM or CB cells on allogeneic
stroma layers for up to 3 weeks resulted in a decrease and a final
loss of in vivo repopulating ability in NOD/SCID mice recipi-
ents; in contrast to the loss of SRC, the cultured cells frequently
contained an equal or higher number of LTC-IC compared with
the fresh cells. Additionally, while this report was being revised,
it was reported that human CD34CD38BM cells, after 6-day
stroma-free suspension cultures containing FL, TPO, and SCF
retained their in vivo repopulating capacity in the SCID/hu bone
assay.34 Also, Xu et al31 recently showed that CB cells could
maintain their in vivo repopulating ability after at least 4 weeks
of coculture on a stromal cell line derived from the aorta-gonad-
mesonephros region of mouse embryo.
Cell populations, culture conditions (stromal cocultures or
stroma-free cultures), as well as the growth factors used in the
various studies are different, therefore the somewhat different
results cannot be compared. However, it is becoming increas-
ingly evident that primitive stem cells, defined by their ability to
completely engraft a myeloablated recipient, can be maintained
in vitro for long periods of time and are likely to undergo
self-renewal divisions.
A very controversial issue is the role played by stroma
cocultures: even though the presence of BM stroma layers has
been shown to increase the frequency of gene transfer into
primitive cells, compared with using viral supernatants and to
prevent the loss of stem cell quality during ex vivo expansion of
peripheral blood stem cells,35-37 other reports suggest that the
long-term repopulating ability of cultured cells decreases using
BM stroma cocultures.38,39
The vast majority of prior studies aimed at developing
clinical applications of expansion protocols have adopted
culture conditions that resulted in a marked expansion of cell
counts, CD34cells, CFC, and even LTC-IC in a short period of
time. However, the expansion was transient, soon followed by a
rapid decline of cell number, of CFC output, and disappearance
of LTC-IC, which indicated the exhaustion of the stem cell pool.
In our culture system, it is possible to obtain very large numbers
of cells and progenitors belonging to the more mature hemato-
poietic compartments and, at the same time, to maintain and
even expand several-fold the primitive in vivo repopulating
stem cells. This information could prove essential to design and
test conditions for ex vivo activation and expansion of imma-
ture hematopoietic cells and for various experimental purposes,
such as required for the development of efficient gene transfer
protocols into hematopoietic cells with retention of repopulat-
ing ability.
REFERENCES
1. Spangrude G, Smith L, Uchida N, Ikuta F, Heimfeld S, Friedman
J, Weissman IL: Mouse hematopoietic stem cells. Blood 78:1395, 1991
Fig 10. Representative Southern blot analysis of individual NOD/
SCID mice transplanted with expanded cells from replicate flasks at
week 10 of expansion. Quadruplicate cultures were initiated with 2
104CD34CB cells. After 10 weeks of expansion, 2 mice were injected
with the corresponding progeny of initial CD34cells (1 mouse per
expansion) lanes 8 to 9; 2 mice with 1/12 of the expanded cells
(corresponding to initial 1,560 CD34cells)lanes6to7;2micewith
1/32 (625 initial cells) lanes 4 to 5; and 2 mice with 1/64 (312 initial
cells) lanes 2 to 3. Lane 1: A nonengrafted mouse (inoculated with 2
104CD34cells at start of cultures, plus 2 105CD34irradiated CB
cells).
EXPANSION OF IN VIVO REPOPULATING STEM CELLS 3747
For personal use only. by guest on June 7, 2013. bloodjournal.hematologylibrary.orgFrom
2. Ogawa M: Differentiation and proliferation of hematopoietic stem
cells. Blood 81:2844, 1993
3. Berardi AC, Wang A, Levine JD, Lopez P, Scadden DT: Func-
tional isolation and characterization of human hematopoietic stem cells.
Science 267:104, 1995
4. To LB, Haylock DN, Simmons PJ, Juttner CA: The biology and
clinical uses of blood stem cells. Blood 89:2233, 1997
5. Sutherland HJ, Lansdorp PM, Henkelman DH, Eaves AC, Eaves
CJ: Functional characterization of individual human hematopoietic
cells cultured at limiting dilution on supportive marrow stromal layers.
