ArticlePDF AvailableLiterature Review

Spaeth E, Klopp A, Dembinski J, Andreeff M, Marini FInflammation and tumor microenvironments: defining the migratory itinerary of mesenchymal stem cells. Gene Therapy 15: 730-738

Authors:
  • Phenogen Sciences Inc

Abstract and Figures

Mesenchymal stem cells (MSC) exhibit tropism for sites of tissue damage as well as the tumor microenvironment. Many of the same inflammatory mediators that are secreted by wounds are found in the tumor microenvironment and are thought to be involved in attracting MSC to these sites. Cell migration is dependent on a multitude of signals ranging from growth factors to chemokines secreted by injured cells and/or respondent immune cells. MSC are likely to have chemotactic properties similar to other immune cells that respond to injury and sites of inflammation. Thus, the well-described model of leukocyte migration can serve as a reasonable example to facilitate the identification of factors involved in MSC migration. Understanding the factors involved in regulating MSC migration to tumors is essential to ultimately develop novel clinical strategies aimed at using MSC as vehicles to deliver antitumor proteins or suppress MSC migration to reduce tumor growth. For example, radiation enhances inflammatory signaling in the tumor microenvironment and may be used to potentiate site-specific MSC migration. Alternatively, restricting the migration of the MSC to the tumor microenvironment may prevent competent tumor-stroma formation, thereby hindering the growth of the tumor. In this review, we will discuss the role of inflammatory signaling in attracting MSC to tumors.
Content may be subject to copyright.
REVIEW
Inflammation and tumor microenvironments: defining
the migratory itinerary of mesenchymal stem cells
E Spaeth, A Klopp, J Dembinski, M Andreeff and F Marini
Molecular Hematology and Therapy, Department of Stem Cell Transplantation and Cellular Therapy, UT-M.D. Anderson Cancer Center,
Houston, TX, USA
Mesenchymal stem cells (MSC) exhibit tropism for sites of
tissue damage as well as the tumor microenvironment. Many
of the same inflammatory mediators that are secreted by
wounds are found in the tumor microenvironment and are
thought to be involved in attracting MSC to these sites. Cell
migration is dependent on a multitude of signals ranging from
growth factors to chemokines secreted by injured cells and/or
respondent immune cells. MSC are likely to have chemo-
tactic properties similar to other immune cells that respond to
injury and sites of inflammation. Thus, the well-described
model of leukocyte migration can serve as a reasonable
example to facilitate the identification of factors involved in
MSC migration.Understanding the factors involved in reg-
ulating MSC migration to tumors is essential to ultimately
develop novel clinical strategies aimed at using MSC as
vehicles to deliver antitumor proteins or suppress MSC
migration to reduce tumor growth. For example, radiation
enhances inflammatory signaling in the tumor microenviron-
ment and may be used to potentiate site-specific MSC
migration. Alternatively, restricting the migration of the MSC
to the tumor microenvironment may prevent competent
tumor-stroma formation, thereby hindering the growth of
the tumor. In this review, we will discuss the role of
inflammatory signaling in attracting MSC to tumors.
Gene Therapy (2008) 15, 730–738; doi:10.1038/gt.2008.39;
published online 10 April 2008
Keywords: mesenchymal stem cell; migration; tumor microenvironment; tumor stroma; inflammatory chemoattractants
Brief description of the mesenchymal
stem cell
Mesenchymal stem cells (MSC) are non-hematopoietic
adult stem cells with multilineage potential. MSC are
defined by plastic adherence, differentiation potential
and cell surface marker expression.
1
MSC, or MSC-like
cells, have been isolated from nearly every organ or
tissue in the body, making it challenging to characterize
the MSC as a completely homogenous population. MSC
contribute to the maintenance and regeneration of
connective tissues and have the capacity to differentiate
within osteoblasts, adipocytes, chondrocytes, myocytes
and cardiomyocytes. MSC express markers including
CD29, CD44, CD51, CD73 (SH3/4), CD105 (SH2), CD166
(ALCAM) and Stro-1, but the expression of specific
combinations of markers appear microenvironment-
dependent, suggesting a strong influence of tissue
context on MSC phenotypes. In general, MSC appear to
be a non-immunogenic population of cells; however, a
few studies demonstrate immune-repressive functions of
MSC through the induction of peripheral tolerance
evident in autoimmune disorders such as multiple
sclerosis.
2
The use of bone marrow-derived MSC have been
employed in support and engraftment of the transplan-
tation of hematopoietic stem cells (HSCs) following
high-dose chemotherapy in an effort to replenish the
destroyed bone marrow cell population.
3
Additionally,
pre-clinical studies have explored the use of MSC in the
reduction of graft-versus-host disease, for tissue repair;
including cerebral injury,
4
bone fracture,
5
myocardial
ischemia/infarction,
6
muscular dystrophy,
7
as well as
tumor homing.
Migration of MSC to tumors is thought to be due to
inflammatory signaling in a tumor resembling that of an
unresolved wound.
8
The innate tropism of MSC for
tumors can be exploited for the delivery of antitumor
agents to the tumor microenvironment. Gene-modified
MSC expressing interferon-bhave been used to signifi-
cantly reduce tumor burden and in some cases extend
survival in murine models of melanoma,
9
lung,
10
breast
cancer
11
and glioma.
12
However, the mechanism and
factors responsible for the targeted tropism of MSC to
these wounded microenvironments remain to be fully
elucidated. MSC are likely to have chemotactic proper-
ties similar to other immune cells that respond to injury
and sites of inflammation. Thus, the well-described
model of leukocyte migration can serve as a reasonable
example to facilitate the identification of factors involved
in MSC migration. Following a discussion of alterations
in the ‘wounded’/tumor microenvironment that are
shown to enhance MSC tumor-specific migration, we
will introduce alternative, injury-induced cell migration
models based on literature reviewing leukocytes and
Received 14 February 2008; accepted 18 February 2008; published
online 10 April 2008
Correspondence: Dr F Marini, Molecular Hematology and Therapy,
Department of Stem Cell Transplantation, UT-M.D. Anderson
Cancer Center, Box081, 1515 Holcombe Boulevard, Houston,
TX 77030, USA.
E-mail: Fmarini@mdanderson.org
Gene Therapy (2008) 15, 730–738
&
2008 Nature Publishing Group All rights reserved 0969-7128/08
$
30.00
www.nature.com/gt
their progenitor cell line, the HSC. These alternative
migration systems will provide rational for the tumor-
specific MSC migration and will lead us into an overview
of the current literature concerning MSC migration
specifically. Ultimately, we will conclude with a discus-
sion of the potential clinical applications of tumor-
directed MSC migration.
Inflammation-targeted homing in the
tumor microenvironment
Inflammation is a cellular response that takes place
under conditions of cellular injury and in sites of tissue
wounding. Over two decades ago, Dvorak and co-
workers described the tumor as an unhealed wound
that produces a continuous source of inflammatory
mediators (cytokines, chemokines and other potential
chemoattractant molecules). Cancer progression has
been correlated with an increase in inflammatory
mediator gene expression, and this is thought to occur
via disruption, damage and cellular turnover occurring
in the tumor microenvironment. This constant produc-
tion of inflammatory mediators perpetuates the main-
tenance and progression of the tumor environment and
becomes a target for the MSC. Tumor-generated inflam-
matory mediators have a role in determining the
conditions of the tumor microenvironment, as they
regulate invasion, motility, extracellular matrix interac-
tion through autocrine effects as well as coordinating cell
movement through paracrine signaling.
13
Tumor-pro-
duced and tumor-induced inflammatory chemokines are
known to have an important role in leukocyte/macro-
phage infiltration into tumors.
14
Based on this evidence,
one can speculate that inflammation-induced chemo-
kines participate in the directed migration of stem cells,
such as MSC, to tumors and inflamed microenviron-
ments. Previous studies have shown the importance of
inflammation to the successful homing of systematically
infused stem cells (HSC) to cardiac tissue,
15
thus reinfor-
cing the notion of inflammation and chemokine production
in migration of MSC. In addition to the secreted
chemotactic molecules secreted by the tumor and its
surrounding stroma, the tumor cells themselves retain a
chemotactic disparity amongst its cellular components.
When fractionated, the tumor cell membrane possesses a
superior chemotaxis-induction potential compared with
the other cellular components such as cytosolic fractions,
including organelles such as the nuclei, mitochondria,
lysosomes, microsomes and ribosomes (Figure 1).
Hypoxia contribution to MSC homing
Many tumors exhibit hypoxia, a state of reduced oxygen
that often parallels and perpetuates inflammation. A
common feature between inflammation and hypoxic
environments is the expression of pro-angiogenic mole-
cules. The hypoxia-induced transcription factor HIF-1a
activates the transcription of genes including vascular
endothelial growth factor (VEGF), macrophage migra-
tion inhibitory factor, tumor necrosis factors (TNF-a),
numerous proinflammatory cytokines and the activation
of the transcription factor nuclear factor kB.
