Download Bone marrow stem cell transplantation for cardiac repair - AJP

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Lymphopoiesis wikipedia , lookup

Transcript
Am J Physiol Heart Circ Physiol 288: H2557–H2567, 2005;
doi:10.1152/ajpheart.01215.2004.
Invited Review
Bone marrow stem cell transplantation for cardiac repair
Husnain Kh Haider and Muhammad Ashraf
Department of Pathology and Laboratory of Medicine, University of Cincinnati, Cincinnati, Ohio
myocardial infarction; cardiomyocyte; plasticity; transdifferentiation
in both small and large animal models
and phase 1 clinical studies in humans have shown the promise
of bone marrow and bone marrow-derived stem cells in the
treatment of diseased myocardium (111). The secret of this
promise lies in the remarkable potential of bone marrow stem
cells to undergo multilineage differentiation in general and
cardiomyogenic potential in particular (66, 108).
Bone marrow consists of a nonhomogeneous population of
cells composed of various lineages with differing abilities. The
hematopoietic stem cell (HSC) fraction of the bone marrow is
responsible for the production of ⬎95% of the body’s blood
cells. They have an extensive capacity of self-renewal. Recent
reports (27, 65) have shown that HSC may also have the
potential to differentiate into other lineages of cells, including
their competence to attain the cardiac phenotype. The work of
Matsuzaki and colleagues (81) suggests high-marrow seeding
efficiency as a specific characteristic of primitive HSC in
addition to their self-renewal and multipotent capacity. They
have demonstrated a high “homing to niche” capacity of these
cells. Over a long period of time, CD34, a cell surface molecule that is expressed on 1–3% of bone marrow cells, has been
used as a convenient positive selection marker for HSC (94).
However, several reports have raised serious questions about
CD34 expression on hematopoietic cells in the mouse (36).
Sato and colleagues (112) have shown that CD34 expression in
murine HSC reflects the activation state, and this may be so in
humans. Moreover, the presence of human CD34⫺ HSC has
been reported as being more immature than CD34⫹ stem cells
(15). Consequently, AC133 is being used as an alternative
selection marker to CD34 antigen. The rationale to use purified
THE RESULTS OF STUDIES
AC133⫹ cells is to avoid injection of a large number of
leukocytes and their progenitors, which have limited plasticity
and the presence of which in large numbers may give rise to an
unwanted inflammatory response at the site of the graft (152).
On the other hand, the non-HSC fraction is mainly composed of mesenchymal stem cells. They are clonogenic and
possess the potential to develop into mature cells that produce
fat, cartilage, bone, tendons, and muscle (108). They can be
isolated from bone marrow and can easily be expanded in an in
vitro cell culture without compromising their stem cell capabilities (86). Because of the ease of the method of their
purification and multipotent nature, they have been extensively
studied in both experimental and clinical settings. There is no
universal surface marker for the identification of mesenchymal
stem cells. They are negative for HSC markers CD31, CD34,
and CD45 and express on their surface CD44, CD90, CD105,
CD106, and CD166 (78). Besides, they have been reported to
express T cell receptor components that enable them to remain
in a state of readiness to enter various pathways of differentiation upon induction (158). Mesenchymal stem cells with
similar potential have also been isolated from tissues other than
bone marrow including peripheral blood and umbilical cord
blood (61). Kawada et al. (59) have recently shown that
CD34⫺ nonhematopoietic bone marrow mesenchymal stem
cells can differentiate into cardiomyocytes. The heterogeneous
mononuclear fraction of the bone marrow (BMMNC) is the
most promising and extensively studied stem cell population
for cellular cardiomyoplasty (24, 47, 54, 56). Besides, CD117
and Sca-1⫹ cells have also been focused to assess their cardiomyogenic potential (72, 73, 76, 80, 152).
MALLEABLE NATURE OF BONE MARROW STEM CELLS
Address for reprint requests and other correspondence: H. Kh Haider, Dept.
of Pathology and Laboratory of Medicine, 231-Albert Sabinway, Univ. of
Cincinnati, Cinncinati, OH 45267-0529 (E-mail: [email protected]).
http://www.ajpheart.org
Intravenous administration of bone marrow-derived mesenchymal stem cells leads to their preferential migration to the
0363-6135/05 $8.00 Copyright © 2005 the American Physiological Society
H2557
Downloaded from http://ajpheart.physiology.org/ by 10.220.32.247 on April 29, 2017
Haider, Kh Husnain, and Muhammad Ashraf. Bone marrow stem cell
transplantation for cardiac repair. Am J Physiol Heart Circ Physiol 288: H2557–
H2567, 2005; doi:10.1152/ajpheart.01215.2004.—Cardiomyocytes respond to
physiological or pathological stress only by hypertrophy and not by an increase in
the number of functioning cardiomyocytes. However, recent evidence suggests that
adult cardiomyocytes have the ability, albeit limited, to divide to compensate for
the cardiomyocyte loss in the event of myocardial injury. Similarly, the presence of
stem cells in the myocardium is a good omen. Their activation to participate in the
repair process is, however, hindered by some as-yet-undetermined biological
impediments. The rationale behind the use of adult stem cell transplantation is to
supplement the inadequacies of the intrinsic repair mechanism of the heart and
compensate for the cardiomyocyte loss in the event of injury. Various cell types
including embryonic, fetal, and adult cardiomyocytes, smooth muscle cells, and
stable cell lines have been used to augment the declining cardiomyocyte number
and cardiac function. More recently, the focus has been shifted to the use of
autologous skeletal myoblasts and bone marrow-derived stem cells. This review is
a synopsis of some interesting aspects of the fast-emerging field of bone marrowderived stem cell therapy for cardiac repair.
Invited Review
H2558
BONE MARROW STEM CELLS FOR HEART
mechanisms, the consensus is on transdifferentiation of bone
marrow cells into cardiomyocytes and endothelial cell phenotypes and angiogenesis (Tables 1–3).
The ability of bone marrow-derived stem cells to break the
barriers of lineage restriction and transdifferentiate to form
different tissue cells are being extensively investigated (6, 37,
46, 53, 133, 135). A series of recent experiments have challenged the long-standing dogma of lineage restriction. The
multipotential nature of bone marrow stem cells to transdifferentiate into various lineages has been shown in vitro (18, 21,
98, 137, 148, 151). Similar results have also been corroborated
in vivo (9, 69, 95, 157). The underlying principle to these
observations is that stem cells form the tissue in which they
reside. Hence, it is thought that bone marrow stem cells under
the influence of the new microenvironment after transplantation start to adopt the new phenotype and produce cells of the
resident tissue (39, 52). However, these findings have been
challenged by several research groups that have independently
advocated the possibility of fusion between donor bone marrow cells with host cells to adopt the phenotype of the host
tissue (134, 153). Recent publications from Alvarez and colleagues (5) have shown fusion between donor bone marrow
cells and Purkinje fibers, cardiomyocytes, and hepatocytes.
Thus there is a prevailing notion that fusion between donor
stem cells and host tissue cells might be misleading the
researchers to conclude that stem cells can cross the boundaries
of lineage restriction.
Back-to-back recent reports from Balsam et al. (11) and Murry
et al. (91) have clearly depicted a lack of ability of donor bone
marrow cells to undergo differentiation into cardiomyocytes after
transplantation. They have shown little evidence for transplanted
bone marrow cells to form cardiomyocytes at the site of the graft
posttransplantation. Similar concern has been shown by earlier
researchers when isolated unfractionated bone marrow cells were
found to be unable to differentiate into cardiomyocytes (13).
These results are presenting a serious challenge to the findings that
have been documented by Orlic and colleagues (101) and thus
require an in-depth assessment of whether bone marrow stem cells
have the required plasticity to transdifferentiate into cardiomyocytes posttransplantation.
CARDIOMYOGENIC TRANSDIFFERENTIATION OF BONE
MARROW STEM CELLS IN VITRO
Cardiomyogenic differentiation of stem cells has been vastly
reported (60, 102, 138). The cells undergoing cardiomyogenic
Table 1. In vitro studies on BMC transdifferentiation into cardiovascular-related lineages
References
Cells
Makino et al. (77)
Fukuda et al. (32)
Fuchs et al. (30)
Toma et al. (137)
Terada et al. (134)
Eisenberg et al. (27)
Fukuhara et al. (33)
Oswald et al. (103)
BMSC
BMSC
BMC
hBMMSC
BMMNC
HPC
BMSC
hBMMSC
Coculture
Pretreatment
No
ESC
Chick embryo heart tissue
Cardiomyocytes
5-Aza
5-Aza
Yes
MyoD transduction
IL-3 for 7 days
RA and ECGS PGE2 and IL-2
VEGF
Differentiation
Myocytes
Myocytes
Myocytes
Myocytes
Myocytes
Myocytes
Endothelial cells
BMC, bone marrow cells; BMSC, bone marrow stromal cells; 5-Aza, 5-azacytidine; hBMMSC, human bone marrow mesenchymal stem cells; BMMNC, bone
marrow mononuclear cells; ESC, embryonic stem cells; IL, interleukin; HPC, hemapoietic progenitor cells; RA retinoic acid; ECGS, endothelial cell growth
supplement.
AJP-Heart Circ Physiol • VOL
288 • JUNE 2005 •
www.ajpheart.org
Downloaded from http://ajpheart.physiology.org/ by 10.220.32.247 on April 29, 2017
infracted myocardium (92), and tissue injury is considered as
one important factor that influences the transdifferentiation of
bone marrow cells to other lineages (1). BMMNCs taken from
myocardial infarction patients have been found to show tropism for damaged cardiac tissue (10a). Similarly, there is a
significant debate over the presence of stem cells from extracardiac origin in the myocardium (12, 110). Despite these
interesting findings, the underlying mechanism as to how the
donor bone marrow stem cells home into the damaged myocardium and contribute toward improved cardiac function remains highly debatable (97, 118). However, it is considered to
be the resultant effect of more than one mechanism. First, bone
marrow cells undergo differentiation into cardiomyocytes, either due to preprogramming or under the influence of signals
from myocardial microenvironment (64, 100, 121, 140). Second, they contribute toward improved wall thickness and a
reduction in the infarct size. Bone marrow stem cells have been
shown to express angiogenic growth factors in a paracrine way
to stimulate neovascularization at the site of the cell graft and
undergo transdifferentiation into endothelial cells to participate
in angiogenesis, thus alleviating myocardial ischemia (42, 132,
139). The paracrine effect may not be the effect of individual
cytokines themselves. Rather, this may be related with their
synergistic interaction that may lead to the beneficial outcome
in terms of neovascularization. Kinnaird and colleagues (62)
have reported a full spectrum of arteriogenic cytokines released
from bone marrow stromal cells. The expression of these
growth factors gets accentuated under stressful hypoxic conditions (8). Mesenchymal stem cells grown under hypoxic
conditions migrated and formed three-dimensional capillarylike structures on a Matrigel matrix. They also showed these
effects when triggered by conditioned media taken from mesenchymal stem cells grown under hypoxic stress. These observations clearly depict the involvement of both paracrine as well
as autocrine mechanisms. The role of paracrine signaling
pathways as a mediator of bone marrow cell therapy has also
been highlighted in vivo in the treatment of tissue ischemia,
which clearly depicts that donor cell incorporation into vessels
is not a prerequisite for their effects in angiogenesis (63).