Proc Natl Acad Sci USA87:3584, 1990
6. Hao QL, Shah AJ, Thienmann FT, Smogorzewska EM, Crooks
GM: Afunctional comparison of CD34CD38cells in cord blood and
bone marrow. Blood 86:3745, 1995
7. Kamel-Reid S, Dick JE: Engraftment of immune-deficient mice
with human hematopoietic stem cells. Science 242:1706, 1988
8. Lapidot T, Pflumio F, Doedens M, Murdoch B, Williams DE, Dick
JE: Cytokine stimulation of multilineage hematopoiesis from immature
human cells engrafted into SCID mice. Science 255:1137, 1992
9. Kyoizumi S, Baum CM, Kaneshima H, McCune JM, Yee EJ,
Namikawa R: Implantation and maintenance of functional human bone
marrow in SCID-hu mice. Blood 79:1704, 1992
10. Nolta JA, Hanley MB, Kohn DB: Sustained human hematopoi-
esis in immunodeficient mice by cotransplantation of marrow stroma
expressing human interleukin-3: Analysis of gene transduction of
long-lived progenitors. Blood 83:3041, 1994
11. Lowry PA, Shultz LD, Greiner DL, Hesselton RM, Kittler ELW,
Tiarks CY, Rao SS, Reilly J, Leif JH, Ramshaw H, Stewart FM,
Quesenberry PJ: Improved engraftment of human cord blood cells in
NOD/LtSz-scid/scid mice after irradiation or multiple-day injections
into unirradiated recipients. Biol Blood Marrow Transplant 2:15, 1996
12. Pflumio F, Izac B, Katz A, Shultz LD, Vainchenker W, Coulom-
bel L: Phenotype and function of human hematopoietic cells engrafting
immune-deficient CB17-severe combined immunodeficiency mice and
nonobese diabetic-severe combined immunodeficiency mice after trans-
plantation of human cord blood mononuclear cells. Blood 88:3731,
1996
13. Cashman JD, Lapidot T, Wang JCY, Doedens M, Shultz LD,
Lansdorp P, Dick JE, Eaves CJ: Kinetic evidence of the regeneration of
multi-lineage hematopoiesis from primitive cells in normal human bone
marrow transplanted into immunodeficient mice. Blood 89:4307, 1997
14. Hogan CJ, Shpall EJ, McNulty O, McNiece I, Dick JE, Shultz
LD, Keller G: Engraftment and development of human CD34-
enriched cells from umbilical cord blood in NOD/LtSz-scid/scid Mice.
Blood 90:85, 1997
15. Dick JE: Normal and leukemic human stem cells assayed in
SCID mice. Semin Immunol 8:197, 1996
16. Conneally E, Cashman J, Petzer A, Eaves C: Expansion in vitro
of transplantable human cord blood stem cells demonstrated using a
quantitative assay of their lympho-myeloid repopulating activity in
nonobese diabetic-scid/scid mice. Proc Natl Acad Sci USA:94:9836,
1997
17. Larochelle A, Vormoor J, Hanenberg H, Wang JCY, Bhatia M,
Lapidot T, MoritzT, Murdoch B, Xiao XL, Kato I, Williams DA, Dick
JE: Identification of primitive human hematopoietic cells capable of
repopulating NOD/SCID mouse bone marrow: Implications for gene
therapy. Nat Med 2:1329, 1996
18. Bhatia M, Wang JCY, Kapp U, Bonnet D, Dick JE: Purification
of primitive human hematopoietic cells capable of repopulating immune-
deficient mice. Proc Natl Acad Sci USA94:5320, 1997
19. Cashman J, Bockhold K, Hogge DE, Eaves AC, Eaves CJ:
Sustained proliferation, multi-lineage differentiation and maintenance
of primitive human hemopoietic cells in NOD/SCID mice transplanted
with human cord blood. Br J Haematol 97:1026, 1997
20. Gluckman E, Broxmeyer HE, Auerbach AD, Friedman H,
Douglas G, Devergie A, Esperou H, Thierry D, Socie G, Lehn P,
Cooper S, English D, Kurtzberg J, Bard J, Boyse E: Hematopoietic
reconstitution in a patient with Fanconi’s anemia by means of umbilical
cord blood from an HLA-identical sibling. N Engl J Med 321:1174,
1989
21. Broxmeyer HE, Gluckman E, Auerbach AD, Douglas GW,
Friedman H, Cooper S, Hangoc G, Kurtzberg J, Bard J, Boyse EA:
Human umbilical cord blood: A clinically useful source of transplant-
able hematopoietic stem/progenitor cells. Int J Cell Cloning 8:76, 1990
22. Hows JM, Bradley BA, Marsh JCW, Luft T, Coutinho L, Testa
NG, Dexter TM: Growth of umbilical cord blood in longterm haemopoi-
etic cultures. Lancet 340:73, 1992
23. Lu L, Xiao M, Shen R-N, Grigsby S, Broxmeyer HE: Enrich-
ment, characterization and responsiveness of single primitive
CD34⫹⫹⫹ human umbilical cord blood hematopoietic progenitors
with high proliferative and replating potential. Blood 81:41, 1993
24. Cairo MS, Wagner JE: Placental and/or umbilical cord blood:An
alternative source of hematopoietic stem cells for transplantation. Blood
90:4665, 1997
25. Petzer AL, Hogge DE, Lansdorp PM, Reid DS, Eaves CJ:
Self-renewal of primitive human hematopoietic cells (long-term-culture-
initiating cells) in vitro and their expansion in defined medium. Proc
Natl Acad Sci USA93:1470, 1996
26. Petzer AL, Zandstra PW, Piret JM, Eaves CJ: Differential
cytokine effects on primitive (CD34CD38-) human hematopoietic
cells: Novel responses to Flt3-ligand and thrombopoietin. J Exp Med
183:2551, 1996
27. Piacibello W, Sanavio F, Garetto L, Severino A, Bergandi D,
Ferrario J, Fagioli F, Berger M, Aglietta M: Extensive amplification and
self-renewal of human primitive hematopoietic stem cells from cord
blood. Blood 89:2644, 1997