16,17
Nuclear
Matrigel Migration Assay
0
50
100
150
200
250
300
350
1
# of GFP Positive Cells
24 Hour
48 Hour
72 Hour
% of Migrated MSC
100
50
C
y
tokines Tumor cells Mixtures
VEGF
PDGF
bFGF
IGF
TGFB
FBS
Live 293s
Live MDA231
Live A375sm
bFGF
Live HEY
Live SKOV3
Cell Membrane - 293s
Cell Membrane - MDA23
Cell Membrane - A375sm
Cell Membrane - OVAR3
Cell Membrane - HEY
Cell Membrane - SKOV3
bFGF & IGF
bFGF & Serum
VEGF & PDGF
PDGF & TGFB
VEGF & TGFB
Cell membrane
Figure 1 Cellular membrane components and mixtures of cytokines are potent mesenchymal stem cell (MSC) attractants. In an in vitro
matrigel assay, the migratory capacity of human MSC was analyzed. MSC preferentially migrated toward selected chemoattractive regions of
the matrigel matrix. The more effective migratory inducers include the growth factors, platelet-derived growth factor and FBS when alone. In
combination, growth factors were more potent than when used as a single chemoattractant. The attractiveness of the tumor cell membrane is
evident in comparison to other chemoattractants tested. The most potent cell membrane fraction was from the SKOV3 tumor cell lines,
followed by the MDA-231 tumor cell lines.
Inflammation-driven mesenchymal stem cell migration
E Spaeth et al
731
Gene Therapy
factor kB is frequently activated in response to inflam-
matory mediators
18
and has been shown to induce
several chemokines (RANTES (CCL5), MIP-2 (CXCL2),
MIP-1a(CCL3), monocyte chemoattractant protein 1
(MCP-1) (CCL2), interleukin-8 (CXCL8)) that are im-
plicated in leukocyte migration.
19–22
Hypoxia in the
tumor microenvironment is a cyclical event, and perpe-
tuates the inflammatory response by ensuring a constant
production of angiogenic and inflammatory mediators.
Briefly, hypoxic conditions result in the generation of
reactive oxygen species, which can increase DNA
damage in neighboring cells. Tumor cells with more
virulent mutations can then proliferate and invade
neighboring cells/tissues resulting in tissue damage,
which increases the demand for nutrients and oxygen,
which continues to be short in supply.
23
The reactive
oxygen species induced in the microenvironment has
also been demonstrated to increase secretion of inflam-
matory cytokines via nitric oxide (NO); NO has been
linked to the regulation/induction of MIP-1a, MCP-1,
macrophage inflammatory protein-related protein-1 and
osteopontin.
24,25
Hypoxia has an important role in
perpetuating the inflammatory process in tumors which
results in the generation of chemokines that are involved
in immune cell and likely MSC migration to tumors.
Archetype for MSC trafficking
The mechanism behind MSC migration is still in its
infancy. However, factors involved in regulating migra-
tion of leukocytes have been studied extensively, and it is
likely that many of the same factors are involved in
regulating MSC migration. A list of receptors expressed
on MSC that have been previously implicated in cell
migration is shown in Table 1. Growth factor, cytokine/
Table 1 Cell surface markers and receptors associated with cell migration that are known to be expressed on MSC
Cell surface receptors
found on MSC
Ligands Present on other cell types
Growth hormone
receptors
EGFR (ErbB) EGF DC, neutrophil
HGFR (c-met) HGF Leukocytes, macrophages
IGF1R IGF1 Leukocytes, HSC
PDGFR (Ra-b) PDGFa/b HSC
VEGFR1 VEGF HSC, monocytes, neutrophils
VEGFR2 VEGF HSC
FGFR2 FGF2 HSC, leukocytes
Tie-2 Ang-1 HSC, leukocytes
Chemokine/cytokine
receptors
CCR1 CCL3, CCL5, CCL7, CCL13, CCL14, CCL15,
CCL16, CCL23
monocyte, T cell, DC
CCR2 CCL2, CCL7, CCL8, CCL13, CCL16 Monocyte, T cell, DC
CCR3 CCL5, CCL7, CCL8, CCL11, CCL13, CCL15,
CCL16, CCL24, CCL26, CCL28
T cell, DC
CCR4 CCL17, CCL22 T cell, macrophage, DC
CCR5 CCL3, CCL4, CCL5, CCL8, CCL11, CCL14,
CCL16
Monocyte, T cell, DC, HSC
CCR6 CCL20 T cell, B cell, DC
CCR7 CCL19, CCL21 T cell, DC
CCR8 CCL1 monocyte, T cell, DC
CCR9 CCL25 T cell
CCR10 CCL27, CCL28 T cell
CXCR1 CXCL6, CXCL7, CXCL8 Neutrophil, monocyte
CXCR2 CXCL1, CXCL2, CXCL3, CXCL5, CXCL6,
CXCL7, CXCL8
Neutrophil, monocyte
CXCR3-A/B CXCL4, CXCL9, CXCL10, CXCL11 T cell, microvascular cells
CXCR4 CXCL12 T cell, B cell, monocyte, macrophage, DC
CXCR5 CXCL13 B cell, Th cells, HSC
CXCR6 CXCL16 CD8 T cells, NK cells, CD4 T cells
CX3CR1 CX3CL1 Macrophage
XCR1 XCL1, XCL2 T cell, NK cell
Adhesion molecules VCAM-1 (VLA-4) b1 integrin/a4 integrin Leukocytes
ICAM-1/3 LFA-1 Leukocytes, DC
ALCAM CD6 Leukocytes
Endoglin (CD105) TGFb1/3 Leukocytes, HSC
TLR1 Lipopeptides Leukocytes
Innate immune TLR2 Peptidoglycans, lipopeptides Monocytes, DC
surveillance TLR3 dsRNA DC
TLR4 LPS Monocytes, DC
TLR5 ECM molecules Monocytes
TLR6 Peptidoglycans Epithelium
The known ligands for each respective receptor are listed in the third column. The fourth column lists various cell types expressing the same
receptor.
Inflammation-driven mesenchymal stem cell migration
E Spaeth et al
732
Gene Therapy
chemokine, adhesion molecules and toll-like receptors
(TLRs) are expressed on MSC, as described by Cham-
berlain et al.,
26
Ringe et al.,
27
Allen et al.
28
and Viola et al.
29
Although the table does not encompass all receptors, it is
a comprehensive list of those receptors that have been
studied on other cell types, including leukocytes and
HSC. The T cell, macrophage and dendritic cells are all
considered relevant to the understanding of MSC
homing because of their functional similarities in
targeting inflamed/injured tissues. The HSC is consid-
ered because it is the precursor to the myeloid and
lymphoid lineages in addition to being resident of the
bone marrow where we also find the MSC.
Leukocytes
There are three main participants in leukocyte traffick-
ing: adhesion molecules, integrins and chemoattrac-
tants.
30
The latter is the most important when
considering the paracrine-mediated gradient trafficking
and migration without cellular contact, such as what has
been observed for MSC migration to sites of tumors and
sites of injury.
Cytokine and chemokine receptors have an important
role in leukocyte and likely MSC migration. A decrease
in leukocyte migration has been observed in a mouse
knockout model of the IL-8 receptors, CXCR1 and
CXCR2.
31
Similarly, neutrophil recruitment in ischemia-
reperfusion models was inhibited by blocking the
CXCR1 and CXCR2 receptors.
32
The presence of both
CXCR1 and CXCR2
27
on MSC suggests that they may
have a similar function in MSC migration. Nearly every
chemokine receptor has been found on the surface of
MSC,
3
while CCR2 and CCR3 are two receptors that may
have a particularly important role in leukocyte and MSC
trafficking. Macrophage trafficking has been shown to be
mediated by CCR2, a chemokine receptor with ligands
including MCP1, 2, 3 and 4 (MCP-1, 2, 3, 4, or CCL2, 8, 7,
13, respectively).
30
Of note, blocking CCR3, the receptor
for eotaxtin (CCL11), RANTES (CCL5), MCP2,
MCP3 and MCP4 (CCL 8, 7, 13) has been effective in
reducing trafficking in leukocytes. Yet another seven
transmembrane receptor, CD97, an epidermal growth
factor receptor appears necessary for the migration of
neutrophils to sites of inflammation
33
and has been
implicated in the promotion of angiogenesis. Increased
expression of CD97 in tumor cell lines, such as colorectal
carcinoma, correlates with the increased migration
potential of leukocytes.
34
Of note, MSC express CD55,
the ligand to CD97, suggesting a potential CD55–CD97
interaction thereby influencing both MSC and leukocyte
migration.
Other than cytokines/chemokines, numerous bioac-
tive molecules can also serve as leukocyte chemoattrac-
tants. Such molecules include lipids, (leukotrienes and
prostaglandins), peptides such as chemerin and other
elements of the extracellular matrix.
35
The adhesion
molecule, CD44, which is widely expressed on leuko-
cytes and parenchymal cells interacts with components
of the extracellular matrix known to be involved in
inflammation including hyaluronic acid, collagen, lami-
nin and fibronectin. The presence of CD44 is thought to
mediate and enhance localized inflammation leading to
the increased migration of leukocytes, it also serves as a
receptor for growth factors, integrins, cytokines (osteo-
pontin), glycosaminoglycans (hyaluronic acid), peptides
(collagen), and is able to both induce inflammation and
serve as a receptor mediator in the homing toward sites
of inflammation.
36,37
Hyaluronan, which is a by-product
of tissue repair, results in persistent inflammation
38
and
in addition to being critical to cell motility during
development, the inhibition of hyaluronan synthesis in
prostate tumors impairs growth and vascularization.
39
Of importance, CD44, the receptor for hyaluronan, is
expressed on MSC, and a recent publication suggests
CD44 roles in the migration of MSC toward injured
kidney tissue.
40
In addition to CD44, hyaluronan by itself
can interact with TLR2 and TLR4, thereby enhancing the
inflammatory response.
38
The participation and involve-
ment of TLRs in MSC migration will be addressed to a
further extent in a subsequent section.