An interesting study from Pak et al. (104) showed that bone
marrow cell transplantation induces cardiac nerve sprouting
and sympathetic hyperinnervation. Another proposed mechanism is the fusion of donor bone marrow cells with host
cardiomyocytes to adopt their phenotype (123). The donor cell
genes may supplement for the lost genetic material from the
host nucleus during the ischemic injury. Out of all these
Invited Review
H2559
BONE MARROW STEM CELLS FOR HEART
Table 2. Small animal model studies used for BMC transplantation for cardiac repair
Model
Rat
Tomita et al. (138)
Wang et al. (143)
Wang et al. (144)
Davani et al. (24)
Cells
Pretreatment
Angiogenesis
BMC
BMSC
BMSC
BMMPC
5-Aza
No
No
No
CAL
CAL
CAL
Doxorubicin
CAL
CAL
BM-MSC
BMSC
BMC ⫹ myoblasts
BMMNC
BMMSC
BMMNC
HGF transduction
5-Aza
No
No
No
No
CAL
CAL
Lin⫺ ckit⫹
SP
No
Yes
Yes
Saito et al. (114)
CAL
BMSC
No
Yes
Toma et al. (137)
Agbulut et al. (3)
Kudo et al. (68)
Hiasa et al. (47)
Murry et al. (91)
Balsam et al. (11)
Normal
Doxorubicin
CAL
CAL
Normal/CAL
CAL
hBMMSC
BMC
Lin⫺ ckit⫹
BMMNC
Lin⫺ ckit⫹
Lin⫺ ckit⫹ and Lin⫺
ckit⫹ Thy1.1loSca⫹
No
No
No
No
No
No
Yes
No
No
No
Duan et al. (26)
Saito et al. (115)
Sakaghuchi et al. (116)
Ishida et al. (54)
Tang et al. (132)
Zhang et al. (156)
Mouse
Orlic et al. (102)
Jackson et al. (55)
Yes
Yes
Yes
Yes
Yes
Yes
Yes
Myocytes
Myocytes
Myocytes
Smooth muscle and
endothelial
phenotype
Myocyte-like cells
Myocytes
Endothelial cells
Endothelial cells
Myocytes
Myocytes and
endothelial cells
Vascular smooth
muscle cells and
myocytes
Myocytes
Myocytes
Myocytes
No
No
No
CAL, coronary artery ligation; HGF, hepatocyte growth factor; SP, side population.
plemented with retinoic acid, DMSO, and 5-azacytidine (5-Aza)
(45). Cardiomyogenic differentiation of bone marrow in vitro has
specifically been achieved using the demethylating agent 5-Aza
(Table 1). Makino et al. (77) reported 5-Aza-induced differentiation of mice bone marrow stromal cells into cardiac-like cells. At
a concentration of 3 ␮mol/l for 1 wk, 5-Aza induced bone marrow
cells into cardiomyogenic cells. These cells stained positive for
myosin, actin, and desmin and showed spontaneous beating at 3
wk after treatment. Electron microscopy revealed a cardiomyocyte-like ultrastructure, such as the presence of sarcomeres, centrally positioned nuclei, and atrial granules.
Coculture of bone marrow stem cells with cardiomyocytes
has been shown to preprogram these cells to adopt a cardiac
phenotype. Our research group (147) has recently shown that
differentiation achieve the cardiomyocyte phenotype through
the expression of specific genes encoding various transcription
factors and structural and regulatory proteins (105). Among
these are included GATA-4, Nkx 2.5, MEF2, and HAND.
However, the exact role of these genes, the timing of their
activation, and the signaling pathways involved in mediating
cardiomyogenic differentiation remain unknown. Besides
these, a recent report (41) has shown that they also express
functional adrenergic and muscrinic receptors that modulate
their function.
Various strategies have been adopted for directed differentiation of stem cells into cardiomyocytes (45). The induction of
cardiomyogenic differentiation of stem cells has been achieved by
culturing bone marrow cells in vitro using culture medium supTable 3. Large animal studies using BMC for cardiac repair
Pig
Kamihata et al. (56)
Fuchs et al. (30)
Shake et al. (121)
Tomita et al. (139)
Min et al. (84)
Pak et al. (104)
Yau et al. (150)
Dog
Hamano et al. (42)
Sheep
Bel et al. (13)
Airey et al. (4)
Model
Cells
Pretreatment
Angiogenesis
LAD ligation
Ameroid ring
60-min LAD occlusion
LAD occlusion
LAD branch ligation
LAD microcoil
embolization
Coil occlusion
Differentiation
BMMNC
BMC
BMMSC
BMSC
hMSC⫹ hFC
BMMSC
No
No
No
5-Aza
No
No
Yes
Yes
Yes
Yes
Yes
Yes
Yes
Yes
Nerve sprouting
BMSC
No
Yes
Yes
LAD ligation
Fresh BMC
No
Yes
Yes
LCx ligation
In utero injection
Fresh BMC
hMSC
No
No
Yes
No
Pukinje fibers
LAD, left anterior descending coronary artery; hMSC, human mesenchymal stem cells; hFC, human fetal cardiomyocytes; LCx, left circumflex coronary
artery.
AJP-Heart Circ Physiol • VOL
288 • JUNE 2005 •
www.ajpheart.org
Downloaded from http://ajpheart.physiology.org/ by 10.220.32.247 on April 29, 2017
Cryoinjury
Normal heart
CAL
CAL
Differentiation
Invited Review
H2560
BONE MARROW STEM CELLS FOR HEART
cell-to-cell contact during coculture of bone marrow cells with
cardiomyocytes is imperative to trigger their cardiomyogenic
transformation.
TRANSDIFFERENTIATION OF BONE MARROW STEM CELLS
IN VIVO
AJP-Heart Circ Physiol • VOL
CLINICAL STUDIES
Since the first human autologous skeletal myoblasts transplantation (82), more research groups have been involved in
phase 1 safety and feasibility studies using adult stem cell
transplantation for cardiac repair, including bone marrow cells.
The underlying principle is to take advantage of the plastic
nature of bone marrow stem cells, which enables them to cross
the lineage restriction and participate in the reparative process
by attaining the cardiac phenotype.
Whereas some of these studies have been carried out as the
sole-therapy procedure using a bone marrow cell implantation
approach (10a, 14, 31, 106, 141), others have been performed
as an adjunct to either coronary artery bypass grafting (34, 109,
126, 127, 154) or percutaneous revascularization (10, 19, 22,
125, 129, 145). The small number of the patients in each study
(without a blinded control group) and their performance as an
adjunct procedure make it difficult to assess and attribute the
288 • JUNE 2005 •
www.ajpheart.org
Downloaded from http://ajpheart.physiology.org/ by 10.220.32.247 on April 29, 2017
Encouraged by the results from in vitro studies, both small
and large animal studies have been carried out to show that the
whole bone marrow and its subpopulations potentially repopulate the injured myocardium and undergo milieu-dependent
differentiation to form endothelial cells and cells with a cardiomyocyte-like phenotype (Tables 2 and 3). It has been
hypothesized that preprogramming donor bone marrow cells to
adopt a cardiomyogenic differentiation pathway before their
transplantation in animal models may have better chances of
their transdifferentiation into cardiomyocytes than transplantation of uncommitted bone marrow cells (16, 138). The heart
function of the 5-Aza-treated bone marrow cell transplanted
group was better than that of other groups. Liechty et al. (74)
demonstrated in vivo site-specific differentiation of human
bone marrow into cardiomyocytes. The cells were directly
injected into the fetal sheep peritoneal cavity. Lin⫺ ckit⫹ bone
marrow cells developed into fetal and neonatal myocytes in
infarcted mice hearts (101). They were positive for cardiac
myosin, GATA-4, MEF2, and Csx/Nkx 2.5. The cells were
also positive for connexin43, indicating intercellular communication between the grafted cells and host tissue. A highly
enriched hematopoietic CD34⫺ “side population” of bone
marrow cells was transplanted into lethally irradiated C57BL/
6-Ly-5.1 mice (55). The engrafted side population bone marrow cells differentiated into cardiomyocytes and endothelial
cells. The derived cardiomyocytes were found primarily in the
peri-infarct region. These studies suggested that signals originating from the cytokine-rich microenvironment of the injured
myocardium and the direct cell-cell interaction between donor
bone marrow cells and host cardiomyocytes triggered their
myogenic differentiation. The injured heart is a more efficient
inducer of bone marrow cells differentiation into cardiomyocytes (64). Shintani et al. (124) demonstrated that endothelial
progenitor cells were mobilized into the peripheral blood and
contributed to neovascularization in the heart during an acute
infarction event in humans. These studies suggested that inflammatory cytokines and factors released by the infarcted
heart into the peripheral blood may have the potential to trigger
circulating bone marrow cells to differentiate into specific cell
lineages. Condorelli et al. (23) observed that the majority of
endothelial cells injected into infarcted mice differentiated into
myosin-positive cells with a cardiomyocytes-like morphology,
whereas only a very few of them differentiated into cardiomyocytes after injection into the normal mice heart. Our research
group (68) has previously shown that transplantation of bone
marrow-derived Lin⫺ ckit⫹ cells significantly reduced infarction and fibrosis in an ischemic mouse heart. The donor bone
marrow cells differentiated into cardiomyocytes and endothelial cells in the ischemically damaged murine heart. Transplantation of bone marrow stromal cells has also been shown to
improve the functioning of the healed infarcted rat heart
through myogenesis and angiogenesis (99). The angiogenic
potential of bone marrow cells posttransplantation may be
enhanced by subjecting the cells to ischemic stress before
transplantation (75). In recent study (117), granulocyte colonystimulating factor (G-CSF)-mobilized human bone marrow
CD34⫹ and CD34⫺ cells were delivered intravenously into an
infracted nude rat heart model of myocardial infarction. The
cells were found to migrate and repopulate the ischemic myocardium and induce neovascularization. There was a resultantly improved cardiac function and reduced remodeling.