28. Piacibello W, Sanavio F, Garetto L, Severino A, Dane`A,
Gammaitoni L, Aglietta M: Differential growth factor requirement of
primitive cord blood hematopoietic stem cell for self-renewal and
amplification vs proliferation and differentiation. Leukemia 12:718,
1998
29. Bhatia M, Bonnet D, Kapp U, Wang CY, Murdoch B, Dick JE:
Quantitative analysis reveals expansion of human hematopoietic repopu-
lating cells after short-term ex vivo culture. J Exp Med 186:619, 1997
30. Gan OI, Murdoch B, Larochelle A, Dick JE: Differential
maintenance of primitive human SCID-repopulating cells, clonogenic
progenitors, and long-term culture-initiating cells after incubation on
human bone marrow stromal cells. Blood 90:641, 1997
31. Xu M, Tsuji K, Ueda T, Mukouyama Y, HaraT,Yang F, Ebihara
Y, Matsuoka S, Manabe A, Kikuchi A, Ito M, Miyajima A, NakahataT:
Stimulation of mouse and human primitive hematopoiesis by murine
embryonic aorta-gonad-mesonephros-derived stromal cell lines. Blood
92:3032, 1998
32. Taswell C: Limiting dilution assays for the determination of
immunocompetent cell frequencies: I. Data analysis. J Immunol 26:
1614, 1981
33. Wang JCY, Doedens M, Dick JE: Primitive human hematopoi-
etic cells are enriched in cord blood compared to adult bone marrow or
mobilized peripheral blood as measured by the quantitative in vivo
SCID-repopulating cell (SRC) assay. Blood 89:3924, 1997
34. Luens KM, Travis MA, Chen BP, Hill BL, Scollay R, Murray LJ:
Thrombopoietin, kit ligand, flk2/flt3 ligand together induce increased
numbers of primitive hematopoietic progenitors from human CD34
Thy-1Lin- cells with preserved ability to engraft SCID-hu bone.
Blood 91:1206, 1998
35. Moore KA, Deisseroth AB, Reading CL, Williams DE, Belmont
3748 PIACIBELLO ET AL
For personal use only. by guest on June 7, 2013. bloodjournal.hematologylibrary.orgFrom
JW: Stromal support enhances cell-free retroviral vector transduction of
human bone marrow long-term culture-initiating cells. Blood 79:1393,
1992
36. Nolta JA, Smogorzewska EM, Kohn DB: Analysis of optimal
conditions for retroviral-mediated transduction of primitive human
hematopoietic cells. Blood 86:101, 1995
37. Breems DA, Blokland EAW, Siebel KE, Mayen AEM, Engels
LJA, Ploemacher RE: Stroma-contact prevents loss of hematopoietic-
stem cell quality during ex vivo expansion of CD34mobilized
peripheral blood stem cells. Blood 91:111, 1998
38. Harrison DE, Lerner CP, Spooncer E: Erythropoietic repopulat-
ing ability of stem cells from long-term marrow culture. Blood 69:1021,
1997
39. Van der Sluijs JP, van den Bos C, Baert MRM, van Beurden CAJ,
Ploemacher RE: Loss of long-term repopulating ability in long-term
bone marrow culture. Leukemia 7:725, 1993
EXPANSION OF IN VIVO REPOPULATING STEM CELLS 3749
For personal use only. by guest on June 7, 2013. bloodjournal.hematologylibrary.orgFrom
... A number of methods to increase the UCB unit cell dose have shown success. These include ex vivo expansion of UCB cells [3][4][5][6] and the use of double UCB transplants. 2,7,8 Bone marrow (BM), peripheral blood stem cell, and UCB grafts are usually injected IV, homing to the BM space after several hours in the circulation. ...
Article
Full-text available
Intrabone (IB) injection of umbilical cord blood has been proposed as a potential mechanism to improve transplant engraftment and prevent graft failure. However, conventional IB techniques produce low retention of transplanted cells in the marrow. To overcome this barrier, we developed an optimized IB (OIB) injection method using low-volume, computer-controlled slow infusion that promotes cellular retention in the marrow. Here, we compare engraftment of CD34+ cells transplanted in a myeloablative rhesus macaque (RM) model using the OIB method compared with IV delivery. RM CD34+ cells obtained by apheresis were split equally for transduction with lentiviral vectors encoding either green fluorescent protein or yellow fluorescent protein reporters. Following conditioning, one marked autologous population of CD34+ cells was injected directly IB using the OIB method and the other was injected via slow IV push into the same animal (n = 3). Daily flow cytometry of blood quantified the proportion of engrafting cells deriving from each source. Marrow retention was examined using positron emission tomography/computed tomography imaging of 89Zirconium (89Zr)-oxine–labeled CD34+ cells. CD34+ cells injected via the OIB method were retained in the marrow and engrafted in all 3 animals. However, OIB-transplanted progenitor cells did not engraft any faster than those delivered IV and contributed significantly less to hematopoiesis than IV-delivered cells at all time points. Rigorous testing of our OIB delivery system in a competitive RM myeloablative transplant model showed no engraftment advantage over conventional IV infusion. Given the increased complexity and potential risks of IB vs IV approaches, our data do not support IB transplantation as a strategy to improve hematopoietic engraftment.