HSC migration
Similar to the leukocytes, the migratory machinery of
HSC are also well characterized in regards to defined
receptors/ligands required for migration and should be
considered in the evaluation of potential candidates for
the elucidation of MSC migration. In fact, one of the most
widely recognized receptor/ligand pairs for HSC traf-
ficking is CXCR4/CXCL12 (SDF1); in addition, CXCR4/
SDF1 appears important in dictating migration of several
tumor cell lines to metastatic sites.
41,42
However, in
contrast to the key role of CXCR4/SDF1 in HSC
migration, a recent publication by Ip and co-workers
showed blocking of the CXCR4 receptor had no impact
on MSC migration, suggesting key differences
between the migration signals of these two stem cells.
43
This controversial receptor indicates that receptor
function differs between cell types and enhances the
importance of examining receptors found mutually
expressed on MSC- and HSC-like CXCR4, CCR5
44
and VEGFR.
45
MSC trafficking—finding the tumor
The past year has revealed a surge of publications
attempting to define the homing properties of MSC. Due
to the more comprehensive command of knowledge in
the field of leukocyte migration, those chemokines and
corresponding receptors chosen for evaluation on MSC is
based on prior data illustrated in leukocyte models.
However, as noted above, the migratory competence
of these receptors clearly varies: CXCR4/CXCL12 are
crucial in bone marrow retention and homing of HSC
where as in MSC, it appears that CXCR4/CXCL12 do not
posses the same migration importance unless enforced
expression of CXCR4 is employed.
43,46,47
Bearing in mind
that each of the following receptors and their respective
ligands (chemokines, cytokines, growth factors, peptides
small biomolecules) have not been implicated as a single
primary mechanism for the migration of MSC; however,
altogether may function in an additive manner. Thus, the
coalescence of known migratory mechanisms is vital to
the understanding of the complete migratory disposition
of the MSC in relationship to cancer biology.
Preconditioning the MSC
Activation of MSC with proinflammatory cytokines (that
is, TNF-a) prior to reinfusion has been demonstrated to
increase MSC in vivo migratory and adhesion capacity
Inflammation-driven mesenchymal stem cell migration
E Spaeth et al
733
Gene Therapy
through the increased expression of receptors. Existing
data suggest that the cytokines IL-1band TNF-aactivate
adherence properties of the MSC including the upregula-
tion of the VCAM-1-VLA-4 adhesion pathway.
48
Addi-
tional receptors known to be upregulated by TNF-a
priming include CCR3 and CCR4; these findings correlate
with the observation of increased in vitro migration to
RANTES (a CCR3 ligand) and macrophage-derived
cytokine (MDC (CCL22)—a CCR4 ligand).
3
Several growth factors have been shown to induce
MSC migration. Insulin-like growth factor 1 (IGF-1)
increases the expression of chemokine receptors on the
MSC, thereby enhancing migration. Li and co-workers
demonstrated that IGF-1-induced upregulation of
CXCR4 expression on MSC increased the migratory
capacity of the cells toward an in vitro SDF-1 gradient
through a phosphoinositide-3 kinase-dependent path-
way without altering the proliferation status of the
MSC.
49
The inability for MSC migration to occur in vivo
through a CXCL12/CXCR4 mechanism was mentioned
previously; accordingly, IGF-1 is capable of stimulating
the expression of other chemokine receptors such as
CCR5—the RANTES (CCL5) receptor.
50
Other growth
factors including basic fibroblast growth factor and
VEGF are associated with angiogenesis and are secreted
under hypoxemic stress. MSC demonstrate an increased
migratory propensity in the presence of basic fibroblast
growth factor through a phosphoinositide-3 kinase/AKT
pathway downstream of the basic fibroblast growth
factor receptor on the MSC.
51
Chemokine/cytokine secretion and the respective
receptors found on MSC
Monocyte-chemoattractant protein 1 (CCL2), a chemo-
kine secreted by tumor cells, was shown to be a potent
chemoattractant for MSC migration toward breast
carcinomas.
52
However, prior studies show conflicting
data on the migratory response to CCL2. The incon-
sistent results may be attributed to differences between
primary cells and passaged cell lines.
27,53
Additionally,
Dwyer and co-workers examined the expression of CCL2
in breast-tumor explants revealing the cytokine presence
not only in the tumor cells, but also surprisingly showing
that the majority of the CCL2 detected was from the
stromal fibroblast population.
27
This finding further
enforces the importance of the tumor microenvironment
participation in MSC migration as well as the tumor cells
themselves.
The cytokines, VEGFaand platelet-derived growth
factor ab (PDGFab) both harness chemoattractant prop-
erties; Ball and co-workers showed that VEGFawas able
to stimulate migration through the PDGF receptor,
confirming the intricacies involved in the induction of
signaling pathways.
54
In support of this, data from our
group demonstrate the combined potential of PDGF and
VEGF acting as chemoattractants inducing MSC migra-
tion in an in vitro matrigel migration assay; this
combination of growth factors is more potent than either
growth factor alone (Figure 1).
Inflammatory chemokine receptor expression on MSC
is influenced by microenvironmental conditions. For
example, the CC-, but not CXC-, chemokine receptors
have been shown to be upregulated by TNF-a.
3
This
upregulation of certain chemokine receptors in response
to cellular signals may have a role in tissue-specific
homing.
3,27,55
Additionally, the cytokines produced by
the MSC themselves may not have a direct role in MSC
migration, but may increase adhesion molecule expres-
sion in preparation for contact with the site of inflamma-
tion/injury as a mechanism of self-conditioning. In vitro
co-culture experiments demonstrate that tumor-secreted
factors influence expression of chemokines and chemo-
kine receptors in MSC (Figure 2). Furthermore, different
tumor cell lines produce different patterns of gene
expression in MSC (Figure 3). Further investigation of
these differentially expressed factors may shed light on
mechanisms of MSC migration to tumors.
Additional receptors implicated in MSC migration
Toll-like receptors are a vital component of the innate
immune response. There are 11 TLR found in humans,
each recognize a conserved yet broad range of molecules
known as pathogen-associated molecular patterns.
Depending on the stimulation, downstream TLR
signaling can regulate the expression of both CC and
CXC chemokines via nuclear factor kB activation.
56
Motif
recognition varies between receptors: the dimers, TLR1/
2 and 2/6 recognize lipopeptides and peptidoglycans;
TLR3 recognizes double-stranded RNA; TLR4 recognizes
lipopolysaccharide; TLR5 recognizes extracellular matrix
molecules; TLR7 and 8 recognize synthetic antiviral
compounds; TLR9 recognizes unmethylated CpG
DNA.
57
TLR signaling results in the activation of several
pathways, MAPK, MyD88, c-Jun N-terminal kinase and
inhibitor of kB kinase leading to the activation of the
downstream activation of transcription factors nuclear
factor kB and AP-1, which ultimately lead to the
transcription of proinflammatory chemokines and cyto-
kines.
57
TLR1–6 have been identified on primary human
MSC by both reverse transcription-PCR and flow
1.E-05
1.E-04
1.E-03
1.E-02
1.E-01
1.E+00
Lo
g
2
(
Fold Difference
)
p Value
-7 -5 -3 -1 1 5 937
Figure 2 Tumor-induced changes in mesenchymal stem cell (MSC)
inflammatory gene expression after 24 h. Expression of 84 genes
involved in inflammation was compared to human MSC
co-cultured with a pancreatic tumor cell line. Individual genes
are plotted as a function of P-value and fold difference in
expression. The blue line represents the P¼0.05 threshold. The
pink lines represent a threshold for fold difference (>2). Seven
chemokine and inflammatory cytokines, shown within the red line,
exhibited a significant increase in expression after non-contact
co-culture conditions with the tumor cell line. See online version
for colour figure.
Inflammation-driven mesenchymal stem cell migration
E Spaeth et al
734
Gene Therapy
cytometry.
58
Tomchuck and co-workers reported that
TLR stimulation enhanced the migratory function of
MSC; the most potent migratory induction occurred with
the stimulation of TLR3. Likewise, the inhibition of TLR3
signaling through a neutralizing antibody decreased
MSC migration capacity by over 50%.
0.00
0.00
0.01
0.10
1.00
10.00
100.00
1000.00
Fold Difference
1
2
3
4
5
6
7
8
9
10
11
12
Column ABCDEFGH
Row
0.01
0.10
1.00
10.00
100.00
1000.00
Fold Difference
1
2
3
4
5
6
7
8
9
10
11
12
Column ABCDEFGH
Row
0.01
0.10
1.00
10.00
100.00
1000.00
Fold Difference
1
2
3
4
5
6
7
8
9
10
11
12
Column
ABCDEFGH
Row
0.00
0.01
0.10
1.00
10.00
100.00
1000.00
Fold Difference
1
2
3
4
5
6
7
8
9
10
11
12
Column
ABCDEFGH
Row
Figure 3 Tumor-specific induction of mesenchymal stem cell (MSC) gene expression. Represented by graphs are the gene expression profiles
from real-time reverse transcription-PCR analysis on 84 inflammatory cytokines and chemokine genes expressed in human MSC. hMSC were
co-cultured with a tumor cell line for 24 h before RNA was extracted for the expression analysis comparison between the MSCs alone and the
MSC in the presence of the tumor paracrine factors. (a) Pediatric sarcoma: RD54; (b) pancreatic cancer: PANC1; (c) breast cancer: MDA-231;
(d) ovarian cancer: SKOV-3.