To achieve controlled delivery of the transplanted bone
marrow cells, Liu et al. (75) used a novel fibrin patch-based
delivery of cells. A fibrin patch coated with autologous bone
marrow cells was placed on the infarct site. The results revealed that the transplanted cells differentiated into cardiomyocyte-like cells and participated in the angiogenic process
and became part of the capillary network.
Taken together, these data suggest that the injured heart
might have a greater contribution to the milieu-dependent
cardiomyocyte differentiation of bone marrow cells. However,
the information on the signaling molecules and responding
molecular mechanisms that recruit bone marrow cells to the
injured heart and transdifferentiation into cardiomyocytes is
still unclear. Myocardial infarction can mobilize bone marrow
cells and recruit them to injured myocardium where they get
stimulated for transformation into new cardiomyocytes.
One important consideration, however, is that the plastic and
malleable nature of unselected multipotent bone marrow may
be disadvantageous and results in unwarranted and unwanted
outcomes. The presence of cells with osteogenic potential or a
multipotent stem cell population together with local milieu of
the cytokine-rich infarcted myocardium may favor osteogenic
differentiation of bone marrow cells and enhance calcium
deposition with an as-yet-undefined regulatory mechanism. It
has been shown that transplantation of unselected bone marrow
cells into the acutely infarcted myocardium may cause significant intramyocardial calcification, thus leading to contractile
dysfunction or arrhythmias (155). The calcification in the bone
marrow cell-transplanted hearts was identified in the periinfarct area or normal myocardium, where the myocardial
structure was relatively well preserved. Similarly, transplantation of undifferentiated mesenchymal stem cells has been
shown to develop into fibroblastic scar tissue (144). These
results underscore the importance of regulating cellular differentiation of adult stem cells in therapeutic applications.
Invited Review
H2561
BONE MARROW STEM CELLS FOR HEART
Table 4. Randomized clinical studies with control groups using BMC
Patients
Study
Control
With cells
Cell type
Assumus (10)
09
20
BMMNC
Chen (22)
35
34
BMMNC
Wollert (145)
30
30
CD34⫹
Composition
7.35 ⫾ 7.31 ⫻ 106
CD34/CD45-positive
cells
Delivery
Mode
Adjunct
Procedure
IC/CPC
PCI
IC
PCI
IC
PCI
24.6 ⫾ 9.4 ⫻ 108
nucleated cells, 9.5 ⫾
6.3 ⫻ 106 CD34⫹
cells, and 3.6 ⫾ 3.4 ⫻
106 hemopoietic
colony-forming cells
Result Summary
Improved regional wall motion in the
infarct area, enhanced LVEF,
profoundly reduced LVESV,
enhanced viability in the infarcted
one by DS-echocardiography, and
quantitative PET scan
Improved wall velocity in the
infracted segments, reduction in
perfusion defects, improved LVEF,
and significantly reduced LVESV
by echocardiography, NOGA
EMM, and PET scanning
Improved LVEF 24-h Holter
monitoring and functional MRI
IC, intracoronary; CPC, circulating progenitor cells; PCI, percutaneous coronary intervention; LVEF, left ventricular ejection fraction; LVESV, left ventricular
end-systolic volume; DS, dobutamine stress; PET, positron emission tomography; NOGA, EMM, electromechanical mapping system.
AJP-Heart Circ Physiol • VOL
288 • JUNE 2005 •
www.ajpheart.org
Downloaded from http://ajpheart.physiology.org/ by 10.220.32.247 on April 29, 2017
sure (129). The control group patients received 6 ml of normal
saline solution without cells using the same method. The
patients underwent [18F]fluorodeoxyglucose PET scanning and
echocardiography on the day of cell transplantation and were
followed up at 3 and 6 mo. The follow-up results were
encouraging and showed significant differences between the
two groups of patients. The percentage of hypokinetic, akinetic, and dyskinetic areas in the left myocardium significantly
decreased (from 32 ⫾ 5% to 13 ⫾ 11%), whereas wall motion
velocity (from 2.17 ⫾ 1.3 to 4.2 ⫾ 2.5 cm/s) and left ventricular ejection fraction (LVEF; from 49 ⫾ 9% to 67 ⫾ 11%)
registered significant increases in the cell transplanted patients
together with significantly decreased perfusion defects as detected by PET. The most encouraging finding of the study was
that the indicators of improvement in cardiac performance
were maintained unruffled until 6 mo of observation.
The Bone Marrow Transfer to Enhance S-T Elevation Infarct Regeneration study, a recent randomized controlled clinical trial involving 60 patients, was reported by Wollert and
colleagues (145). The patients were randomized to receive
optimum medical treatment and PCI together with autologous
bone marrow cell transplantation (n ⫽ 30) or without cell
transplantation as the control group (n ⫽ 30). A total of 24.6 ⫻
108 freshly isolated CD34⫹ cells were transplanted by intracoronary infusion using a balloon catheter in the infarct related
artery. Mean global LVEF showed a 7% increase in the stem
cell transfer group compared with 0.7% in the medical therapy
group (P ⫽ 0.0026). Study limitations included a lack of
sham-operated control subjects and an inability to determine
the mechanisms of action of bone marrow cells.
Zeiher’s group, in Germany, has documented preliminary
results of the Transfer of Progenitor Cells and Regeneration
Enhancement in Acute Myocardial Infarction study. The randomized study, which previously reported 20 patients, included
8 more patients to the cohort (10, 19). Interestingly, the study
involves a comparison between BMMNC and circulating progenitor cells (CPC) in blood for cardiac repair together with an
internal reference group of 11 patients who were included as a
real benefit and improvement in cardiac function to bone
marrow cell transplantation. Invariably, all the studies have
used autologous bone marrow cell transplantation. However,
they differ in the processing, culture, and selection of the
subpopulation before transplantation. For example, unlike
Galinanes et al. (34), who used unmanipulated autologous bone
marrow (34), and Tse et al. (141), who used unselected
BMMNC for transplantation, others have tried to purify a
subpopulation of BMMNC (106, 126, 129, 145). NOGA has
been used for electromechanical mapping to discriminate between the ischemic and viable regions and for targeted delivery
of cells (106, 141). The methods for the end-point measurement have been variable and include treadmill exercise duration, consumption of nitroglycerine, single photon emission
computed tomography (SPECT) perfusion imaging, electromechanical mapping system (NOGA, EMM), echocardiography,
[18F]fluorodeoxyglucose positron emission tomography (PET),
and magnetic resonance imaging. In most cases, the cell
transplantation procedure was uneventful, and the patients
recovered well without any postprocedural complications. No
arrhythmic changes have been reported on Holter monitoring
during any study. Only two deaths, one each in the experimental and control groups, were reported by Perin et al. (106).
However, both these deaths were unrelated to the cell transplantation procedure. Despite their observational status and
design limitations, their outcome reveals the safety and feasibility of the approach. Taken together, the results of the phase
1 studies have been encouraging. Table 4 summarizes the bone
marrow cell transplantation reports with the largest number of
patients published in refereed journals.
The largest study, with 78 patients, was reported by Chen et
al. (22). The patients were randomized to receive bone marrow
cells (n ⫽ 34) and only percutaneous coronary intervention
(n ⫽ 35) as the control group. After being cultured for 10 days
in vitro, 8 –10 ⫻ 109 cells/ml of autologous bone marrow cells
were resuspended in 6 ml of heparinized saline solution and
transfused into the infarct related target coronary artery
through an inflated over-the-balloon catheter with high pres-
Invited Review
H2562
BONE MARROW STEM CELLS FOR HEART
BONE MARROW STEM CELL MOBILIZATION
BMMNC taken from myocardial infarction patients have
been found to show tropism for damaged cardiac tissue in vitro
(28). Moreover, there is evidence that repair of cardiac and
vascular tissue by bone marrow stem cells are naturally occurring processes after injury (35, 124). Besides tissue-specific
resident stem cells, a very low level of progenitor cells move
from their bone marrow niche into peripheral blood under
physiological conditions as a part of the homeostatic mechanism. However, in patients with acute myocardial infarction,
circulating mononuclear cells are significantly increased (124).
It is thought that tissue ischemia mobilizes mononuclear cells
from the bone marrow to the peripheral blood, and the mobilized mononuclear cells home specifically to participate in
neovascularization and differentiate into mature endothelial
cells (130). It has been demonstrated that after myocardial
infarction, mesenchymal stem cells are recruited to the injured
heart, where they undergo milieu-dependent differentiation and
may participate in the pathophysiological mechanisms of
postinfarct remodeling, angiogenesis, and maturation of the
scar (17). However, the process is far from being adequate to
compensate for the massive loss of cardiomyocytes and needs
to be supported for its inadequacy from an outside intervention.
Attempts are being made to pharmacologically augment the
process of mobilization of progenitor cells from the bone
marrow niche for their homing into the infarcted myocardium
(96).
Stem cell mobilization using various cytokines and growth
factors is routinely practiced in hematological procedures. The
use of cytokines for induction of endogenous bone marrow
stem cells to home into the infarcted myocardium is gaining
popularity. It is less invasive and obviates the use of cells from
nonautologous sources and hence the risk of transmission of
infectious agents and immunological rejection of the cell graft.
These advantages of cytokine-induced mobilization of endogenous bone marrow stem cells make it a superior choice for
regenerating infarcted myocardium. Various cytokines and
growth factors either alone or in combination together can
release HSC from the bone marrow into the peripheral blood
(7, 85, 90, 100, 119, 149). G-CSF is currently the most
commonly used agent for HSC mobilization (131). This is
AJP-Heart Circ Physiol • VOL
based on the fact that G-CSF is an effective chemoattractant for
bone marrow stem cells and enhances the infiltration of the side
population after myocardial infarction in mice (2). The outcome of the mobilization protocol is significantly influenced by
the dose and route of G-CSF administration. Similarly, a
comparative study (113) between continuous administration
and bolus administration of G-CSF has shown that the former
mode of administration reduced activation of polymorphonuclear cells. Treatment with G-CSF also induces an angiogenic
response (120). Despite its effectiveness in translocation of
bone marrow cells from their niche, the high dose of G-CSF
that is required to achieve bone marrow cell mobilization is not
without side effects (49, 58, 79). To overcome this problem, a
combination of G-CSF with stem cell factor-1 (SCF-1) has
been used (146). SCF is expressed constitutively in bone
marrow stromal cells, fibroblasts, and endothelial cells and is
essential to the survival, proliferation, adhesion/migration, and
differentiation of HSC (20, 43). The synergistic interaction
between G-CSF and other cytokines in mobilizing progenitor
cells from the bone marrow has been demonstrated by multiple
studies in animals and humans (38, 70, 89). Recent studies
support the ability of HSC induced by cytokine(s) to transdifferentiate into cardiomyocytes; however, nothing is actually
known regarding the process of transendothelial migration of
HSC to the myocardium. In the hematopoietic microenvironment, bone marrow-derived endothelial cells may play an
important role in the trafficking and maintenance of progenitor
and stem cells due to adhesive interactions and paracrine
secretion of hematopoietic growth factors.