... During the past few years, an accumulation of data has demonstrated that the Mpl receptor and its ligand, TPO, which were first described to be involved in the primary regulation of megakaryopoiesis and thrombopoiesis, also played an important role at the level of adult hematopoietic stem cells. 11,12,[36][37][38][39][40][41] This concept has been supported by the effects of inactivation of the Mpl/TPO pathway. 16,17 c-mpl / adult mice are not only deficient in hematopoietic progenitors, but also demonstrate a dramatic decrease in hematopoietic stem cells. ...
Article
The Mpl receptor plays an important role at the level of adult hematopoietic stem cells, but little is known of its function in embryonic and fetal hematopoiesis. We investigated the signals sent by the MPL cytoplasmic domain in fetal liver hematopoietic progenitors and during embryonic stem (ES) cell hematopoietic commitment. Mpl was found to be expressed only from day 6 of ES cell differentiation into embryoid bodies. Therefore, we expressed Mpl in undifferentiated ES cells or in fetal progenitors and studied the effects on hematopoietic differentiation. To avoid the inadvertent effect of thrombopoietin, we used a chimeric receptor, PM-R, composed of the extracellular domain of the prolactin receptor (PRL-R) and the transmembrane and cytoplasmic domains of Mpl. This allowed activation of the receptor with a hormone that is not involved in hematopoietic differentiation and assessment of the specificity of responses to Mpl by comparing PM-R with another PRL-R chimeric receptor that includes the cytoplasmic domain of the erythropoietin receptor (EPO-R) ([PE-R]). We have shown that the cytoplasmic domain of the Mpl receptor transduces exclusive signals in fetal liver hematopoietic progenitors as compared with that of EPO-R and that it promotes hematopoietic commitment of ES cells. Our findings demonstrate for the first time the specific role of Mpl in early embryonic or fetal hematopoietic progenitors and stem cells.
... The SRC frequency was calculated from the proportions of negative mice in each cohort, using the L-CALC™ software (STEMCELL™ Technologies, Vancouver, BC, Canada), which employed Poisson statistics and the method of maximum likelihood. After log transformation, the ratio of frequencies was computed as reported earlier [38][39][40]. ...
Article
Full-text available
Background: Hematopoietic stem cell (HSC) transplantation has been widely applied to the treatment of malignant blood diseases. However, limited number of functional HSCs hinders successful transplantation. The purpose of our current study is to develop a new and cost-efficient medium formulation that could greatly enhance the expansion of HSCs while retaining their long-term repopulation and hematopoietic properties for effective clinical transplantation. Methods: Enriched human CD34+ cells and mobilized nonhuman primate peripheral blood CD34+ cells were expanded with a new, cost-efficient expansion medium formulation, named hematopoietic expansion medium (HEM), consisting of various cytokines and nutritional supplements. The long-term repopulation potential and hematologic-lineage differentiation ability of expanded human cells were studied in the non-obese diabetic/severe combined immunodeficiency mouse model. Furthermore, the efficacy and safety studies were performed by autologous transplantation of expanded primate cells in the nonhuman primate model. Results: HEM could effectively expand human CD34+ cells by up to 129 fold within 9 days. Expanded HSCs retained long-term repopulation potential and hematologic-lineage differentiation ability, as indicated by (1) maintenance (over unexpanded HSCs) of immunophenotypes of CD38-CD90+CD45RA-CD49f+ in CD34+ cells after expansion; (2) significant presence of multiple human hematopoietic lineages in mouse peripheral blood and bone marrow following primary transplantation; (3) enrichment (over unexpanded HSCs) in SCID-repopulating cell frequency measured by limiting dilution analysis; and (4) preservation of both myeloid and lymphoid potential among human leukocytes from mouse bone marrow in week 24 after primary transplantation or secondary transplantation. Moreover, the results of autologous transplantation in nonhuman primates demonstrated that HEM-expanded CD34+ cells could enhance hematological recovery after myelo-suppression. All primates transplanted with the expanded autologous CD34+ cells survived for over 18 months without any noticeable abnormalities. Conclusions: Together, these findings demonstrate promising potential for the utility of HEM to improve expansion of HSCs for clinical application.
Article
Full-text available
The application of umbilical cord blood (UCB) as an important source of hematopoietic stem and progenitor cells (HSPCs) for hematopoietic reconstitution in the clinical context has steadily grown worldwide in the past 30 years. UCB has advantages that include rapid availability of donors, less strict HLA-matching demands, and low rates of graft-versus-host disease (GVHD) versus bone marrow (BM) and mobilized peripheral blood (PB). However, the limited number of HSPCs within a single UCB unit often leads to delayed hematopoietic engraftment, increased risk of transplant-related infection and mortality, and proneness to graft failure, thus hindering wide clinical application. Many strategies have been developed to improve UCB engraftment, most of which are based on 2 approaches: increasing the HSPC number ex vivo before transplantation and enhancing HSPC homing to the recipient BM niche after transplantation. Recently, several methods have shown promising progress in UCB engraftment improvement. Here, we review the current situations of UCB manipulation in preclinical and clinical settings and discuss challenges and future directions.