Renilla luciferase
4T1 tumor
Firefly luciferase
MSC
0
5000
10000
15000
20000
25000
30000
35000
40000
45000
50000
left
(irradiated)
right
(unirradiated)
counts
Figure 4 Targeted tropism of mesenchymal stem cell (MSC) using focal irradiation. (a) Ten days after 4T1 tumors were implanted into the
bilateral hindlimbs of Balb/C mice, tumors were imaged with renilla luciferase (upper panel). The right hindlimb was irradiated with 2 Gy
(approximate irradiated volume represented by white triangle). MSC were injected i.v. the following day and imaged with firefly luciferase
48 h later (lower panel). (b) Quantitation of MSC in tumors.
Inflammation-driven mesenchymal stem cell migration
E Spaeth et al
735
Gene Therapy
Manipulation of MSC migration
Augmentation of MSC migration via genetic
manipulation of MSC
With the mechanistic understanding behind MSC migra-
tion being slowly deciphered, the potential for enhancing
MSC-targeted therapies appears promising. The genetic
manipulation of MSC to overexpress target receptors
should enhance their migration to site-specific locations.
The introduction of exogenous DNA into MSC, as
reviewed by Damme and co-workers, enables enforced
expression/secretion of a desired therapeutic factor into
the targeted environment.
59
One could imagine the
potential in the overexpression of TLR3 and CCR2, the
receptor for CCL2 (MCP-1) on MSC may improve their
migration efficiency to specific tumor cells. Such a
specific and directed approach will prove promising to
the field of gene therapy for the treatment of cancers,
allowing the cellular targeting of specific tumors,
inflammatory diseases and other tissue injuries including
myocardial infarction and ischemic cerebral damage.
The diverse collection of receptors present on MSC
suggest the formation of a cascade of competitive events
that enable a hierarchy of chemoattractants that are
responsible for a step-wise chemotaxis to the tumor
microenvironment. Conflicting chemotactic signals will
lead to a biased migration to previously encountered
chemoattractants in leukocyte-trafficking models.
60
This
heterologous signaling pathway activation based on the
variation of surface receptors expression may justify both
the preconditioning of MSC as discussed previously and
the manipulation of surface receptor expression to
enhance chemotaxis.
Alteration of MSC migration via changes in external
environment
As discussed previously, inflammatory mediators have
been shown to increase targeted trafficking. Inflamma-
tion induction as an element of targeted treatment
enables a broader scope of secreted inflammatory
molecules that may influence migration. This global
enhancement of inflammatory chemokines, cytokines,
along with tissue damage by-products including, lipids
(leukotriene), glycosaminoglycans (hyaluran), enzymes,
free radicals, complement and fibrinopeptides will
exacerbate the migratory response seen in MSC. Mice
with irradiated tumors, as compared with unirradiated
tumors, show an increase in MSC migration. Klopp and
co-workers recently demonstrated this in an elegant
experiment upon which bilateral hind leg tumor im-
plants were used. The tumor in the left hind leg
remained an internal control, whereas the right hind
leg received local irradiation treatment after which
intravenous injection of MSC revealed a higher number
of MSC localizing to the irradiated tumor (Figure 4).
Similar experiments using HSC have shown an increased
migratory propensity post-irradiation treatment of glio-
ma cells. The attraction of HSC to irradiated glioma cells
was attributed to an increase in stress signaling that
induced hypoxia-induced transcription factor 1atran-
scriptional activity dependent on a functional TGF-b
signaling cascade to induce CXCL12 promoter activity.
61
Low-dose irradiation promotes VEGF release that is
dependent on matrix metalloproteinase 9 expression,
which is also upregulated under stress responses such as
radiation treatment.
62
The angiogenic factor, VEGF, is
secreted by both the tumor cells and the tumor stroma,
and is a known migratory inducer of both HSC and
MSC.
63,64
In the presence of irradiated tumor cell-
conditioned media, MSC increase their expression of
VEGFa, VEGFc and PDGFb. The expression of these
growth factors may be an autocrine feedback loop
mechanism to enhance receptor expression, or to counter
balance an increased apoptotic mRNA expression seen in
these cells.
Future directions
Targeted migration of MSC to tumor sites will have a
significant impact on the field of antitumor therapy. MSC
exhibit an intrinsic homing property enabling them to
direct migration to sites of inflammation. The exploita-
tion of this process will be a valuable asset to directed
therapy. Their capability to express exogenous gene
products, genetic stability and allogeneic properties
make MSC excellent delivery vehicles for antitumor
therapy, previously demonstrated not only in tumor
models but also for other diseases such as graft-versus-
host, multiple sclerosis and arthritis.
9,65,66
The induced expression of receptors critical to the
migratory competence of MSC to tumors will allow an
increased number of MSC to reach the target location.
The increase in migratory efficiency will improve the
therapeutic value of the overall system.
Conclusions
There have been many advances in determining the
factors involved in the migration of MSC to ‘wounded’/
inflammatory/tumor environments; however, their full
potential as therapeutic-vehicle candidates can only be
utilized when the mechanistic understanding behind
their migration is elucidated. Many different receptors
have been implicated in the homing of MSC: (1) the
broad activation of growth factor receptors that activate
further chemokine receptor expression like CXC and CC
receptors; (2) the activation of TLR that also target
downstream expression of CXC and CC receptors; (3) the
activation of adhesion molecules and (4) integrins that
may or may not be implicated in the direct role of
paracrine cell movement. The key players implicated
in MSC migration to date include the chemokines
MCP-1 (CCL2),
52
CXCL8,
27
RANTES (CCL5);
3
LL-37,
58
integrinb1,
43
receptors CD44,
40
CCR2,
3
CCR3,
3
and the
receptor tyrosine kinases for the following growth
factors, IGF-1,
3
PDGF-bb,
67
HGF,
68
and VEGF.
54
The ability to extract from previously described cell-
migration models in the elucidation of MSC homing
properties is an arduous task. Many common receptors
have been identified on MSC; however, multiple ligands
and co-receptors have the ability to alter the downstream
signaling pathway through coupling, crosstalk or in-
hibitory mechanisms thereby rendering an alternative
mechanistic feature for a common ligand/receptor pair
depending on the cell-lineage. The future of targeted
therapy using MSC will depend on the exploitation of
these previous ligand/receptor interactions for the
Inflammation-driven mesenchymal stem cell migration
E Spaeth et al
736
Gene Therapy
enhancement of the existing intrinsic migratory/homing
propensity.
Acknowledgements
This work was supported in part by grants from the
National Cancer Institute (CA-1094551 and CA-116199
for FCM, CA-55164, CA-16672, and CA-49639 for MA)
and by the Paul and Mary Haas Chair in Genetics (MA).
ES, AK and FCM are supported in part by grants from
the Susan G Komen Breast Cancer Foundation.
References
1 Prockop DJ. Marrow stromal cells as stem cells for nonhemato-
poietic tissues. Science 1997; 276: 71–74.
2 Zappia E, Casazza S, Pedemonte E, Benvenuto F, Bonanni I,
Gerdoni E et al. Mesenchymal stem cells ameliorate experi-
mental autoimmune encephalomyelitis inducing T-cell anergy.
Blood 2005; 106: 1755–1761.
3 Lopez Ponte A, Marais E, Gallay N, Langonne
´A, Delorme B,
He
´rault O et al. The in vitro migration capacity of human bone
marrow mesenchymal stem cells: comparison of chemokine and
growth factor chemotactic activities. Stem Cells 2007; 25: 1737–1745.
4 Li Y, Chen J, Wang L, Zhang L, Lu M, Chopp M. Intracerebral
transplantation of bone marrow stromal cells in a 1-methyl-4-
phenyl-1,2,3,6-tetrahydropyridine mouse model of parkinson’s
disease. Neurosci Lett 2001; 316: 67–70.
5 Niedzwiedzki T, Dabrowski Z, Miszta H, Pawlikowski M. Bone
healing after bone marrow stromal cell transplantation to the
bone defect. Biomaterials 1993; 14: 115–121.
6 Kawada H, Fujita J, Kinjo K, Matsuzaki Y, Tsuma M, Miyatake H
et al. Nonhematopoietic mesenchymal stem cells can be
mobilized and differentiate into cardiomyocytes after myocar-
dial infarction. Blood 2004; 104: 3581–3587.
7 Dezawa M, Ishikawa H, Itokazu Y, Yoshihara T, Hoshino M,
Takeda S et al. Bone marrow stromal cells generate muscle cells
and repair muscle degeneration. Science 2005; 309: 314–317.
8 Dvorak HF. Tumors: wounds that do not heal. Similarities
between tumor stroma generation and wound healing. N Engl J
Med 1986; 315: 1650–1659.
9 Studeny M, Marini FC, Dembinski JL, Zompetta C, Cabreira-
Hansen M, Bekele BN et al. Mesenchymal stem cells: potential
precursors for tumor stroma and targeted-delivery vehicles for
anticancer agents. J Natl Cancer Inst 2004; 96: 1593–1603.
10 Kanehira M, Xin H, Hoshino K, Maemondo M, Mizuguchi H,
Hayakawa T et al. Targeted delivery of NK4 to multiple lung
tumors by bone marrow-derived mesenchymal stem cells.
Cancer Gene Ther 2007; 14: 894–903.
11 Hall B, Dembinski J, Sasser AK, Studeny M, Andreeff M,
Marini F. Mesenchymal stem cells in cancer: tumor-associated
fibroblasts and cell-based delivery vehicles. Int J Hematol 2007; 86:
8–16.
12 Nakamizo A, Marini F, Amano T, Khan A, Studeny M, Gumin J
et al. Human bone marrow-derived mesenchymal stem cells in
the treatment of gliomas. Cancer Res 2005; 65: 3307–3318.