It is hypothesized that bone marrow cells are integrated in
their bone marrow niche with the bone marrow stroma. For the
translocation of cells from their bone marrow niche under the
influence of various cytokines, it is imperative to break these
cytoadhesive interactions before they are blood borne (67).
Besides others, CXCR-4 is a cell surface antigen expressed on
hematopoietic progenitor cells including the primitive, pluripotent CD34⫹ subpopulation and is related to efficient SDF1-induced migration in vitro (88, 107). Stromal cell derived
factor-1 (SDF-1), through its receptor CXCR-4, is believed to
be superior in stem cell mobilization, angiogenesis, and cell
survival. Besides, G-CSF-induced HSC mobilization may also
involve the SDF-1/CXCR-4 axis (107). Homing in of the
mobilized stem cells to the injured myocardium is milieu
dependent. Ischemia enhances the homing-in process. A multitude of local and systemic factors influence the homing-in of
stem cells, including VEGF and its receptor-2, c-kit, and stem
cell factors (93).
FUTURE DIRECTIONS
Stem cell therapy in general (44, 50, 51) and therapy based
on bone marrow-derived stem cells in particular (31, 105, 127)
is quickly emerging as an alternative strategy in cardiovascular
therapeutics. Proof-of-concept animal studies and phase 1
clinical trials have already shown the safety and feasibility of
this approach. However, there are some fundamental issues
that need to be resolved to streamline research in this area. The
ability of bone marrow cells to transdifferentiate into cardiomyocytes and integrate into the host tissue is being questioned.
The proponents of fusion as the mechanism for the survival of
donor cells posttransplantation are arguing for the development
288 • JUNE 2005 •
www.ajpheart.org
Downloaded from http://ajpheart.physiology.org/ by 10.220.32.247 on April 29, 2017
control for comparison of results. BMMNC (CD34⫹/CD45⫹)
were purified from autologous bone marrow aspirates of each
patient undergoing BMMNC transplantation, and CPC were
purified and expanded ex vivo for 3 days for each patient
undergoing CPC transplantation from 250 ml venous blood.
Except for one patient who suffered anterior wall infarction 3
days after cell transplantation, the whole exercise was without
any problem. A 4 mo follow-up revealed improvement in
global cardiac function; however, it did not differ between
patients receiving BMMNC or CPC. The control group patients without cell therapy revealed no significant changes in
the respective parameters of observation. The study results
highlight the safety and efficacy of the procedure, like many of
the previously reported studies. Moreover, it also showed that
transplantation of CPC, the collection and availability of which
is relatively easy and less invasive compared with BMMNC, is
as effective as BMMNC in improving contractile function of
the heart and coronary artery flow reserve.
Invited Review
BONE MARROW STEM CELLS FOR HEART
AJP-Heart Circ Physiol • VOL
provide an alternative option with superior attributes in cell
therapy for cardiac repair.
The results of the phase 1 clinical studies have been encouraging. However, in the absence of a blinded control group, and
with a small number of patients involved in most of these
studies, it is difficult to assert the beneficial effects of stem cell
therapy. Furthermore, it is important to define the patient
population that can benefit from cell therapy. For this purpose,
a more concerted effort between the various research groups
involved is needed.
REFERENCES
1. Abedi M, Greer DA, Colvin GA, Demers DA, Dooner MS, Harpel
JA, Pimentel J, Menon MK, and Quesenberry PJ. Tissue injury in
marrow transdifferentiation. Blood Cells Mol Dis 32: 42– 46, 2004.
2. Adachi Y, Imagawa J, Suzuki Y, Yogo K, Fukazawa M, Kuromaru
O, and Saito Y. G-CSF treatment increases side population cell infiltration after myocardial infarction in mice. J Mol Cell Cardiol 36:
707–710, 2004.
3. Agbulut O, Menot ML, Li Z, Marotte F, Paulin D, Hagege AA,
Chomienne C, Samuel JL, and Menasche P. Temporal patterns of bone
marrow cell differentiation following transplantation in doxorubicin
induced cardiomyopathy. Cardiovasc Res 58: 451– 459, 2003.
4. Airey JA, Almeida-Porada G, Colletti EJ, Porada CD, Chamberlain
J, Movsesian M, Sutko JL, and Zanjani ED. Human mesenchymal
stem cells form Purkinje fibers in fetal sheep heart. Circulation 109:
1401–1407, 2004.
5. Alvarez-Donaldo M, Pardal R, Garcia-Verdugo JM, Fike JR, Lee
HO, Pfeiffer K, Lois C, Morrison SJ, and Alvarez-Buylla A. Fusion
of bone-marrow-derived cells with Purkinje neurons, cardiomyocytes
and hepatocytes. Nature 425: 968 –973, 2003.
6. Anderson DJ, Gage FH, and Weissman IL. Can stem cell cross lineage
boundaries? Nat Med 7: 393–395, 2001.
7. Andrews RG, Bensinger WI, Knitter GH, Bartelmez SH, Longin K,
Bernstein ID, Appelbaum FR, and Zsebo KM. The ligand for c-kit,
stem cell factor, stimulates the circulation of cells that engraft lethally
irradiated baboons. Blood 80: 2715–2720, 1992.
8. Annabi B, Lee YT, Turcotte S, Naud E, Desrosiers RR, Champagne
M, Eliopoulos N, Galipeau J, and Beliveau R. Hypoxia promotes
murine bone-marrow-derived stromal cell migration and tube formation.
Stem Cells 21: 337–347, 2003.
9. Asahara T, Masuda H, Takahashi T, Kalka C, Pastore C, Silver M,
Kearne M, Magner M, and Isner JM. Bone marrow origin of endothelial progenitor cells responsible for postnatal vasculogenesis in physiological and pathological neovascularization. Circ Res 85: 221–228,
1999.
10. Assmus B, Schachinger V, Teupe C, Britten M, Lehmann R, Dobert
N, Grunwald F, Aicher A, Urbich C, Martin H, Hoelzer D, Dimmeler
S, and Zeiher AM. Transplantation of progenitor cells and regeneration
enhancement in acute myocardial infarction (TOPCARE-AMI). Circulation 106: 3009 –3017, 2002.
10a.Aviles FF, San Roman JA, Garcia Frade J, Valdes M, Sanchez A, de
la Fuente L, Penarrubia MJ, Fernandez ME, Tejedor P, Duran JM,
Hernandez C, Sanz R, and Garcia Sancho J. [Intracoronary stem cell
transplantation in acute myocardial infarction]. Rev Esp Cardiol 57:
201–208, 2004.
11. Balsam LB, Wagers AJ, Christensen JL, Kofidis T, Weissman IL,
and Robbins RC. Haematopoietic stem cells adopt mature haematopoietic fates in ischemic myocardium. Nature 428: 668 – 673, 2004.
12. Bayes-Genis A, Muniz-Diaz EM, Catasus L, Arilla M, Rodriguez C,
Sierra J, Madoz PJ, and Cinca J. Cardiac chimerism in recipients of
peripheral-blood and bone marrow stem cells. Eur J Heart Fail 6:
399 – 402, 2004.
13. Bel A, Messas E, Agbulut O, Richard P, Samuel JL, Bruneval P,
Hagege AA, and Menasche P. Transplantation of autologous fresh bone
marrow into infarcted myocardium: a word of caution. Circulation 108:
247–252, 2003.
14. Beran G, Glogar D, and Lang IM. Improved myocardial viability
following intramyocardial autologous bone marrow injection after acute
myocardial infarction (Abstract). Heart 89: 930, 2003.
288 • JUNE 2005 •
www.ajpheart.org
Downloaded from http://ajpheart.physiology.org/ by 10.220.32.247 on April 29, 2017
of hybrid cells as a result of fusion between donor cells and
host cardiomyocytes, thus exhibiting a dual phenotype. Subsequently, the hybrid cells take on the function of the existing
cells and proliferation results from the hybrid cells rather than
stem cell transdifferentiation. However, they have failed to
explain the extensive tissue regeneration that so many earlier
studies have shown to occur after cell transplantation. These
issues regarding the mechanistic basis of cell transplantation
need extensive and in-depth studies. Resolving this issue is
imperative because fused cells may have a different therapeutic
potential and prognostic outcome compared with differentiated
cells.
Combining bone marrow stem cell transplantation together
with the delivery of therapeutic genes to the injured myocardium is an attractive alternative strategy. A previous study (40)
has shown the effectiveness of overexpression of angiogenic
genes together with stem cell transplantation in animal models.
Genetic modulation of bone marrow stromal cells to overexpress the endothelial nitric oxide synthase gene did not interfere with their differentiation potential (25). Transplantation of
mesenchymal stem cells overexpressing Akt has been shown to
improve the survival of the donor cell and prevent ventricular
remodeling together with improved cardiac function (78).
Age-related poor proliferative and differentiation potential
of bone marrow stem cells is an important parameter that needs
special consideration. Hence, in the case of old patients, the
pros and cons of using autologous bone marrow cells for
transplantation need to be carefully weighed. It has been shown
that bone marrow undergoes age-related changes with respect
to proliferative capacity and multilineage potential (87, 128). A
clear relevance has been established between an intrinsic asyet-undefined genetically controlled program of impaired stem
cell functioning with aging. Interestingly, the stem cell pool in
short-lived DBA/2 (D2) mice was reduced during aging, in
contrast to long-lived C57BL/6 (B6) mice (57). Besides,
growth of bone marrow stromal cells is better from bone
marrow aspirates of younger patients (122). In the light of
these observations, the age of the donor of bone marrow stem
cells for transplantation has a strong influence on the outcome
of the procedure.
Mobilization of bone marrow cells for cardiac repair is an
interesting approach. Use of G-CSF has shown promise but has
untoward effects that may limit its clinical application (49,
136). The combination of G-CSF with other cytokines and
growth factors has been used for mobilization effects and to
overcome this problem (93, 142). Studies have also shown that
stem cells show a uniquely selective response to various
cytokines and combinations of cytokines (146). The knowledge about a specific subpopulation of bone marrow cells that
can differentiate into cardiomyocytes combined with the novel
concept of lineage-specific mobilization may help to achieve
better results with fewer side effects of the cytokine therapy.