Article
Thymic repopulation by transplanted hematopoietic progenitor cells (HPC) is likely to be important for long-term immune reconstitution and for successful gene therapy of diseases affecting the T-cell lineage. However, the T-cell progenitor potential of HPC, cultured in vitro for cell number expansion and gene transfer remains largely unknown. Here, we cultured highly purified human umbilical cord blood (CB) CD34+CD38− or CD34+CD38+ cells for up to 5 weeks in stroma-free cultures supplemented with various combinations of the cytokines thrombopoietin (TPO), stem cell factor (SCF), flt3/flk-2 ligand (FL), interleukin-3 (IL-3), and IL-6 and investigated thymus-repopulating ability of expanded cells in vitro and in vivo. After up to 5 weeks of culture in IL-3 + SCF + IL-6 or TPO + FL + SCF supplemented medium, the progeny of CD34+CD38− CB cells generated T cells and natural killer cells in the thymus. Limiting dilution experiments demonstrated increase in the number of T-cell progenitors during culture. After 3 weeks of culture, gene marked CD34+CD38− CB cells injected in the human thymus fragment transplanted in severe combined immunodeficient (SCID) mice (SCID-hu) generated thymocytes expressing the retroviral encoded marker gene GFP in vivo. Thus, our results show that the progeny of CD34+CD38− CB cells cultured for extensive periods, harbor thymus-repopulating cells that retain T-cell progenitor potential after expansion and gene transfer.
Article
The marrow repopulating potential (MRP) of different sources of human hematopoietic stem cells (HSCs) was directly compared using an in vivo assay in which severe combined immunodeficient disease (SCID) mice were implanted with human fetal bones. HSCs from 2 human lymphocyte antigen (HLA)-mismatched donors were injected individually or simultaneously into the fetal bones of a 3rd distinct HLA type and donor and recipient myeloid and lymphoid cells were identified after 8 to 10 weeks. The study compared the MRP of umbilical cord blood (CB) and adult bone marrow (ABM) CD34+ cells as well as grafts of each type expanded ex vivo. Equal numbers of CB and ABM CD34+ cells injected individually demonstrated similar abilities to establish multilineage hematopoiesis. However, when CB and ABM cells were transplanted simultaneously, the engraftment of CB cells was markedly superior to ABM. CB and ABM CD34+ cells were expanded ex vivo using either a porcine microvascular endothelial cell (PMVEC)-based coculture system or a stroma-free expansion system. Primary CB CD34+ cells or CD34+ cells expanded in either culture system demonstrated a similar ability to engraft. However, the MRP of expanded grafts simultaneously injected with primary CB cells was uniformly inferior to primary CB cells. CD34+ cell grafts expanded in the stroma-free system, furthermore, outcompeted CD34+ cells expanded using the PMVEC coculture system. The triple HLA-mismatched SCID-hu model represents a novel in vivo stem cell assay system that permits the direct demonstration of the functional consequences of ex vivo HSC expansion and ontogeny-related differences in HSCs.
Article
Human SCID repopulating cells (SRC) are defined based on their functional ability to repopulate the bone marrow of NOD/SCID mice with both myeloid and lymphoid cell populations. The frequency of SRC in umbilical cord blood cells is 1 in 9.3 × 105mononuclear cells. We report that as few as 8 × 104 human cord blood mononuclear cells transplanted into NOD/SCID/B2mnull mice resulted in mutlilineage differentiation in the murine bone marrow, revealing a more than 11-fold higher SRC frequency than in NOD/SCID mice. Moreover, as few as 2 to 5 × 103 CD34+ cells recovered from the bone marrow of primary transplanted NOD/SCID mice were sufficient for engrafting secondary NOD/SCID/B2mnull mice with SRC, suggesting SRC self-renewal. Thus, by using NOD/SCID/B2mnull mice as recipients, we established a functional assay for human stem cells capable of engrafting the bone marrow of primary and secondary transplanted immune-deficient mice with SRC, providing a model that better resembles autologous stem cell transplantation.