13 Balkwill F. Cancer and the chemokine network. Nat Rev Cancer
2004; 4: 540–550.
14 Zernecke A, Weber KSC, Erwig LP, Kluth DC, Schro
¨ppel B, Rees
AJ et al. Combinatorial model of chemokine involvement in
glomerular monocyte recruitment: role of CXC chemokine
receptor 2 in infiltration during nephrotoxic nephritis. J Immunol
2001; 166: 5755–5762.
15 Malek S, Kaplan E, Wang JF, Ke Q, Rana JS, Chen Y et al.
Successful implantation of intravenously administered stem
cells correlates with severity of inflammation in murine
myocarditis. Pflug Arch Eur J Physiol 2006; 452: 268–275.
16 Winner M, Koong AC, Rendon BE, Zundel W, Mitchell RA.
Amplification of tumor hypoxic responses by macrophage
migration inhibitory factor-dependent hypoxia-inducible factor
stabilization. Cancer Res 2007; 67: 186–193.
17 Ye J, Gao Z, Yin J, He Q. Hypoxia is a potential risk factor for
chronic inflammation and adiponectin reduction in adipose
tissue of ob/ob and dietary obese mice. Am J Physiol Endocrinol
Metab 2007; 293: E1118–E1128.
18 Pold M, Zhu LX, Sharma S, Burdick MD, Lin Y, Lee PP et al.
Cyclooxygenase-2-dependent expression of angiogenic CXC
chemokines ENA-78/CXC ligand (CXCL) 5 and interleukin-8/
CXCL8 in human non-small cell lung cancer. Cancer Res 2004; 64:
1853–1860.
19 Ubogu EE, Callahan MK, Tucky BH, Ransohoff RM. Determi-
nants of CCL5-driven mononuclear cell migration across the
blood-brain barrier. Implications for therapeutically modulating
neuroinflammation. J Neuroimmunol 2006; 179: 132–144.
20 Gregory JL, Morand EF, McKeown SJ, Ralph JA, Hall P, Yang YH
et al. Macrophage migration inhibitory factor induces macro-
phage recruitment via CC chemokine ligand 2. J Immunol 2006;
177: 8072–8079.
21 Montecucco F, Bianchi G, Bertolotto M, Viviani G, Dallegri F,
Ottonello L. Insulin primes human neutrophils for CCL3-
induced migration: crucial role for JNK 1/2. Ann N Y Acad Sci
2006; 1090: 399–407.
22 Lehman N, Di Fulvio M, McCray N, Campos I, Tabatabaian F,
Gomez-Cambronero J. Phagocyte cell migration is mediated by
phospholipases PLD1 and PLD2. Blood 2006; 108: 3564–3572.
23 Laconi E. The evolving concept of tumor microenvironments.
BioEssays 2007; 29: 738–744.
24 Guo HT, Cai CQ, Schroeder RA, Kuo PC. Nitric oxide is
necessary for CC-class chemokine expression in endotoxin-
stimulated ANA-1 murine macrophages. Immunol Lett 2002; 80:
21–26.
25 Takahashi F, Takahashi K, Maeda K, Tominaga S, Fukuchi Y.
Osteopontin is induced by nitric oxide in RAW 264.7 cells.
IUBMB Life 2000; 49: 217–221.
26 Chamberlain G, Fox J, Ashton B, Middleton J. Concise review:
mesenchymal stem cells: their phenotype, differentiation capa-
city, immunological features, and potential for homing. Stem
Cells 2007; 25: 2739–2749.
27 Ringe J, Strassburg S, Neumann K, Endres M, Notter M,
Burmester G-R. Towards in situ tissue repair: human mesench-
ymal stem cells express chemokine receptors CXCR1, CXCR2
and CCR2, and migrate upon stimulation with CXCL8 but not
CCL2. J Cell Biochem 2007; 101: 135–146.
28 Allen SJ, Crown SE, Handel TM. Chemokine: Receptor structure,
interactions, and antagonism. Annu Rev Immunol 2007; 25:
787–820.
29 Viola A, Luster AD. Chemokines and their receptors: drug
targets in immunity and inflammation. Annu Rev Pharmacol
Toxicol 2008; 48: 171–197.
30 Luster AD, Alon R, von Andrian UH. Immune cell migration in
inflammation: present and future therapeutic targets. Nat
Immunol 2005; 6: 1182–1190.
31 Becker MD, O’Rourke LM, Blackman WS, Planck SR, Rosen-
baum JT. Reduced leukocyte migration, but normal rolling and
arrest, in interleukin-8 receptor homologue knockout mice.
Invest Ophthalmol Vis Sci 2000; 41: 1812–1817.
32 Sekido N, Mukaida N, Harada A, Nakanishi I, Watanabe Y,
Matsushima K. Prevention of lung reperfusion injury in rabbits
by a monoclonal antibody against interleukin-8. Nature 1993;
365: 654–657.
33 Leemans JC, te Velde AA, Florquin S, Bennink RJ, de Bruin K, van
Lier RA et al. The epidermal growth factor-seven transmembrane
Inflammation-driven mesenchymal stem cell migration
E Spaeth et al
737
Gene Therapy
(EGF-TM7) receptor CD97 is required for neutrophil migration
and host defense. JImmunol2004; 172: 1125–1131.
34 Galle J, Sittig D, Hanisch I, Wobus M, Wandel E, Loeffler M et al.
Individual cell-based models of tumor-environment interac-
tions: multiple effects of CD97 on tumor invasion. Am J Pathol
2006; 169: 1802–1811.
35 Zabel BA, Zuniga L, Ohyama T, Allen SJ, Cichy J, Handel TM et
al. Chemoattractants, extracellular proteases, and the integrated
host defense response. Exp Hematol 2006; 34: 1021–1032.
36 Pisterna GV, Siragusa M. CD44 presence in inflamed pulp tissue.
J Endodontics 2007; 33: 1203–1207.
37 Chaulet H, Desgranges C, Renault MA, Dupuch F, Ezan G,
Peiretti F et al. Extracellular nucleotides induce arterial
smooth muscle cell migration via osteopontin. Circ Res 2001;
89: 772–778.
38 Jiang D, Liang J, Noble PW. Hyaluronan in tissue injury and
repair. Annu Rev Cell Dev Biol 2007; 23: 435–461.
39 Simpson MA, Wilson CM, McCarthy JB. Inhibition of prostate
tumor cell hyaluronan synthesis impairs subcutaneous growth
and vascularization in immunocompromised mice. Am J Pathol
2002; 161: 849–857.
40 Herrera MB, Bussolati B, Bruno S, Morando L, Mauriello-
Romanazzi G, Sanavio F et al. Exogenous mesenchymal stem
cells localize to the kidney by means of CD44 following acute
tubular injury. Kidney Int 2007; 72: 430–441.
41 Wright DE, Bowman EP, Wagers AJ, Butcher EC, Weissman IL.
Hematopoietic stem cells are uniquely selective in their
migratory response to chemokines. J Exp Med 2002; 195:
1145–1154.
42 Sutton A, Friand V, Brule-Donneger S, Chaigneau T, Ziol M,
Sainte-Catherine O et al. Stromal cell-derived factor-1/
chemokine (C-X-C motif) ligand 12 stimulates human hepatoma
cell growth, migration, and invasion. Mol Cancer Res 2007; 5:
21–33.
43 Ip JE, Wu Y, Huang J, Zhang L, Pratt RE, Dzau VJ. Mesenchymal
stem cells use integrin beta1 not CXC chemokine receptor 4 for
myocardial migration and engraftment. Mol Biol Cell 2007; 18:
2873–2882.
44 Schwabe RF, Bataller R, Brenner DA. Human hepatic stellate
cells express CCR5 and RANTES to induce proliferation
and migration. Am J Physiol Gastrointest Liver Physiol 2003; 285:
G949–G958.
45 Novo E, Cannito S, Zamara E, Valfre
`di Bonzo L, Caligiuri A,
Cravanzola C et al. Proangiogenic cytokines as hypoxia-
dependent factors stimulating migration of human hepatic
stellate cells. Am J Pathol 2007; 170: 1942–1953.
46 Le
´vesque JP, Hendy J, Takamatsu Y, Simmons PJ, Bendall LJ.
Disruption of the CXCR4/CXCL12 chemotactic interaction
during hematopoietic stem cell mobilization induced by gcsf
or cyclophosphamide. J Clin Invest 2003; 111: 187–196.
47 Shi M, Li J, Liao L, Chen B, Li B, Chen L et al. Regulation of
CXCR4 expression in human mesenchymal stem cells by
cytokine treatment: Role in homing efficiency in NOD/SCID
mice. Haematologica 2007; 92: 897–904.
48 Segers VFM, VanRiet I, Andries LJ, Lemmens K, Demolder MJ,
De Becker AJ et al. Mesenchymal stem cell adhesion to cardiac
microvascular endothelium: activators and mechanisms. Am J
Physiol Heart Circ Physiol 2006; 290: H1370–H1377.
49 Li Y, Yu X, Lin S, Li X, Zhang S, Song YH. Insulin-like growth
factor 1 enhances the migratory capacity of mesenchymal stem
cells. Biochem Biophys Res Commun 2007; 356: 780–784.
50 Mira E, Lacalle RA, Gonza
´lez MA, Go
´mez-Mouto
´n C, Abad JL,
Bernad A et al. A role for chemokine receptor transactivation in
growth factor signaling. EMBO Rep 2001; 2: 151–156.
51 Schmidt A, Ladage D, Schinkothe T, Klausmann U, Ulrichs C,
Klinz FJ et al. Basic fibroblast growth factor controls migration
in human mesenchymal stem cells. Stem Cells 2006; 24:
1750–1758.