Like numerous other tissues that house their resident stem
cells, there is strong evidence for the presence of a myocardial
stem cell-like population that retains the characteristics of other
stem cells. However, with a decreased ability to change their
genotype and gene expression profile, they are considered to
have myocardium-restricted functions (48, 83). Hence, besides
transplantation and mobilization strategies, activation of the
resident stem cells for participation in the repair process may
H2563
Invited Review
H2564
BONE MARROW STEM CELLS FOR HEART
AJP-Heart Circ Physiol • VOL
35.
36.
37.
38.
39.
40.
41.
42.
43.
44.
45.
46.
47.
48.
49.
50.
51.
52.
53.
54.
55.
cardium is safe and enhances cardiac function in humans. Cell Transplant 13: 7–13, 2004.
Gill M, Dias S, Hattori K, Rivera ML, Hicklin D, Witte L, Girardi L,
Yurt R, Himel H, and Rafii S. Vascular trauma induces rapid but
transient mobilization of VEGF2(⫹) AC133(⫹) endothelial precursor
cells. Circ Res 88: 167–174, 2001.
Goodell MA, Rosenzweig M, Kim H, Marks DF, DeMaria M, Paradis G, Grupp SA, Sieff CA, Mulligan RC, and Johnson RP. Dye
efflux studies suggest that hematopoietic stem cells expressing low or
undetectable levels of CD34 antigen exist in multiple species. Nat Med 3:
1337–1345, 1997.
Graf T. Differentiation plasticity of hematopoietic cells. Blood 99:
3089 –3101, 2002.
Grafte-Faure S, Levesque C, Ketata E, Jean P, Vasse SC, and
Vannier JM. Recruitment of primitive peripheral blood cells: synergism
of interleukin 12 with interleukin 6 and stromal cell-derived factor-1.
Cytokine 12: 1–7, 2000.
Grove JE, Bruscia E, and Krause D. Plasticity of bone marrow derived
stem cells. Stem Cells 22: 487–500, 2004.
Haider Kh H, Lei Y, Shujia J, Ge R, Law PK, Chua T, Wong P, and
Sim EKW. Angiomyogenesis for cardiac repair using human myoblasts
as carriers of human vascular endothelial growth factor. J Mol Med 82:
539 –549, 2004.
Hakuno D, Fukuda K, Makino S, Konishi F, Tomita Y, Manabe T,
Suzuki Y, Umezawa A, and Ogawa S. Bone marrow-derived regenerated cardiomyocytes (CMG Cells) express functional adrenergic and
muscarinic receptors. Circulation 105: 380 –386, 2002.
Hamano K, Li TS, Kobayashi T, Hirata K, Yano M, Kohno M, and
Matsuzaki M. Therapeutic angiogenesis induced by local autologous
bone marrow cell implantation. Ann Thorac Surg 73: 1210 –1215, 2002.
Hartman M, Piliponsky AM, Temkin V, and Levi-Schaffer F. Human
peripheral blood eosinophils express stem cell factor. Blood 97: 1086 –
1091, 2001.
Hattan N, Warltier D, Gu W, Kolz C, Chilian WM, and Weihrauch
D. Autologous vascular smooth muscle cell-based myocardial gene
therapy to induce coronary collateral growth. Am J Physiol Heart Circ
Physiol 287: H488 –H493, 2004.
Heng BC, Haider KhH, Sim EK, Cao T, and Ng SC. Strategies for
directing the differentiation of stem cells into the cardiomyogenic lineage
in vitro. Cardiovasc Res 62: 34 – 42, 2004.
Herzog EL, Chai L, and Krause DS. Plasticity of marrow-derived stem
cells. Blood 102: 3483–3493, 2003.
Hiasa K, Egashira K, Kitamoto S, Ishibashi M, Inoue S, Ni W, Zhao
Q, Nagata S, Katoh M, Sata M, and Takeshita A. Bone marrow
mononuclear cell therapy limits myocardial infarct size through vascular
endothelial growth factor. Basic Res Cardiol 99: 1– 8, 2004.
Hierlihy AM, Seale P, Lobe CG, Rudnicki MA, and Megeney LA.
The post-natal heart contains a myocardial stem cell population. FEBS
Lett 530: 239 –243, 2002.
Hill JM, Paul JD, Powell TM, McCoy JP, Dunbar CE, Horne M,
Csako G, and Cannon RO. Efficacy and risk of granulocyte colony
stimulating factor administration in patients with severe coronary artery
disease (Abstract). Circulation 108, Suppl IV: 478, 2003.
Hodgson DM, Behfar A, Zingman LV, Kane GC, Perez-Terzic C,
Alekseev AE, Pucéat M, and Terzic A. Stable benefit of embryonic
stem cell therapy in myocardial infarction. Am J Physiol Heart Circ
Physiol 287: H471–H479, 2004.
Horackova M, Chen AR, Armour JA, Cattini PA, Livingston R, and
Byczko Z. Cell transplantation for treatment of acute myocardial infarction: unique capacity for repair by skeletal muscle satellite cells. Am J
Physiol Heart Circ Physiol 287: H1599 –H1608, 2004.
Horwitz EM. Stem cell plasticity: the growing potential of cellular
therapy. Arch Med Res 34: 600 – 606, 2003.
Huttmann A, Li L, and Duhrsen U. Bone marrow derived stem cells
and plasticity. Ann Hematol 604: 599 – 604, 2003.
Ishida M, Tomita S, Nakatani T, Fukuhara S, Hamamoto M, Nagaya
N, Ohtsu Y, Suga M, Yutani C, Yagihara T, Yamada K, and
Kitamura S. Bone marrow mononuclear cell transplantation had beneficial effects on doxorubicin-induced cardiomyopathy. J Heart Lung
Transplant 23: 436 – 445, 2004.
Jackson KA, Majka SM, Wang H, Pocius J, Hartley CJ, Majesky
MW, Entman ML, Michael LH, Hirschi KK, and Goodell MA.
Regeneration of ischemic cardiac muscle and vascular endothelium by
adult stem cells. J Clin Invest 107: 1395–1402, 2001.
288 • JUNE 2005 •
www.ajpheart.org
Downloaded from http://ajpheart.physiology.org/ by 10.220.32.247 on April 29, 2017
15. Bhatia M, Bonnet D, Murdoch B, Gan OI, and Dick JE. A newly
discovered class of human hematopoietic cells with SCID-repopulating
activity. Nat Med 4: 1038 –1045, 1998.
16. Bittira B, Kuang JQ, Al-Khaldi A, Shum-Tim D, and Chiu RCJ. In
vitro pre-programming of marrow stromal cells for myocardial regeneration. Ann Thorac Surg 74: 1154 –1160, 2002.
17. Bittira B, Shum-Tim D, Al-Khaldi A, and Chiu RCJ. Mobilization
and homing of bone marrow stromal cells in myocardial infarction. Eur
J Cardiothorac Surg 24: 393–398, 2003.
18. Brazekon TR, Rossi FM, Kesher GI, and Blau HM. From bone
marrow to brain: expression of neuronal phenotypes in adult mice.
Science 290: 1775–1779, 2000.
19. Britten MB, Abolmaali ND, Assmus B, Lehmann R, Honold J,
Schmitt J, Vogl TJ, Martin H, Schachinger V, Dimmeler S, and
Zeiher AM. Infarct remodeling after intra coronary progenitor cell
treatment in patients with acute myocardial infarction (TOPCARE-AMI):
mechanistic insights from serial contrast-enhanced magnetic resonance
imaging. Circulation 108: 2212–2218, 2003.
20. Broudy VC. Stem cell factor and hematopoiesis. Blood 90: 1345–1364,
1997.
21. Carter JE and Schuchman EH. Mesenchymal stem cell therapy for
neurodegenerative disease (Abstract). Neurobiol Aging 21: 152, 2000.
22. Chen SL, Fang WW, Ye F, Liu YH, Qian J, Shan SJ, Zhang JJ,
Chunhua RZ, Liao LM, Lin S, and Sun JP. Effect on left ventricular
function of intracoronary transplantation of autologous bone marrow
mesenchymal stem cells in patients with acute myocardial infarction.
Am J Cardiol 94: 92–95, 2004.
23. Condorelli G, Borello U, De Angelis L, Latronico M, Sirabella D,
Coletta M, Galli R, Balconi G, Follenzi A, Frati G, Cusella De
Angelis MG, Gioglio L, Amuchastegui S, Adorini L, Naldini L,
Vescovi A, Dejana E, and Cossu G. Cardiomyocytes induce endothelial
cells to trans-differentiate into cardiac muscle: implications for myocardium regeneration. Proc Natl Acad Sci USA 98: 10733–10738, 2001.
24. Davani S, Marandin A, Mersin N, Royer B, Kantelip B, Herve P,
Etievent JP, and Kantelip JP. Mesenchymal progenitor cells differentiate into an endothelial phenotype, enhance vascular density, and improve heart function in a rat cellular cardiomyoplasty model. Circulation
108: 253–258, 2003.
25. Deng W, Bivalacqua TJ, Chattergoon NN, Hyman AL, Jeter JR Jr,
and Kadowitz PJ. Adenoviral gene transfer of eNOS: high-level expression in ex vivo expanded marrow stromal cells. Am J Physiol Cell
Physiol 285: C1322–C1329, 2003.
26. Duan HF, Wu CT, Wu DL, Lu Y, Liu HJ, Ha XQ, Zhang QW, Wang
H, Jia XX, and Wang LS. Treatment of myocardial ischemia with bone
marrow-derived mesenchymal stem cells over-expressing hepatocyte
growth factor. Mol Ther 8: 467– 474, 2003.
27. Eisenberg LM, Burns L, and Eisenberg CA. Hematopoietic cells from
bone marrow have the potential to differentiate into cardiomyocytes in
vitro. Anat Rec A Discov Mol Cell Evol Biol 274: 870 – 882, 2003.
28. Fernandez-Aviles F, San Roman JA, Garcia-Frade J, Fernandez
ME, Penarrubia MJ, de la Fuente L, Gomez-Bueno M, Cantalapiedra A, Fernandez J, Gutierrez O, Sanchez PL, Hernandez C,
Sanz R, Garcia-Sancho J, and Sanchez A. Experimental and clinical
regenerative capability of human bone marrow cells after myocardial
infarction. Circ Res 95: 742–748, 2004.
30. Fuchs S, Baffour R, Zhou YF, Shou M, Pierre A, Tio FO, Weissman
NJ, Leon MB, Epstein SE, and Kornowski R. Transendocardial delivery of autologous bone marrow enhances collateral perfusion and
regional function in pigs with chronic experimental myocardial ischemia.
J Am Coll Cardiol 37: 1726 –1732, 2001.