Article
The low frequency of transplantable hematopoietic stem cells in adult human bone marrow (BM) and other differences from cord blood stem cells have impeded studies to optimize the retroviral transduction of stem cells from adult sources. To address this problem, first a cytokine combination was defined that would both maximize the kinetics of adult BM CD34+CD38− cell mitogenesis and minimize the period of prestimulation required for the transduction of these cells by a MSCV-GFP/neor virus in tissue culture dishes in the absence of fibronectin. Three days of stimulation with flt3-ligand, Steel factor, interleukin (IL)-3, and hyper-IL-6 proved both necessary and sufficient to obtain 83% ± 2% GFP+ CD34+CD38− cells, 75% ± 10% G418-resistant clonogenic progenitors, and 50% ± 20% transduced long-term culture-initiating cells as recovered 48 hours after a single exposure to virus. Moreover, this was accompanied by a several-fold increase in viral receptor (pit-1) messenger RNA transcripts in the target cells. Using this prestimulation protocol, repeated daily exposure to new virus (3×) did not alter the proportion of transduced cells over that obtained with a single exposure. Adult human BM cells able to engraft immunodeficient (NOD/SCID-β2M−/−) mice were also efficiently transduced (10%-20% GFP+ human lymphoid and myeloid cells present 6-8 weeks after transplant) using a 6-day prestimulation and infection protocol. A clinically useful efficiency of retrovirus-mediated gene transfer to transplantable adult human BM stem cells can thus be obtained with a protocol that allows their semisynchronous activation into cycle and concomitant increased expression of virus receptor transcripts before virus exposure.
Article
Recent experiments point to the great value of lentiviral vectors for the transduction of human hematopoietic stem cells (hHSCs). Vectors used so far, however, have been poorly satisfying in terms of either biosafety or efficiency of transgene expression. Herein is described the results obtained with human immunodeficiency virus–based vectors optimized in both of these aspects. It is thus shown that vectors containing the EF1α and, to a lesser extent, the phosphoglycerate kinase (PGK) promoter, govern high-level gene expression in human hematopoietic progenitors as well as derived hematopoietic lineages of therapeutic relevance, such as erythrocytes, granulocytes, monocytes, dendritic cells, and megakaryocytes. EF1α promoter-containing lentiviral vectors can also induce strong transgene expression in primary T lymphocytes isolated from peripheral blood. A self-inactivating design did not affect the performance of EF1α promoter-based vectors but significantly reduced expression from the PGK promoter. This negative effect could nevertheless be largely rescued by inserting the post-transcriptional regulatory element of woodchuck hepatitis virus upstream of the vector 3′ long terminal repeat. These results have important practical implications for the genetic treatment of lymphohematologic disorders as well as for the study of hematopoiesis via the lentivector-mediated modification of hHSCs.
Article
An understanding of mechanisms regulating hematopoietic stem cell engraftment is of pivotal importance to the clinical use of cultured and genetically modified transplants. Human cord blood (CB) cells with lymphomyeloid repopulating activity in NOD/SCID mice were recently shown to undergo multiple self-renewal divisions within 6 days in serum-free cultures containing Flt3-ligand, Steel factor, interleukin 3 (IL-3), IL-6, and granulocyte colony-stimulating factor. The present study shows that, on the fifth day, the transplantable stem cell activity is restricted to the G1fraction, even though both colony-forming cells (CFCs) and long-term culture-initiating cells (LTC-ICs) in the same cultures are approximately equally distributed between G0/G1and S/G2/M. Interestingly, the G0 cells defined by their low levels of Hoechst 33342 and Pyronin Y staining, and reduced Ki67 and cyclin D expression (representing 21% of the cultured CB population) include some mature erythroid CFCs but very few primitive CFCs, LTC-ICs, or repopulating cells. Although these findings suggest a cell cycle–associated change in in vivo stem cell homing, the cultured G0/G1 and S/G2/M CD34+ CB cells exhibited no differences in levels of expression of VLA-4, VLA-5, or CXCR-4. Moreover, further incubation of these cells for 1 day in the presence of a concentration of transforming growth factor β1 that increased the G0/G1 fraction did not enhance detection of repopulating cells. The demonstration of a cell cycle–associated mechanism that selectively silences the transplantability of proliferating human hematopoietic stem cells poses both challenges and opportunities for the future improvement of ex vivo–manipulated grafts.
Article
Full-text available
Many experimental and clinical protocols are being developed that involve ex vivo culture of human hematopoietic cells on stroma or in the presence of cytokines. However, the effect of these manipulations on primitive hematopoietic cells is not known. Our severe combined immune-deficient mouse (SCID)-repopulating cell (SRC) assay detects primitive human hematopoietic cells based on their ability to repopulate the bone marrow (BM) of immune-deficient non-obese diabetic/SCID (NOD/SCID) mice. We have examined here the maintenance of SRC, colony-forming cells (CFC), and long-term culture-initiating cells (LTC-IC) during coculture of adult human BM or umbilical cord blood (CB) cells with allogeneic human stroma. Transplantation of cultured cells in equivalent doses as fresh cells resulted in lower levels of human cell engraftment after 1 and 2 weeks of culture for BM and CB, respectively. Similar results were obtained using CD34+-enriched CB cells. By limiting dilution analysis, the frequency of SRC in BM declined sixfold after 1 week of culture. In contrast to the loss of SRC as measured by reduced repopulating capacity, the transplanted inocula of cultured cells frequently contained equal or higher numbers of CFC and LTC-IC compared with the inocula of fresh cells. The differential maintenance of CFC/LTC-IC and SRC suggests that SRC are biologically distinct from the majority of these in vitro progenitors. This report demonstrates the importance of the SRC assay in the development of ex vivo conditions that will allow maintenance of primitive human hematopoietic cells with repopulating capacity.