52 Dwyer RM, Potter-Beirne SM, Harrington KA, Lowery AJ,
Hennessy E, Murphy JM et al. Monocyte chemotactic protein-1
secreted by primary breast tumors stimulates migration of
mesenchymal stem cells. Clin Cancer Res 2007; 13: 5020–5027.
53 Wang L, Li Y, Chen J, Gautam SC, Zhang Z, Lu M et al. Ischemic
cerebral tissue and MCP-1 enhance rat bone marrow stromal cell
migration in interface culture. Exp Hematol 2002; 30: 831–836.
54 Ball SG, Shuttleworth CA, Kielty CM. Vascular endothelial
growth factor can signal through platelet-derived growth factor
receptors. J Cell Biol 2007; 177: 489–500.
55 Honczarenko M, Le Y, Swierkowski M, Ghiran I, Glodek AM,
Silberstein LE. Human bone marrow stromal cells express a
distinct set of biologically functional chemokine receptors. Stem
Cells 2006; 24: 1030–1041.
56 Boyd JH, Divangahi M, Yahiaoui L, Gvozdic D, Qureshi S, Petrof
BJ. Toll-like receptors differentially regulate CC and CXC
chemokines in skeletal muscle via NF-kappaB and calcineurin.
Infect Immun 2006; 74: 6829–6838.
57 West AP, Koblansky AA, Ghosh S. Recognition and signaling by
toll-like receptors. Annu Rev Cell Dev Biol 2006; 22: 409–437.
58 Tomchuck SL, Zwezdaryk KJ, Coffelt SB, Waterman RS, Danka
ES, Scandurro AB. Toll-like receptors on human mesenchymal
stem cells drive their migration and immunomodulating
responses. Stem Cells 2007; 26: 99–107.
59 Hamada H, Kobune M, Nakamura K, Kawano Y, Kato K,
Honmou O et al. Mesenchymal stem cells (MSC) as therapeutic
cytoreagents for gene therapy. Cancer Sci 2005; 96: 149–156.
60 Foxman EF, Kunkel EJ, Butcher EC. Integrating conflicting
chemotactic signals: the role of memory in leukocyte navigation.
J Cell Biol 1999; 147: 577–587.
61 Tabatabai G, Frank B, Mohle R, Weller M, Wick W. Irradiation
and hypoxia promote homing of haematopoietic progenitor
cells towards gliomas by TGF-beta-dependent HIF-1alpha-
mediated induction of CXCL12. Brain 2006; 129 (Pt 9):
2426–2435.
62 Heissig B, Rafii S, Akiyama H, Ohki Y, Sato Y, Rafael T et al.
Low-dose irradiation promotes tissue revascularization
through VEGF release from mast cells and MMP-9-
mediated progenitor cell mobilization. J Exp Med 2005; 202:
739–750.
63 Rabbany SY, Heissig B, Hattori K, Rafii S. Molecular pathways
regulating mobilization of marrow-derived stem cells for tissue
revascularization. Trends Mol Med 2003; 9: 109–117.
64 Schichor C, Birnbaum T, Etminan N, Schnell O, Grau S, Miebach
Set al. Vascular endothelial growth factor A contributes to
glioma-induced migration of human marrow stromal cells
(hMSC). Exp Neurol 2006; 199: 301–310.
65 Kim SK, Kim SU, Park IH, Bang JH, Aboody KS, Wang KC et al.
Human neural stem cells target experimental intracranial
medulloblastoma and deliver a therapeutic gene leading to
tumor regression. Clin Cancer Res 2006; 12: 5550–5556.
66 Evans CH, Robbins PD, Ghivizzani SC, Wasko MC, Tomaino
MM, Kang R et al. Gene transfer to human joints: progress
toward a gene therapy of arthritis. Proc Natl Acad Sci USA 2005;
102: 8698–8703.
67 Fiedler J, Ro
¨derer G, Gu
¨nther KP, Brenner RE. BMP-2, BMP-4,
and PDGF-bb stimulate chemotactic migration of primary
human mesenchymal progenitor cells. J Cell Biochem 2002; 87:
305–312.
68 Forte G, Minieri M, Cossa P, Antenucci D, Sala M, Gnocchi V
et al. Hepatocyte growth factor effects on mesenchymal stem
cells: proliferation, migration, and differentiation. Stem Cells
2006; 24: 23–33.
Inflammation-driven mesenchymal stem cell migration
E Spaeth et al
738
Gene Therapy
... Notably, MSC-derived extracellular vesicles (EVs) have also been found to facilitate immunosuppression, promote M2 macrophage polarization, and induce Treg cell formation . TME produce inflammatory reactions to recruit MSCs (Spaeth et al., 2008). Tumors secrete a variety of chemokines and cytokines, which have been demonstrated to engage with receptors on MSCs, including stromal cell-derived factor-1(SDF-1), epidermal growth factor (EGF), PDGF, monocyte chemotactic protein-1 (MCP-1), IL-8, and IL-1β, and TNF-α. ...
Article
Full-text available
Cholangiocarcinoma (CCA) is a highly aggressive malignant tumor that originates from the biliary system. With restricted treatment options at hand, the challenging aspect of early CCA diagnosis leads to a bleak prognosis. Besides the intrinsic characteristics of tumor cells, the generation and progression of CCA are profoundly influenced by the tumor microenvironment, which engages in intricate interactions with cholangiocarcinoma cells. Of notable significance is the role of extracellular vesicles as key carriers in enabling communication between cancer cells and the tumor microenvironment. This review aims to provide a comprehensive overview of current research examining the interplay between extracellular vesicles and the tumor microenvironment in the context of CCA. Specifically, we will emphasize the significant contributions of extracellular vesicles in molding the CCA microenvironment and explore their potential applications in the diagnosis, prognosis assessment, and therapeutic strategies for this aggressive malignancy.
... Since destroying the cancer cells would require the presence of therapeutic MPs at the tumor site, we studied the possibility of transporting these particles using adipose-derived mesenchymal cells (ADSCs) as carriers. These cells are known to preferentially target tumors, inflammations, or wound-healing sites, making them a convenient modality to carry therapeutic substances [17]. Magneto-mechanically actuating MP-loaded ADSCs, after insertion in the human osteosarcoma cell area, induce their death and the release of the MPs both from the surface and inside of the former cells, to be subsequently internalized by HOS. ...
Article
Full-text available
Magnetic nanoparticles (MPs) are emerging as powerful and versatile tools for biotechnology, including cancer research and theranostic applications. Stem cell-mediated magnetic particle delivery has been previously recognized as a modality to target sites of malignancies. Here, we propose the use of adipose-derived mesenchymal cells (ADSC) for the targeted delivery of Fe-Cr-Nb-B magnetic particles to human osteosarcoma (HOS) cells and magneto-mechanical actuation (MMA) for targeting and destroying HOS cells. We show that MPs are easily incorporated by ADSCs and HOS cells, as confirmed by TEM images and a ferrozine assay. MP-loaded ADSCs display increased motility towards tumor cells compared with their unloaded counterparts. MMA of MP-loaded ADSCs induces HOS destruction, as confirmed by the MTT and live/dead assays. MMA enables the release of the MPs towards cancer cells, producing a significant decrease (about 80%) in HOS viability immediately after application. In contrast, normal human dermal fibroblasts’ (NHDFs) viability exposed to similar conditions remains high, showing a differential behavior of normal and malignant cells to MP load and MMA exposure. Taken together, the method could derive successful strategies for in vivo applications in targeting and destroying malignant cells while protecting normal cells.
Article
Full-text available
Autophagy is a cytoplasmic defense mechanism that cells use to break and reprocess their intracellular components. This utilization of autophagy is regarded as a savior in nutrient-deficient and other stressful conditions. Hence, autophagy keeps contact with and responds to miscellaneous cellular tensions and diverse pathways of signal transductions, such as growth signaling and cellular death. Importantly, autophagy is regarded as an effective tumor suppressor because regular autophagic breakdown is essential for cellular maintenance and minimizing cellular damage. However, paradoxically, autophagy has also been observed to promote the events of malignancies. This review discussed the dual role of autophagy in cancer, emphasizing its influence on tumor survival and progression. Possessing such a dual contribution to the malignant establishment, the prevention of autophagy can potentially advocate for the advancement of malignant transformation. In contrast, for the context of the instituted tumor, the agents of preventing autophagy potently inhibit the advancement of the tumor. Key regulators, including calpain 1, mTORC1, and AMPK, modulate autophagy in response to nutritional conditions and stress. Oncogenic mutations like RAS and B-RAF underscore autophagy's pivotal role in cancer development. The review also delves into autophagy's context-dependent roles in tumorigenesis, metastasis, and the tumor microenvironment (TME). It also discusses the therapeutic effectiveness of autophagy for several cancers. The recent implication of autophagy in the control of both innate and antibody-mediated immune systems made it a center of attention to evaluating its role concerning tumor antigens and treatments of cancer.
Chapter
Schwann cells (SC), endothelial cells (EC), pericytes, and cancer-associated fibroblasts (CAF) are non-immune cells present in the tumor microenvironment (TME) of neuroblastoma (NB) tumors. These cells communicate with neuroblastoma (NB) cells through contact-dependent and contact-independent mechanisms which stimulate the production of soluble cytokines and chemokines that contribute to a pro-tumorigenic inflammatory reaction. Their roles in the TME of NB are substantially different and can be anti- as well as pro-tumorigenic. There is recent evidence that although these cells are not part of the immune system, they communicate with immune cells, modulating the immune response and often promoting immune escape. As our knowledge on the contribution of non-immune stromal cells in NB continues to expand, therapeutic approaches targeting these cells are considered. In this chapter, the role of SC, EC, and CAF, their mechanisms of communication with NB cells and immune cells, and recent clinical trials targeting these cells or their activity are reviewed in the context of recent literature.