31. Fuchs S, Satler LF, Kornowski R, Okubagzi P, Weisz G, Baffour R,
Waksman R, Weissman NJ, Cerqueira M, Leon MB, and Epstein
SE. Catheter-based autologous bone marrow myocardial injection in
no-option patients with advanced coronary artery disease: a feasibility
study. J Am Coll Cardiol 41: 1721–1724, 2003.
32. Fukuda K. Development of regenerative cardiomyocytes from mesenchymal stem cells for cardiovascular tissue engineering. Artif Organs 25:
187–193, 2001.
33. Fukuhara S, Tomita S, Yamashiro S, Morisaki T, Yutani C, Kitamura S, and Nakatani T. Direct cell-cell interaction of cardiomyocytes
is key for bone marrow stromal cells to go into cardiac lineage in vitro.
J Thorac Cardiovasc Surg 125: 1470 –1480, 2003.
34. Galinanes M, Loubani M, Davies J, Chin D, Pasi J, and Bell PR.
Auto-transplantation of un-manipulated bone marrow into scarred myo-
Invited Review
BONE MARROW STEM CELLS FOR HEART
AJP-Heart Circ Physiol • VOL
73.
74.
75.
76.
77.
78.
79.
80.
81.
82.
83.
84.
85.
86.
87.
88.
89.
90.
91.
angiogenesis induced by bone marrow cell implantation. Am J Physiol
Heart Circ Physiol 285: H931–H937, 2003.
Li TS, Hayashi M, Liu ZL, Ito H, Mikamo A, Furutani A, Matsuzaki
M, and Hamano K. Low angiogenic potency induced by the implantation of ex vivo expanded CD117(⫹) stem cells. Am J Physiol Heart Circ
Physiol 286: H1236 –H1241, 2004.
Liechty KW, MacKenzie TC, Shaaban AF, Radu A, Moseley AM,
Deans R, Marshak DR, and Flake AW. Human mesenchymal stem
cells engraft and demonstrate site-specific differentiation after in utero
transplantation in sheep. Nat Med 6: 1282–1286, 2000.
Liu J, Hu Q, Wang Z, Chengsu Xu C, Wang X, Gong G, Mansoor A,
Lee J, Hou M, Zeng L, Zhang JR, Jerosch-Herold M, Guo T, Bache
RJ, and Zhang J. Autologous stem cell transplantation for myocardial
repair. Am J Physiol Heart Circ Physiol 287: H501–H511, 2004.
Lum LG, Fok H, Sievers R, Abedi M, Queensberry PJ, and Lee RJ.
Targetting Lin-Sca⫹ heamatopietic stem cells with bispecific antibodies
to injured myocardium. Blood Cells Mol Dis 32: 82– 87, 2004.
Makino S, Fukuda K, Miyoshi S, Konishi F, Kodama H, Pan J, Sano
M, Takahashi T, Hori S, Abe H, Hata J, Umezawa A, and Ogawa S.
Cardiomyocytes can be generated from marrow stromal cells in vitro.
J Clin Invest 103: 697–705, 1999.
Mangi AA, Noiseux N, Kong D, He H, Rezvani M, Ingwall JS, and
Dzau VJ. Mesenchymal stem cells modified with Akt prevent remodeling and restore performance of infarcted hearts. Nat Med 9: 1195–1201,
2003.
Matsubara H. Risk to the coronary arteries of intracoronary stem cell
infusion and GCSF cytokine therapy. Lancet 363: 746 –747, 2004.
Matsuura K, Nagai T, Nishigaki N, Oyama T, Nishi J, Wada H, Sano
M, Toko H, Akazawa H, Sato T, Nakaya H, Kasanuki H, and
Komuro I. Adult cardiac Sca-1-positive cells differentiate into beating
cardiomyocytes. J Biol Chem 279: 11384 –11391, 2004.
Matsuzaki Y, Kinjo K, Mulligan RC, and Okano H. Unexpectedly
efficient homing capacity of purified murine hematopoietic stem cells.
Immunity 20: 87–93, 2004.
Menasche P, Hagege AA, Scorsin M, Pouzet B, Desnos M, Duboc D,
Schwartz K, Vilquin JT, and Marolleau JP. Myoblast transplantation
for heart failure. Lancet 357: 279 –280, 2001.
Messina E, De Angelis L, Frati G, Morrone S, Chimenti S, Fiordaliso
F, Salio M, Battaglia M, Latronico MV, Coletta M, Vivarelli E, Frati
L, Cossu G, and Giacomello A. Isolation and expansion of adult cardiac
stem cells from human and murine heart. Circ Res 95: 911–921, 2004.
Min JY, Sullivan MF, Yang Y, Zhang JP, Converso KL, Morgan JP,
and Xiao YF. Significant improvement of heart function by co transplantation of human mesenchymal stem cells and fetal cardiomyocytes in
post-infarcted pigs. Ann Thorac Surg 74: 1568 –1575, 2002.
Minatoguchi S, Takemura G, Chen XH, Wang N, Uno Y, Koda M,
Arai M, Misao Y, Lu C, Suzuki K, Goto K, Komada A, Takahashi T,
Kosai K, Fujiwara T, and Fujiwara H. Acceleration of the healing
process and myocardial regeneration may be important as a mechanism
of improvement of cardiac function and remodeling by postinfarction
granulocyte colony-stimulating factor treatment. Circulation 109: 2572–
2580, 2004.
Minguell JJ, Erices A, and Conget P. Mesenchymal stem cells. Exp
Biol Med 226: 507–520, 2001.
Moerman EJ, Teng K, Lipschitz DA, and Lecka-Czernik B. Aging
activates adipogenic and suppresses osteogenic programs in mesenchymal marrow stroma/stem cells: the role of PPAR-gamma2 transcription
factor and TGF-beta/BMP signaling pathways. Aging Cell 3: 379 –389,
2004.
Möhle R, Bautz F, Rafii S, Moore MAS, Brugger W, and Kanz L. The
chemokine receptor CXCR-4 is xpressed on CD34⫹ ematopoietic progenitors and leukemic cells and mediates transendothelial migration
induced by stromal cell-derived factor-1. Blood 91: 4523– 4530, 1998.
Molineux G, McCrea C, Yan XQ, Kerzic P, and McNiece I. Flt-3
ligand synergizes with granulocyte colony-stimulating factor to increase
neutrophil numbers and to mobilize peripheral blood stem cells with
longterm repopulating potential. Blood 89: 3998 – 4004, 1997.
Moore MA, Hattori K, Heissig B, Shieh JH, Dias S, Crystal RG, and
Rafii S. Mobilization of endothelial and hematopoietic stem and progenitor cells by adenovector-mediated elevation of serum levels of SDF-1,
VEGF, and angiopoietin-1. Ann NY Acad Sci 938: 45– 47, 2001.
Murry CE, Soonpaa MH, Reinecke H, Nakajima H, Nakajima HO,
Rubart M, Pasumarthi KB, Virag JI, Bartelmez SH, Poppa V,
Bradford G, Dowell JD, Williams DA, and Field LJ. Haematopoietic
288 • JUNE 2005 •
www.ajpheart.org
Downloaded from http://ajpheart.physiology.org/ by 10.220.32.247 on April 29, 2017
56. Kamihata H, Matsubara H, Nishiue T, Fujiyama S, Tsutsumi Y,
Ozono R, Masaki H, Mori Y, Iba O, Tateishi E, Kosaki A, Shintani
S, Murohara T, Imaizumi T, and Iwasaka T. Implantation of bone
marrow mononuclear cells into ischemic myocardium enhances collateral
perfusion and regional function via side supply of angioblasts, angiogenic ligands, and cytokines. Circulation 104: 1046 –1052, 2001.
57. Kamminga LM, van Os R, Ausema A, Noach EJ, Weersing E, Dontje
B, Vellenga E, and de Haan G. Impaired hematopoietic stem cell
functioning after serial transplantation and during normal aging. Stem
Cells 23: 82–92, 2005.
58. Kang HJ, Kim HS, Zhang SY, Park KW, Cho HJ, Koo BK, Kim YJ,
Soo Lee D, Sohn DW, Han KS, Oh BH, Lee MM, and Park YB.
Effects of intracoronary infusion of peripheral blood stem-cells mobilized with granulocyte-colony stimulating factor on left ventricular systolic function and restenosis after coronary stenting in myocardial infarction: the MAGIC cell randomized clinical trial. Lancet 363: 751–756,
2004.
59. Kawada H, Fujita J, Kinjo K, Matsuzaki Y, Tsuma M, Miyatake H,
Muguruma Y, Tsuboi K, Itabashi Y, Ikeda Y, Ogawa S, Okano H,
Hotta T, Ando K, and Fukuda K. Non-hematopoietic mesenchymal
stem cells can be mobilized and differentiate into cardiomyocytes after
myocardial infarction. Blood 104: 3581–3587, 2004.
60. Kehat I, Kenyagin-Karsenti D, Snir M, Segev H, Amit M, Gepstein
A, Livne E, Binah O, Itskovitz-Eldor J, Gepstein L. Human embryonic stem cells can differentiate into myocytes with structural and
functional properties of cardiomyocytes. J Clin Invest 108: 407– 414,
2001.
61. Kim DK, Fujiki Y, Fukushima T, Ema H, Shibuya A, and Nakauchi
H. Comparison of hematopoietic activities of human bone marrow and
umbilical cord blood CD34 positive and negative cells. Stem Cells 17:
286 –294, 1999.
62. Kinnaird T, Stabile E, Burnett MS, Lee CW, Barr S, Fuchs S, and
Epstein SE. Marrow-derived stromal cells express genes encoding a
broad spectrum of arteriogenic cytokines and promote in vitro and in vivo
arteriogenesis through paracrine mechanisms. Circ Res 94: 678 – 685,
2004.
63. Kinnaird T, Stabile E, Burnett MS, Shou M, Lee CW, Barr S, Fuchs
S, and Epstein SE. Local delivery of marrow-derived stromal cells
augments collateral perfusion through paracrine mechanisms. Circulation 109: 1543–1549, 2004.
64. Kocher AA, Schuster MD, Szabolcs MJ, Takuma S, Burkhoff D,
Wang J, Homma S, Edwards NM, and Itescu S. Neovascularization of
ischemic myocardium by human bone-marrow-derived angioblasts prevents cardiomyocyte apoptosis, reduces remodeling and improves cardiac function. Nat Med 7: 430 – 436, 2001.
65. Kondo M, Wagers AJ, Manz MG, Prohaska SS, Scherer DC, Beilhack GF, Shizuru JA, and Weissman IL. Biology of hematopoietic
stem cells and progenitors: implications for clinical application. Annu
Rev Immunol 21: 759 – 806, 2003.