Article
Full-text available
Human fetal bone fragments implanted in the immunodeficient C.B-17 scid/scid (SCID) mouse were shown to sustain active human hematopoiesis in vivo. Human progenitor cell activity was maintained for as long as 20 weeks after implantation and was associated with multilineage differentiation in the engrafted bone. Thus, the bone implants provided stem cells as well as the microenvironment requisite for their long- term maintenance and multilineage differentiation. Administration of human erythropoietin (Epo) stimulated human erythropoiesis in human bone implants. This animal model may facilitate direct analysis of a wide variety of physiologic and pathologic conditions of human bone marrow (BM) in vivo.
Article
Full-text available
The clinical manifestations of Fanconi's anemia, an autosomal recessive disorder, include progressive pancytopenia, a predisposition to neoplasia, and nonhematopoietic developmental anomalies [1-3]. Hypersensitivity to the clastogenic effect of DNA-cross-linking agents such as diepoxybutane acts as a diagnostic indicator of the genotype of Fanconi's anemia, both prenatally and postnatally [3-6]. Prenatal HLA typing has made it possible to ascertain whether a fetus is HLA-identical to an affected sibling [7].We report here on hematopoietic reconstitution in a boy with severe Fanconi's anemia who received cryo-preserved umbilical-cord blood from a sister shown by prenatal testing to be unaffected by the disorder, to have a normal karyotype, and to be HLA-identical to the patient. We used a pretransplantation conditioning procedure developed specifically for the treatment of such patients [8]; this technique makes use of the hypersensitivity of the abnormal cells to alkylating agents that cross-link DNA [9,10] and to irradiation [11] In this case, the availability of cord blood obviated the need for obtaining bone marrow from the infant sibling.This use of cord blood followed the suggestion of one of us that blood retrieved from umbilical cord at delivery, usually discarded, might restore hematopoiesis – a proposal supported by preparatory studies by some of us [12] and consistent with reports on the presence of hematopoietic stem and multipotential (CFU-GEMM), erythroid (BFU-E), and granulocyte-macrophage (CFU-GM) progenitor cells in human umbilical-cord blood (see the references cited by Broxmeyer et al. [12]).
Article
We report here on a novel stromal cell line, AGM-S3, derived from the aorta-gonad-mesonephros (AGM) region of a 10.5 days postcoitum (dpc) mouse embryo. The AGM-S3 cells promoted production of hematopoietic progenitors and day-12 spleen colony-forming cells from Lin−c-Kit+Sca-1+ murine primitive hematopoietic cells. They also supported for 6 weeks generation of human multipotential progenitors from cord blood CD34+CD38− primitive hematopoietic cells. Human long-term repopulating hematopoietic stem cells (LTR-HSC) with the potential to reconstitute hematopoiesis in NOD/SCID mice were maintained on AGM-S3 cells for at least 4 weeks. Flow cytometric analysis showed that CD13, vascular cellular adhesion molecule-1, and Sca-1 were expressed on AGM-S3 cells. Because stem cell factor, interleukin-6 (IL-6), and oncostatin M, but not IL-3, IL-11, leukemia- inhibitory factor, granulocyte colony-stimulating factor, granulocyte-macrophage colony-stimulating factor, thrombopoietin, and Flk2 ligand were detected in reverse transcription-polymerase chain reaction analysis of AGM-S3 cells, the cells seem to express species-cross reactive molecule(s) other than the cytokines examined and which act on primitive hematopoietic progenitor/stem cells. This cell line is expected to elucidate molecular mechanisms regulating early hematopoiesis and pave the way for developing strategies for expansion of human transplantable HSC. © 1998 by The American Society of Hematology.
Article
Available evidence indicates that qualitative changes in hematopoietic stem cells and progenitors, such as the decision of stem cells to self- renew or differentiate, or selection of lineage potentials by the multipotential progenitors during differentiation (commitment), are intrinsic properties of the progenitors and are stochastic in nature. In-contrast, proliferative kinetics of the progenitors, namely survival and expansion of the progenitors, appear to be controlled by a number of interacting cytokines. While proliferation and maturation of committed progenitors is controlled by late-acting lineage-specific factors such as Ep, M-CSF, G-CSF, and IL-5, progenitors at earlier stages of development are controlled by a group of several overlapping cytokines. IL-3, GM-CSF, and IL-4 regulate proliferation of multipotential progenitors only after they exit from G0 and begin active cell proliferation. Triggering of cycling by dormant primitive progenitors and maintenance of B-cell potential of the primitive progenitors appears to require interactions of early acting cytokines including IL-6, G-CSF, IL-11, IL-12, LIF, and SF. Currently, this simple model fits our understanding of the interactions of growth factors with hematopoietic progenitors. Naturally the model risks oversimplification of a very complex process. However, because the model is testable, it will hopefully challenge investigators to design new experiments to examine its validity.