Article
Full-text available
Background: Neural Precursor Cell Expressed Developmentally Down-Regulated Protein 1 (NEDD1) serves as a crucial factor in promoting cellular mitosis by directly facilitating wheel assembly and daughter centriole biogenesis at the lateral site of parent centrioles, ultimately driving centrosome replication. The amplification of centrosomes and the abnormal expression of centrosome-associated proteins contribute to the invasion and metastasis of non-small cell lung cancer cells. However, the specific mechanism by which NEDD1 contributes to the progression of lung adenocarcinoma (LUAD) remains unexplored. Therefore, the objective of this study is to uncover the role played by NEDD1 in LUAD. Methods: To verify the expression of NEDD1 in pan-carcinoma. The feasibility of NEDD1 as a prognostic marker for LUAD in TCGA and GEO databases was verified. Subsequently, Cox proportional hazard regression analysis was used to screen the prognostic factors of LUAD, so as to analyze the correlation between prognostic factors and NEDD1 expression. For another, NEDD1-related genes were screened for pathway enrichment analysis to verify their possible functions. In addition, the expression of NEDD1 in LUAD was verified by qPCR and IHC, then siRNA was used to construct NEDD1-knocked lung cancer cells for subsequent cytobehavioral experiments. Finally, the distribution of NEDD1 in single-cell samples was revealed, and then the correlation between its overexpression and LUAD immune escape and drug resistance was analyzed. Results: LUAD exhibits upregulation of NEDD1, which in turn promotes the proliferation, migration, invasion, and epithelial-mesenchymal transition of lung cancer cells, thereby contributing to a poor prognosis. Furthermore, the overexpression of NEDD1 is closely associated with immune escape and drug resistance in LUAD. Conclusion: NEDD1 serves as a reliable prognostic marker for LUAD, and its upregulation is associated with increased immune escape and drug resistance. Given these findings, NEDD1 holds potential as a novel therapeutic target for the treatment of LUAD.
Article
Full-text available
Lung cancer’s intractability is enhanced by its frequent resistance to (chemo)therapy and often high relapse rates that make it the leading cause of cancer death worldwide. Improvement of therapy efficacy is a crucial issue that might lead to a significant advance in the treatment of lung cancer. Oncolytic viruses are desirable combination partners in the developing field of cancer immunotherapy due to their direct cytotoxic effects and ability to elicit an immune response. Systemic oncolytic virus administration through intravenous injection should ideally lead to the highest efficacy in oncolytic activity. However, this is often hampered by the prevalence of host-specific, anti-viral immune responses. One way to achieve more efficient systemic oncolytic virus delivery is through better protection against neutralization by several components of the host immune system. Carrier cells, which can even have innate tumor tropism, have shown their appropriateness as effective vehicles for systemic oncolytic virus infection through circumventing restrictive features of the immune system and can warrant oncolytic virus delivery to tumors. In this overview, we summarize promising results from studies in which carrier cells have shown their usefulness for improved systemic oncolytic virus delivery and better oncolytic virus therapy against lung cancer.
Article
Full-text available
Chrozophora plicata has been extensively utilized in India for the management of numerous disorders. The effective Phytoconstituents derived from the Ethyl Acetate Fraction of Chrozophora plicata [EAFCP] have been identified as Camptothecin Agathisflavone, Rutin, Procynidine B, and Apigenin. These Phytoconstituents have been detected in the EAFCP through qualitative analysis using LC-Q-TOF-MS/MS. The anti-inflammatory properties of Chrozophora plicata are yet to be determined. Therefore, the aim of this study was to utilize a network pharmacology-based methodology to predict potential therapeutic targets of EAFCP in the setting of inflammation. The identification of inflammation targets was followed by the acquisition of verified targets of EAFCP. The key therapeutic targets of EAFCP against inflammation were found by creating a target-functional PPI network, GO studies were conducted on the core therapeutic targets in order to assess the essential signalling pathways involved in the anti-inflammatory effects of EAFCP. A total of 38 significant hub targets associated with EAFCP's anti-inflammatory effects were identified. The key proteins were retrieved for the docking investigation based on the findings, which aided in anticipating the potential interaction between components and targets. The in vivo study revealed that EAFCP had a notable efficiency in decreasing paw edema induced by carrageenan in rats. The evidence we have gathered collectively offers clarification about the anti-inflammatory activity of EAFCP, which is predominantly linked to the suppression of the Cox 1, 2 pathway. The aforementioned findings highlight potential therapeutic targets that could be utilized for the anti-inflammatory activity of EAFCP.
Article
Targeting actionable mutations in oncogene‐driven cancers and the evolution of immuno‐oncology are the two prominent revolutions that have influenced cancer treatment paradigms and caused the emergence of precision oncology. However, intertumoral and intratumoral heterogeneity are the main challenges in both fields of precision cancer treatment. In other words, finding a universal marker or pathway in patients suffering from a particular type of cancer is challenging. Therefore, targeting a single hallmark or pathway with a single targeted therapeutic will not be efficient for fighting against tumor heterogeneity. Mesenchymal stem cells (MSCs) possess favorable characteristics for cellular therapy, including their hypoimmune nature, inherent tumor‐tropism property, straightforward isolation, and multilineage differentiation potential. MSCs can be loaded with various chemotherapeutics and oncolytic viruses. The combination of these intrinsic features with the possibility of genetic manipulation makes them a versatile tumor delivery vehicle that can be used for in vivo selective tumor delivery of various chemotherapeutic and biological therapeutics. MSCs can be used as biofactory for the local production of chemical or biological anticancer agents at the tumor site. MSC‐mediated immunotherapy could facilitate the sustained release of immunotherapeutic agents specifically at the tumor site, and allow for the achievement of therapeutic concentrations without the need for repetitive systemic administration of high therapeutic doses. Despite the enthusiasm evoked by preclinical studies that used MSC in various cancer therapy approaches, the translation of MSCs into clinical applications has faced serious challenges. This manuscript, with a critical viewpoint, reviewed the preclinical and clinical studies that have evaluated MSCs as a selective tumor delivery tool in various cancer therapy approaches, including gene therapy, immunotherapy, and chemotherapy. Then, the novel nanotechnology and bioengineering approaches that can improve the potency of MSC for tumor targeting and overcoming challenges related to their low localization at the tumor sites are discussed.
Article
Full-text available
Background The use of umbilical cord mesenchymal stem cells (UC-MSCs) is a burgeoning method for the treatment of liver cirrhosis. However, the secretory phenotype and regulatory ability of UC-MSCs are easily affected by their microenvironment. Ensuring a specific microenvironment to enhance the UC-MSCs phenotype is a potential strategy for improving their therapeutic efficacy. The aim of this study was to explore therapeutic UC-MSCs phenotypes for improving liver fibrosis. Methods RNA-sequencing was used to analyze the response pattern of UC-MSCs after exposure to the serum of cirrhotic patients with HBV. Using immunohistochemistry, quantitative polymerase chain reaction, and immunofluorescence techniques, we evaluated the therapeutic effect of UC-MSCs pretreated with interferon alpha 2 (IFN-α2) (pre-MSCs) in an animal model of cirrhosis. Immunoblotting, ELISA, and other techniques were used to analyze the signaling pathways underlying the IFN-induced changes in UC-MSCs. Results UC-MSCs exposed to the serum of patients with hepatitis B-induced cirrhosis showed an enhanced response to type I IFN. The activated type I IFN signal induced the highest secretion of colony-stimulating factor 3 (CSF-3), interleukin (IL)-8, and chemokine (C–C motif) ligand 20 (CCL20) by the UC-MSCs. Pre-MSCs showed a higher therapeutic efficacy than untreated UC-MSCs in an animal model of liver fibrosis. Immunohistochemical analysis revealed that pre-MSCs could recruit neutrophils resulting in an increase in the secretion of matrix metalloprotease 8 that alleviated fibrosis. When neutrophils in animals were depleted, the therapeutic effect of pre-MSCs on fibrosis was inhibited. IFN-α2 altered the secretory phenotype of UC-MSCs by activating phosphorylated signal transducer and activator of transcription 1 and 2 (p-STAT1 and p-STAT2). Conclusions Pre-MSCs exhibited enhanced secretion of CSF-3, IL-8, and CCL20 and recruited neutrophils to alleviate fibrosis. This new strategy can improve cell therapy for liver cirrhosis. Graphical abstract
Article
Chemotherapy remains one of the most prevailing strategies for cancer treatment. However, its treatment effect is hampered by drug resistance, nonspecific tumor targeting, and severe toxic side effects. Combination chemotherapy with synergistic effect has become an attractive tumor therapy. N6-methyladenosine (m6A) regulators determine the fate of m6A-modified transcripts and play vital roles in cancer development and drug resistance. Gene therapy such as small interfering RNA (siRNA) is a promising strategy to reduce the abnormal gene expression of m6A regulators. However, its poor selectivity and high systemic toxicity necessitate the use of delivery vectors to target specific cells and tissues. Here, we constructed a dual-functional targeted nanodrug platform for the synergetic m6A-associated epigenetic regulation and chemotherapy of ovarian cancer. We encapsulated siRNA targeting the m6A reader YT521-B homology (YTH) N6-methyladenosine RNA-binding protein 1 (YTHDF1) and docetaxel (DTX), the first-line chemotherapeutic agent of ovarian cancer, into mesenchymal stem cell-derived small extracellular vesicles (MsEVs). This nanosystem exhibits significant tumor targeting and endo/lysosomal escape of siYTHDF1. It effectively depletes YTHDF1 and suppresses the protein translation of eukaryotic translation initiation factor 3 subunit C (EIF3C) in an m6A-dependent manner. The combination of YTHDF1-targeting epigenetic regulation significantly enhances the anti-tumor effect of DTX and effectively inhibits ovarian cancer progression without causing significant systemic toxicity. This co-delivery nanoplatform offers a promising approach for combinational cancer treatment, showing improved anti-tumor efficacy through the synergistic effects of epigenetic regulation and chemotherapeutic inhibition.