66. Krause DS, Theise ND, Collector MI, Henegariu O, Hwang S,
Gardner R, Neutzel S, and Sharkis SJ. Multi-organ, multi-lineage
engraftment by a single bone marrow-derived stem cell. Cell 105:
369 –377, 2001.
67. Kronenwett R, Martin S, and Haas R. The role of cytokines and
adhesion molecules for mobilization of peripheral blood stem cells. Stem
Cells 18: 320 –330, 2000.
68. Kudo M, Wang Y, Wani MA, Xu M, Ayub A, and Ashraf M.
Implantation of bone marrow stem cells reduces the infarction and
fibrosis in ischemic mouse heart. J Mol Cell Cardiol 35: 1113–1119,
2003.
69. Lagasse E, Connors H, Al-Dhalimy M, Reitsma M, Dohse M, Osborne L, Wang X, Finegold M, Weissman IL, and Grompe M.
Purified hematopoietic stem cells can differentiate into hepatocytes in
vivo. Nat Med 6: 1229 –1234, 2000.
70. Lataillade JJ, Clay D, Dupuy C, Rigal S, Jasmin C, Bourin P, and Le
Bousse-Kerdiles MC. Chemokine SDF-1 enhances circulating CD34(⫹)
cell proliferation in synergy with cytokines: possible role in progenitor
survival. Blood 95: 756 –768, 2000.
71. Li TS, Hamano K, Suzuki K, Ito H, Zempo N, and Matsuzaki M.
Improved angiogenic potency by implantation of ex vivo hypoxia prestimulated bone marrow cells in rats. Am J Physiol Heart Circ Physiol
283: H468 –H473, 2002.
72. Li TS, Hamano K, Nishida M, Hayashi M, Ito H, Mikamo A, and
Matsuzaki M. CD117⫹ stem cells play a key role in therapeutic
H2565
Invited Review
H2566
92.
93.
94.
95.
96.
98.
99.
100.
101.
102.
103.
104.
105.
106.
107.
108.
109.
stem cells do not transdifferentiate into cardiac myocytes in myocardial
infarcts. Nature 428: 664 – 668, 2004.
Nagaya N, Fujii T, Iwase T, Ohgushi H, Itoh T, Uematsu M,
Yamagishi M, Mori H, Kangawa K, and Kitamura S. Intravenous
administration of mesenchymal stem cells improves cardiac function in
rats with acute myocardial infarction through angiogenesis and myogenesis. Am J Physiol Heart Circ Physiol 287: H2670 –H2676, 2004.
Nakamura Y, Tajima F, Ishiga K, Yamazaki H, Oshimura M, Shiota
G, and Murawaki Y. Soluble c-kit receptor mobilizes hematopoietic
stem cells to peripheral blood in mice. Exp Hematol 32: 390 –396, 2004.
Nakauchi H. Hematopoietic stem cells: are they CD34-positive or
CD34-negative? Nat Med 4: 1009 –1010, 1998.
Nilsson SK, Dooner MS, Weier HU, Frenkel B, Lian JB, Stein GS,
and Quesenberry PJ. Cells capable of bone production engraft from
whole bone marrow transplants in non-ablated mice. J Exp Med 189:
729 –734, 1999.
Norol F, Merlet P, Isnard R, Sebillon P, Bonnet N, Cailliot C,
Carrion C, Ribeiro M, Charlotte F, Pradeau P, Mayol JF, Peinnequin A, Drouet M, Safsafi K, Vernant JP, and Herodin F. Influence
of mobilized stem cells on myocardial infarct repair in a nonhuman
primate model. Blood 102: 4361– 4368, 2003.
Nygren JM, Jovinge S, Breitbach M, Sawen P, Roll W, Hescheler J,
Taneera J, Fleischmann BK, and Jacobsen SE. Bone marrow-derived
hematopoietic cells generate cardiomyocytes at a low frequency through
cell fusion, but not transdifferentiation. Nat Med 10: 494 –501, 2004.
Oh SH, Muzzonigro TM, Bae SH, LaPlante JM, Hatch H, and
Petersen BE. Adult bone marrow-derived cells transdifferentiating into
insulin-producing cells for the treatment of type-1 diabetes. Lab Invest
84: 607– 617, 2004.
Olivares EL, Ribeiro VP, de Castro JPS, Ribeiro KC, Mattos EC,
Goldenberg RCS, Mill JG, Dohmann HF, dos Santos RR, de Carvalho ACC, and Masuda MO. Bone marrow stromal cells improve
cardiac performance in healed infarcted rat hearts. Am J Physiol Heart
Circ Physiol 287: H464 –H470, 2004.
Orlic D, Kajstura J, Chimenti S, Limana F, Jakoniuk I, Quaini F,
Nadal-Ginard B, Bodine DM, Leri A, and Anversa P. Mobilized bone
marrow cells repair the infarcted heart, improving function and survival.
Proc Natl Acad Sci USA 98: 10344 –10349, 2001.
Orlic D, Kajstura J, Chimenti S, Jakoniuk I, Anderson SM, Li B,
Pickel J, McKay R, Nadal-Ginard B, Bodine DM, Leri A, and
Anversa P. Bone marrow cells regenerate infarcted myocardium. Nature
410: 701–705, 2001.
Orlic D, Kajstura J, Chimenti S, Bodine DM, Leri A, and Anversa P.
Transplanted adult bone marrow cells repair myocardial infarcts in mice.
Ann NY Acad Sci 938: 221–229, 2001.
Oswald J, Boxberger S, Jorgensen B, Feldmann S, Ehninger G,
Bornhauser M, and Werner C. Mesenchymal stem cells can be differentiated into endothelial cells in vitro. Stem Cells 22: 377–384, 2004.
Pak HN, Qayyum M, Kim DT, Hamabe A, Miyauchi Y, Lill MC,
Frantzen M, Takizawa K, Chen LS, Fishbein MC, Sharifi BG, Chen
PS, and Makkar R. Mesenchymal stem cell injection induces cardiac
nerve aprouting and increased tenacin expression in swine model of
myocardial infarction. J Cardiovasc Electrophysiol 14: 841– 848, 2003.
Peng CF, Wei Y, Levsky JM, McDonald TV, Childs G, and Kitsis
RN. Microarray analysis of global changes in gene expression during
cardiac myocyte differentiation. Physiol Genomics 9: 145–155, 2002.
Perin EC, Dohmann HF, Borojevic R, Silva SA, Sousa AL, Mesquita
CT, Rossi MI, Carvalho AC, Dutra HS, Dohmann HJ, Silva GV,
Belem L, Vivacqua R, Rangel FO, Esporcatte R, Geng YJ, Vaughn
WK, Assad JA, Mesquita ET, and Willerson JT. Transendocardial,
autologous bone marrow cell transplantation for severe, chronic ischemic
heart failure. Circulation 107: 2294 –2302, 2003.
Petit I, Szyper-Kravitz M, Nagler A, Lahav M, Peled A, Habler L,
Ponomaryov T, Taichman RS, Arenzana-Seisdedos F, Fujii N, Sandbank J, Zipori D, and Lapidot T. G-CSF induces stem cell mobilization
by decreasing bone marrow SDF-1 and up-regulating CXCR4. Nat
Immun 3: 687– 694, 2002.
Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca
JD, Moorman MA, Simonetti DW, Craig S, and Marshak DR.
Multillineage potential of adult human mesenchymal stem cells. Science
284: 143–147, 1999.
Prosper F, Rabago G, Perez AA, Gavira JJ, Garcia MJ, Velloso
MJG, Barba J, Sanchez PL, Canizo C, Marti JM, Hernandez M,
Holgado NL, Gonzalez JM, Luengo CM, Alegria E, and Herreros J.
AJP-Heart Circ Physiol • VOL
110.
111.
112.
113.
114.
115.
116.
117.
118.
119.
120.
121.
122.
123.
124.
125.
126.
127.
128.
129.
Combine autologous myoblast transplantation with coronary revascularization in patients with non-acute myocardial infarction. J Heart Lung
Transplant Suppl S62, 2004.
Quaini F, Urbanek K, Beltrami AP, Finato N, Beltrami CA, NadalGinard B, Kajstura J, Leri A, and Anversa P. Chimerism of the
transplanted heart. N Engl J Med 346: 5–15, 2002.
Rezai N, Podor TJ, and McManus BM. Bone marrow cells in the repair
and modulation of heart and blood vessels: emerging opportunities in
native and engineered tissue and biomechanical materials. Artif Organs
28: 142–151, 2004.
Sato T, Laver JH, and Ogawa M. Reversible expression of CD34 by
murine hematopoietic stem cells. Blood 94: 2548 –2554, 1999.
Sato Y, Goto Y, Sato S, Endo S, and Sohara Y. Continuous subcutaneous injection reduces polymorphonuclear leukocyte activation by granulocyte colony-stimulating factor. Am J Physiol Lung Cell Mol Physiol
286: L143–L148, 2004.
Saito T, Kuang JG, Bittira B, Al-Kahlidi A, and Chiu RCJ. Xenotransplant cardiac chimera: immune tolerance of adult stem cells. Ann
Thorac Surg 74: 19 –24, 2002.
Saito T, Kuang JQ, Lin CC, and Chiu RC. Transcoronary implantation
of bone marrow stromal cells ameliorates cardiac function after myocardial infarction. J Thorac Cardiovasc Surg 126: 114 –123, 2003.
Sakaguchi G, SakakibaraY, Tambara K, Premaratne G, Lin X,
Nishimura K, and Komeda M. Bone marrow cell enhances the efficacy
of skeletal myoblast transplantation in rats with ischemic cadiomyopathy
(Abstract). J Heart Lung Transplant 22: S79, 2003.
Schuster MD, Kocher AA, Seki T, Martens TP, Xiang G, Homma S,
and Itescu S. Myocardial neovascularization by bone marrow angioblasts results in cardiomyocyte regeneration. Am J Physiol Heart Circ
Physiol 287: H525–H532, 2004.
Scot EW. Stem cell plasticity or fusion: two approaches to the targeted
cell therapy. Blood Cells Mol Dis 32: 65– 67, 2004.
Sean JM, Wright DE, and Weissman LI. Cyclophosphamide/granulocyte colony-stimulating factor induces hematopoietic stem cells to proliferate prior to mobilization. Proc Natl Acad Sci USA 94: 1908 –1913,
1997.
Seiler C, Pohl T, Wustmann K, Hutter D, Nicolet PA, Windecker S,
Eberli FR, and Meier B. Promotion of collateral growth by granulocytemacrophage colony-stimulating factor in patients with coronary artery
disease: a randomized, double-blind, placebo-controlled study. Circulation 104: 2012–2017, 2001.