Article
To characterize the growth of cord blood progenitor cells, single nonadherent, low-density, T-lymphocyte-depleted CD34 cells were sorted by flow cytometer with an autoclone device into single wells containing culture medium and cytokines. These cells were evaluated for proliferation and for replating ability of their progeny. This latter effect is used as a measure of self-renewal capacity. Colony formation was assessed in 1 degree wells containing various cytokines, alone and in combination, and single colonies deriving after 21 days in semisolid medium were replated into 2 degree wells in the presence of the combination of purified preparations of recombinant human steel factor (SF, a c-kit ligand), granulocyte-macrophage colony-stimulating factor (GM-CSF), granulocyte colony-stimulating factor (G-CSF), interleukin-3 (IL-3), and erythropoietin (Epo). Replating of single colonies was performed also for 3 degrees, 4 degrees, and 5 degrees cultures. In the presence of serum, colony formation was observed in > 66% of the wells stimulated with the combination of Epo, SF, GM-CSF, G-CSF, and IL-3, and more than 39% of the colonies formed in these 1 degree wells were very large in size (> 2.5 mm in diameter, dense in the center, and containing > 10(4) cells/colony). The replating efficiency of these large colonies was up to 93% with generation of subsequent colonies of very large size. Replating could be shown for up to five generations. The cells in these colonies were large, nonspecific esterase positive, and contained large amounts of cytoplasm with one or more nuclei containing several nucleoli per nucleus. Smaller colonies (1 to 2.5 mm in diameter and dense in the center) containing similar cells and making up an additional 14% of the colonies formed in 1 degree wells also showed extensive replating capacity, including generation of larger colonies. These colony-forming cells are likely similar to the murine macrophage high-proliferative potential colony-forming cells. The cells giving rise to these colonies are present in about eightfold higher frequency in cord blood than in adult bone marrow. These cells may at least in part be associated with the successful hematopoietic repopulating capacity of umbilical cord blood cells.
Article
Hemopoietic precursors are heterogeneous with respect to their capacity for self-renewal and long-term repopulating ability. Bone marrow cultures produce a variety of precursors over many weeks, including CFU- S; however, it is important to determine whether these populations retain the functional ability shown by fresh marrow. The most primitive precursor or stem cells have the most long-term repopulating ability. We here describe direct measurements of this ability in cells from marrow cultures by using competitive repopulation assays. Cultured adherent cells repeatedly showed less capacity than fresh marrow cells to repopulate erythropoiesis in irradiated recipients, whereas cultured suspension cells consistently had less capacity than adherent cells. Concentrations of macroscopic CFU-S measured at nine or 12 days were similar in cultured adherent and suspension cells and generally lower than those in fresh marrow. In every experiment, the long-term repopulating ability of the marrow cells used was substantially reduced after transfer into tissue culture. Thus, primitive stem cells may not proliferate in such cultures despite extensive production of CFU-S and more differentiated cell types.
Article
Available evidence indicates that qualitative changes in hematopoietic stem cells and progenitors, such as the decision of stem cells to self- renew or differentiate, or selection of lineage potentials by the multipotential progenitors during differentiation (commitment), are intrinsic properties of the progenitors and are stochastic in nature. In-contrast, proliferative kinetics of the progenitors, namely survival and expansion of the progenitors, appear to be controlled by a number of interacting cytokines. While proliferation and maturation of committed progenitors is controlled by late-acting lineage-specific factors such as Ep, M-CSF, G-CSF, and IL-5, progenitors at earlier stages of development are controlled by a group of several overlapping cytokines. IL-3, GM-CSF, and IL-4 regulate proliferation of multipotential progenitors only after they exit from G0 and begin active cell proliferation. Triggering of cycling by dormant primitive progenitors and maintenance of B-cell potential of the primitive progenitors appears to require interactions of early acting cytokines including IL-6, G-CSF, IL-11, IL-12, LIF, and SF. Currently, this simple model fits our understanding of the interactions of growth factors with hematopoietic progenitors. Naturally the model risks oversimplification of a very complex process. However, because the model is testable, it will hopefully challenge investigators to design new experiments to examine its validity.
Article
Cryopreserved human umbilical-cord (HUC) blood is an alternative to bone marrow as a source of haemopoietic "stem" cells for HLA-identical transplantation of children with leukaemia or Fanconi's anaemia. We have studied the in-vitro growth potential of HUC blood in clonogenic assays and in longterm haemopoietic cultures. Clonogenic assays showed that HUC blood produced as many haemopoietic-cell colonies as normal adult bone marrow and a higher proportion of primitive-cell colonies. In longterm culture on preformed irradiated marrow stroma, both progenitor-cell production and lifespan of cultures were significantly greater in HUC blood than in normal bone marrow (p = 0.0007). Our findings indicate that the quality and quantity of HUC-blood-derived haemopoietic "stem" cells are better than those of normal bone marrow. Therefore, single HUC-blood donations are probably sufficient for adults requiring transplantation for leukaemia and other haemopoietic disorders. Banking of HLA-typed HUC blood to facilitate transplantation of patients who lack a family donor should be considered.