Article
Full-text available
Introduction and Aims: In our previous studies we found that exercise partially prevents chronic kidney disease (CKD)-induced muscle atrophy. Effective therapeutic strategies to treat Chronic Kidney Disease (CKD)-induced muscle atrophy are urgently needed. Low frequency electrical stimulation (LFES) induces muscle contraction which mimics acupuncture and exercise, and may be effective in preventing muscle atrophy. Methods: CKD was induced in 20-25g mice by 5/6th nephrectomy. CKD and control mice were treated with LFES for 15 days. Results: LFES prevented soleus and EDL muscle weight loss in CKD mice (p<0.05, LFES/CKD vs. CKD) as well as loss of hind-limb muscle grip. LFES countered the CKD-induced decline in the IGF-1 signaling pathway, leading to increased protein synthesis, decreased protein degradation and increased myogenesis markers. There is an acute (immediate) response phase during which inflammation cytokines (IFN and IL-6) increased. M1 macrophage markers (pro-inflammation: IL-1β) were also increased acutely, but M2 markers (anti-inflammation: arg-1, IL-10 and IL-4) were increased 2-days after initiation of LFES. In addition, microRNA-1 and -206 were decreased in the acute phase after starting LFES. Conclusions: We conclude that LFES ameliorates CKD-induced skeletal muscle atrophy by up-regulation of the IGF-1 signaling pathway which improves protein metabolism and promotes myogenesis. The potential mechanisms leading to upregulation of IGF are 1) decrease of microRNA-1 and -206 in the early phase of LEFS resulting in reversal of the inhibition of IGF-1 production by these microRNAs or 2) direct secretion of IGF-1 by M2 macrophages in the later phase inflammatory response.
Article
Full-text available
Leukocytes navigate through complex chemoattractant arrays, and in so doing, they must migrate from one chemoattractant source to another. By evaluating directional persistence and chemotaxis during neutrophil migration under agarose, we show that cells migrating away from a local chemoattractant, against a gradient, display true chemotaxis to distant agonists, often behaving as if the local gradient were without effect. We describe two interrelated properties of migrating cells that allow this to occur. First, migrating leukocytes can integrate competing chemoattractant signals, responding as if to the vector sum of the orienting signals present. Second, migrating cells display memory of their recent environment: cells' perception of the relative strength of orienting signals is influenced by their history, so that cells prioritize newly arising or newly encountered attractants. We propose that this cellular memory, by promoting sequential chemotaxis to one attractant after another, is in fact responsible for the integration of competitive orienting signals over time, and allows combinations of chemoattractants to guide leukocytes in a step-by-step fashion to their destinations within tissues.
Article
Full-text available
MSCs are nonhematopoietic stromal cells that are capable of differentiating into, and contribute to the regeneration of, mesenchymal tissues such as bone, cartilage, muscle, ligament, tendon, and adipose. MSCs are rare in bone marrow, representing ∼1 in 10,000 nucleated cells. Although not immortal, they have the ability to expand manyfold in culture while retaining their growth and multilineage potential. MSCs are identified by the expression of many molecules including CD105 (SH2) and CD73 (SH3/4) and are negative for the hematopoietic markers CD34, CD45, and CD14. The properties of MSCs make these cells potentially ideal candidates for tissue engineering. It has been shown that MSCs, when transplanted systemically, are able to migrate to sites of injury in animals, suggesting that MSCs possess migratory capacity. However, the mechanisms underlying the migration of these cells remain unclear. Chemokine receptors and their ligands and adhesion molecules play an important role in tissue-specific homing of leukocytes and have also been implicated in trafficking of hematopoietic precursors into and through tissue. Several studies have reported the functional expression of various chemokine receptors and adhesion molecules on human MSCs. Harnessing the migratory potential of MSCs by modulating their chemokine-chemokine receptor interactions may be a powerful way to increase their ability to correct inherited disorders of mesenchymal tissues or facilitate tissue repair in vivo. The current review describes what is known about MSCs and their capacity to home to tissues together with the associated molecular mechanisms involving chemokine receptors and adhesion molecules. Disclosure of potential conflicts of interest is found at the end of this article.
Article
Full-text available
Adult bone marrow (BM)-derived stem cells, including hematopoietic stem cells (HSCs) and MSCs, represent an important source of cells for the repair of a number of damaged tissues. In contrast to HSCs, the soluble factors able to induce MSC migration have not been extensively studied. In the present work, we compared the in vitro migration capacity of human BM-derived MSCs, preincubated or not with the inflammatory cytokines interleukin 1beta (IL1beta) and tumor necrosis factor alpha (TNFalpha), in response to 16 growth factors (GFs) and chemokines. We show that BM MSCs migrate in response to many chemotactic factors. The GFs platelet-derived growth factor-AB (PDGF-AB) and insulin-like growth factor 1 (IGF-1) are the most potent, whereas the chemokines RANTES, macrophage-derived chemokine (MDC), and stromal-derived factor-1 (SDF-1) have limited effect. Remarkably, preincubation with TNFalpha leads to increased MSC migration toward chemokines, whereas migration toward most GFs is unchanged. Consistent with these results, BM MSCs express the tyrosine kinase receptors PDGF-receptor (R) alpha, PDGF-Rbeta, and IGF-R, as well as the RANTES and MDC receptors CCR2, CCR3, and CCR4 and the SDF-1 receptor CXCR4. TNFalpha increases CCR2, CCR3, and CCR4 expression (as opposed to that of CXCR4), together with RANTES membrane binding. These data indicate that the migration capacity of BM MSCs is under the control of a large range of receptor tyrosine kinase GFs and CC and CXC chemokines. Most chemokines are more effective on TNFalpha-primed cells. Our results suggest that the mobilization of MSCs and their subsequent homing to injured tissues may depend on the systemic and local inflammatory state. Disclosure of potential conflicts of interest is found at the end of this article.
Article
Full-text available
Successful tumors - that is, tumors that grow progressively in the host - are obligate parasites. They have developed the capacity to preempt and subvert the wound-healing response of the host as a means to acquire the stroma they need to grow and expand. They mimic wounds by depositing an extravascular fibrin-fibronectin gel. Such gels, in tumors as at sites of local injury, signal the host to marshal the wound-healing response. This response is stereotyped and similar in both tumors and wounds. In tumors, however, the fibrin-fibronectin matrix signal that evokes the wound-healing response is not self-limited; it continues to operate and new gel is continuously deposited. Thus, tumors appear to the host in the guise of wounds or, more correctly, of an unending series of wounds that continually initiate healing but never heal completely.
Article
 The present article shows that a short-term exposure of purified human neutrophils to recombinant insulin conferred on these cells both the ability to migrate and the capacity to mobilize [Ca2+]i in response to CCL3, a chemokine per se ineffective with native neutrophils. Furthermore, the effects of recombinant insulin were reproduced by short-term incubation with sera from adult patients with metabolic syndrome, known to be characterized by a hyperinsulinemic state. A strict linear correlation (P < 0.01) between sera insulin levels and sera's ability to induce neutrophil locomotion was indeed found. Our data also suggest that (i) insulin primed neutrophils for migration to CCL3 via the selective activation of JNK 1/2, as shown by the use of inhibitors and kinase activation assay; (ii) the activation of Src kinases was necessary but not sufficient for CCL3-induced locomotory activity; (iii) PI3K-Akt, ERK 1/2, and p38 MAPK were not involved in insulin-induced migratory competence. In summary, we provided evidence that the exposition of neutrophils to insulin, as it occurs in hyperinsulinemic conditions, confers the competence of the cells to migrate in response to CCL3, known to be generated near atherosclerotic plaques. As neutrophils have been recently suggested to be involved in breaking unstable atherosclerotic plaques, the present findings contribute to the understanding of the pathophysiology of plaque instability. Finally, biochemical analysis herein carried out raises the hypothesis of JNK 1/2 as an attractive therapeutic target.
Article
Re-establishing blood flow to ischaemic tissues causes greater injury than that induced during the ischaemic period. This type of tissue injury, reperfusion injury, is involved in frostbite, multiple organ failure after hypovolaemia and in myocardial infarction. Depletion of neutrophils alleviates reperfusion injury, implying a causal role of neutrophil infiltration. Among members of the recently discovered family of chemotactic cytokines (chemokines), interleukin-8 (IL-8) is a major neutrophil chemotactic and activating factor produced by various types of human cells. We investigated its pathophysiological role in a rabbit model of a lung reperfusion injury. Reperfusion of ischaemic lung caused neutrophil infiltration and destruction of pulmonary structure, as well as local production of IL-8. Furthermore, the administration of a neutralizing monoclonal antibody against IL-8 prevented neutrophil infiltration and tissue injury, proving a causal role of locally produced IL-8 in this model.