Shake JG, Gruber PJ, Baumgartner WA, Senechal G, Meyers J,
Redmond JM, Pittenger MF, and Martin BJ. Mesenchymal stem cell
implantation in a swine myocardial infarct model: engraftment and
functional effects. Ann Thorac Surg 73: 1919 –1926, 2002.
Shamsul BS, Aminuddin BS, Ng MH, and Ruszymah BH. Age and
gender effect on the growth of bone marrow stromal cells in vitro. Med
J Malaysia 59: 196 –197, 2004.
Shi D, Reinecke H, Murry CE, and Torok-Storb B. Myogenic fusion
of human bone marrow stromal cells, but not hematopoietic cells. Blood
104: 290 –294, 2004.
Shintani S, Murohara T, Ikeda H, Ueno T, Honma T, Katoh A,
Sasaki K, Shimada T, Oike Y, and Imaizumi T. Mobilization of
endothelial progenitor cells in patients with acute myocardial infarction.
Circulation 103: 2776 –2779, 2001.
Siminiak T, Grygielska B, Jerzykowska O, Fiszer D, Kalmucki P,
Rzezniczak J, and Kurpisz M. Autologous bone marrow stem cell
transplantation in acute myocardial infarction-report of two cases. Kardiol Pol 59: 502–510, 2003.
Stamm C, Westphal B, Kleine HD, Petzsch M, Kittner C, Klinge H,
Schumichen C, Nienaber CA, Freund M, and Steinhoff G. Autologous bone marrow stem cell transplantation for myocardial regeneration.
Lancet 361: 45– 46, 2003.
Stamm C, Kleine HD, Westphal B, Petzsch M, Kittner C, Nienaber
CA, Freund M, and Steinhoff G. CABG and bone marrow stem cell
transplantation after myocardial infarction. Thorac Cardiovasc Surg 52:
152–158, 2004.
Stenderup K, Justesen J, Clausen C, and Kassem M. Aging is
associated with decreased maximal life span and accelerated senescence
of bone marrow stromal cells. Bone 33: 919 –926, 2003.
Strauer BE, Brehm M, Zeus T, Kostering M, Hernandez A, Sorg RV,
Kogler G, and Wernet P. Repair of infarcted myocardium by autologous intracoronary mononuclear bone marrow cell transplantation in
humans. Circulation 106: 1913–1918, 2002.
288 • JUNE 2005 •
www.ajpheart.org
Downloaded from http://ajpheart.physiology.org/ by 10.220.32.247 on April 29, 2017
97.
BONE MARROW STEM CELLS FOR HEART
Invited Review
BONE MARROW STEM CELLS FOR HEART
AJP-Heart Circ Physiol • VOL
145.
146.
147.
148.
149.
150.
151.
152.
153.
154.
155.
156.
157.
158.
iologic and therapeutic implications. J Thorac Cardiovasc Surg 122:
699 –705, 2001.
Wollert KC, Meyer GP, Lotz J, Ringes-Lichtenberg S, Lippolt P,
Breidenbach C, Fichtner S, Korte T, Hornig B, Messinger D, Arseniev L, Hertenstein B, Ganser A, and Drexler H. Intracoronary
autologous bone-marrow cell transfer after myocardial infarction: the
BOOST randomised controlled clinical trial. Lancet 364: 141–148, 2004.
Wright DE, Edward P, Bowman EP, Wagers AJ, Butcher EC, and
Weissman IL. Hematopoietic Stem Cells are uniquely selective in their
migratory response to chemokines. J Exp Med 195: 1145–1154 2002.
Xu M, Wani M, Dai YS, Wang J, Yan M, Ayub A, and Ashraf M.
Differentiation of bone marrow stromal cells into the cardiac phenotype
requires intercellular communication with myocytes. Circulation 110:
2658 –2665, 2004.
Xu W, Zhang X, Qian H, Zhu W, Sun X, Hu J, Zhou H, and Chen
Y. Mesenchymal stem cells from adult human bone marrow differentiates into a cardiomyocyte phenotype in vitro. Exp Biol Med (Maywood)
229: 623– 631, 2004.
Yan XQ, Briddell R, Hartley C, Stoney G, Samal B, and McNiece I.
Mobilization of long-term hematopoietic reconstituting cells in mice by
the combination of stem cell factor plus granulocyte colony-stimulating
factor. Blood 84: 795–799, 1994.
Yau TM, Tomita S, Weisel RD, Jia ZQ, Tumiati LC, Mickle DA, and
Li RK. Beneficial effect of autologous cell transplantation on infarcted
heart function: comparison between bone marrow stromal cells and heart
cells. Ann Thorac Surg 75: 169 –176, 2003.
Yeh ETH, Zhang S, Wu HD, Korbling M, Willerson JT, and Estrov
Z. Transdifferentiation of human peripheral blood CD34⫹ enriched cell
population into cardiomyocytes endothelial cells and smooth muscle
cells in vivo. Circulation 108: 2070 –2073, 2003.
Yin AH, Miraglia S, Zanjani ED, Almeida-Porada G, Ogawa M,
Leary AG, Olweus J, Kearney J, and Buck DW. AC133, a novel
marker for human hematopoietic stem and progenitor cells. Blood 90:
5002–5012, 1997.
Ying QL, Nichols J, Evans EP, and Smith AG. Changing potency by
spontaneous fusion. Nature 416: 545–548, 2002.
Yoon KJ, Kim HO, Kwak YL, Kang SM, Jang YS, Lim SH, Ahn JY,
and Li RK. Autologous bone marrow cell transplantation combined with
off-pump coronary artery bypass grafting in human ischemic myocardium. Yonsei Med J 45, Suppl: S73, 2004.
Yoon YS, Park JS, Tkebuchava T, Luedeman C, and Losordo DW.
Unexpected severe calcification after transplantation of bone marrow
cells in acute myocardial infarction. Circulation 109: 3154 –3157, 2004.
Zhang S, Guo J, Zhang P, Liu Y, Jia Z, Ma K, Li W, Li L, and Zhou
C. Long-term effects of bone marrow mononuclear cell transplantation
on left ventricular function and remodeling in rats. Life Sci 74: 2853–
2864, 2004.
Zhao LR, Duan WM, Reyes M, Keene CD, Verfaillie CM, and Low
WC. Human bone marrow stem cells exhibit neural phenotypes and
ameliorate neurological deficits after grafting into the ischemic brain of
rats. Exp Neurol 174: 11–20, 2002.
Zipori D. Mesenchymal stem cells: harnessing cell plasticity to tissue
and organ repair. Blood Cells Mol Dis 33: 211–215, 2004.
288 • JUNE 2005 •
www.ajpheart.org
Downloaded from http://ajpheart.physiology.org/ by 10.220.32.247 on April 29, 2017
130. Takahashi T, Kalka C, Masuda H, Chen D, Silver M, Kearney M,
Magner M, Isner JM, and Asahara T. Ischemia- and cytokine-induced
mobilization of bone marrow-derived endothelial progenitor cells for
neovascularization. Nat Med 5: 434 – 438, 1999.
131. Takano H, Ohtsuka M, Akazawa H, Toko H, Harada M, Hasegawa
H, Nagai T, and Komuro I. Pleiotropic effects of cytokines on acute
myocardial infarction: G-CSF as a novel therapy for acute myocardial
infarction. Curr Pharm Des 9: 1121–1127, 2003.
132. Tang YL, Zhao Q, Zhang YC, Cheng L, Liu M, Shi J, Yang YZ, Pan
C, Ge J, and Phillips MI. Autologous mesenchymal stem cell transplantation induce VEGF and neovascularization in ischemic myocardium. Regul Pept 117: 3–10, 2004.
133. Tao H and Ma DD. Evidence for transdifferentiation of human bone
marrow-derived stem cells: recent progress and controversies. Pathology
35: 6 –13, 2003.
134. Terada N, Hamazaki T, Oka M, Hoki M, Mastalerz DM, Nakano Y,
Meyer EM, Morel L, Petersen BE, and Scott EW. Bone marrow cells
adopt the phenotype of other cells by spontaneous cell fusion. Nature
416: 542–545, 2002.
135. Theise ND and d’Inversno M. Understanding cell lineages as complex
adaptive systems. Blood Cells Mol Dis 32: 17–20, 2004.
136. Togel F, Isaac J, and Westenfelder C. Hematopoietic stem cell mobilization-associated granulocytosis severely worsens acute renal failure.
J Am Soc Nephrol 15: 1261–1267, 2004.
137. Toma C, Pittenger MF, Cahill KS, Byrne BJ, and Kessler PD. Human
mesenchymal stem cells differentiate to a cardiomyocyte phenotype in
the adult murine heart. Circulation 105: 93–98, 2002.
138. Tomita S, Li RK, Weisel RD, Mickle DA, Kim EJ, Sakai T, and Jia
ZQ. Atologous transplantation of bone marrow cells improves damaged
heart function. Circulation 100: II247–II256, 1999.
139. Tomita S, Mickle DA, Weisel RD, Jia ZQ, Tumiati LC, Allidina Y,
Liu P, and Li RK. Improved heart function with myogenesis and
angiogenesis after autologous porcine bone marrow stromal cell transplantation. J Thorac Cardiovasc Surg 123: 1132–1140, 2002.
140. Tomita S, Ishida M, Nakatani T, Fukuhara S, Hisashi Y, Ohtsu Y,
Suga M, Yutani C, Yagihara T, Yamada K, and Kitamura S. Bone
marrow is a source of regenerated cardiomyocytes in doxorubicininduced cardiomyopathy and granulocyte colony-stimulating factor enhances migration of bone marrow cells and attenuates cardiotoxicity of
doxorubicin under electron microscopy. J Heart Lung Transplant 23:
577–584, 2004.
141. Tse HF, Kwong YL, Chan JKF, Lo G, Ho CL, and Lau CK.
Angiogenesis in ischemic myocardium by intramyocardial autologous
bone marrow mononuclear cell implantation. Lancet 361: 47– 49, 2003.
142. Velders GA, Pruijt JF, Verzaal P, van Os R, van Kooyk Y, Figdor
CG, de Kruijf EJ, Willemze R, and Fibbe WE. Enhancement of
G-CSF-induced stem cell mobilization by antibodies against the beta 2
integrins LFA-1 and Mac-1. Blood 100: 327–333, 2002.
143. Wang JS, Shum-Tim D, Galipeau J, Chedrawy E, Eliopoulos N, and
Chiu RCJ. Marrow stromal cells for cellular cardiomyoplasty: Feasibility and potential clinical advantages. J Thorac Cardiovasc Surg 120:
999 –1006, 2000.
144. Wang JS, Shum-Tim D, Chedrawy E, and Chiu RCJ. The coronary
delivery of marrow stromal cells for myocardial regeneration: pathophys-
H2567