ArticlePDF Available

Gao, J, Dennis, JE, Muzic, RF, Lundberg, M and Caplan, AI. The dynamic in vivo distribution of bone marrow-derived mesenchymal stem cells after infusion. Cells Tissues Organs (Print) 169: 12-20

Authors:

Abstract and Figures

Bone marrow-derived mesenchymal stem cells (MSCs) have the potential to differentiate along different mesenchymal lineages including those forming bone, cartilage, tendon, fat, muscle and marrow stroma that supports hematopoiesis. This differentiation potential makes MSCs candidates for cell-based therapeutic strategies for mesenchymal tissue injuries and for hematopoietic disorders by both local and systemic application. In the present study, rat marrow-derived MSCs were ex vivo culture-expanded, labeled with (111)In-oxine, and infused into syngeneic rats via intra-artery (i.a.), intravenous (i.v.) and intraperitoneal cavity (i.p.) infusions. In addition, for i.a. and i.v. infusions, a vasodilator, sodium nitroprusside, was administered prior to the cell infusion and examined for its effect on MSC circulation. The dynamic distribution of infused MSCs was monitored by real-time imaging using a gamma camera immediately after infusion and at 48 h postinfusion. After 48 h, radioactivity in excised organs, including liver, lungs, kidneys, spleen and long bones, was measured in a gamma well counter and expressed as a percentage of injected doses. After both i.a. and i.v. infusion, radioactivity associated with MSCs was detected primarily in the lungs and then secondarily in the liver and other organs. When sodium nitroprusside was used, more labeled MSCs cleared the lungs resulting in a larger proportion detected in the liver. Most importantly, the homing of labeled MSCs to the marrow of long bones was significantly increased by the pretreatment with vasodilator. These results indicate multiple homing sites for injected MSCs and that the distribution of MSCs can be influenced by administration of vasodilator.
Content may be subject to copyright.
Original Paper
Cells Tissues Organs 2001;169:12–20
The Dynamic in vivo Distribution of
Bone Marrow-Derived Mesenchymal
Stem Cells after Infusion
Jizong Gao
a
James E. Dennis
a
Raymond F. Muzic
b
Magnus Lundberg
a
Arnold I. Caplan
a
a
Skeletal Research Center, Department of Biology and
b
Department of Radiology, Case Western Reserve University,
Cleveland, Ohio, USA
Accepted after revision: July 25, 2000
Dr. Arnold I. Caplan
Skeletal Research Center, Department of Biology
Case Western Reserve University
2080 Adelbert Road, Cleveland, OH 44106-7080 (USA)
Tel. +1 216 368 3562, Fax +1 216 368 4077
ABC
Fax + 41 61 306 12 34
E-Mail karger@karger.ch
www.karger.com
© 2001 S. Karger AG, Basel
Accessible online at:
www.karger.com/journals/cto
Key Words
Mesenchymal stem cells
W Indium-111-oxine labeling W
Infusion W Imaging W Rat
Abstract
Bone marrow-derived mesenchymal stem cells (MSCs)
have the potential to differentiate along different mes-
enchymal lineages including those forming bone, carti-
lage, tendon, fat, muscle and marrow stroma that sup-
ports hematopoiesis. This differentiation potential
makes MSCs candidates for cell-based therapeutic strat-
egies for mesenchymal tissue injuries and for hemato-
poietic disorders by both local and systemic application.
In the present study, rat marrow-derived MSCs were ex
vivo culture-expanded, labeled with
111
In-oxine, and in-
fused into syngeneic rats via intra-artery (i.a.), intrave-
nous (i.v.) and intraperitoneal cavity (i.p.) infusions. In
addition, for i.a. and i.v. infusions, a vasodilator, sodium
nitroprusside, was administered prior to the cell infusion
and examined for its effect on MSC circulation. The
dynamic distribution of infused MSCs was monitored by
real-time imaging using a gamma camera immediately
after infusion and at 48 h postinfusion. After 48 h,
radioactivity in excised organs, including liver, lungs,
kidneys, spleen and long bones, was measured in a gam-
ma well counter and expressed as a percentage of
injected doses. After both i.a. and i.v. infusion, radioac-
tivity associated with MSCs was detected primarily in the
lungs and then secondarily in the liver and other organs.
When sodium nitroprusside was used, more labeled
MSCs cleared the lungs resulting in a larger proportion
detected in the liver. Most importantly, the homing of
labeled MSCs to the marrow of long bones was signifi-
cantly increased by the pretreatment with vasodilator.
These results indicate multiple homing sites for injected
MSCs and that the distribution of MSCs can be in-
fluenced by administration of vasodilator.
Copyright © 2001 S. Karger AG, Basel
Abbreviations used in this paper
CFU-F fibroblastic colony-forming cell
DMEM-LG Dulbecco’s modified Eagle’s medium-low glucose
FBS fetal bovine serum
i.a. intra-artery infusion
i.p. intraperitoneal cavity infusion
i.v. intravenous infusion
MSCs mesenchymal stem cells
VCAM-1 vascular cell adhesion molecule-1
Imaging of Infused MSCs
Cells Tissues Organs 2001;169:12–20
13
Introduction
Bone marrow stroma is a complex mesenchymal con-
nective tissue that contains a subset of cells termed mes-
enchymal stem cells (MSCs) [Caplan, 1991]. MSCs have
the potential to differentiate into different phenotypic
lineages forming bone, cartilage, muscle, tendon, liga-
ment, fat, muscle and marrow stroma [Bruder et al.,
1998a, b; Johnstone et al., 1998; Young et al., 1998; Pit-
tenger et al., 1999]. Culture-expanded MSCs have been
tested in preclinical models for the repair of bone [Bruder
et al., 1998a], cartilage [Johnstone and Yoo, 1999] and
tendon/ligament [Young et al., 1998] by local delivery of
MSCs within an appropriate matrix. Because MSCs are
able to differentiate into osteoblasts, a potential thera-
peutic application of MSCs would be to modulate or cure
age-related osteoporosis where decreased bone mass is
postulated to result from diminished osteogenesis [Parfitt
et al., 1995; Jilka et al., 1996]. In the case of osteoporosis,
local introduction of MSCs may not be efficient since
osteoporosis is a systemic disease. With this in mind, sys-
temic administration of MSCs would be required as a
therapeutic procedure.
Another function of marrow-derived MSCs is to sup-
port hematopoiesis by providing a supportive connective
tissue microenvironment, including the synthesis of cer-
tain regulators and cytokines for hematopoietic stem cell
proliferation and differentiation [Dexter et al., 1977; Gor-
don et al., 1990; Haynesworth et al., 1996; Weimar et al.,
1998]. Disruption of the marrow architecture with he-
morrhaging, loss of fat and loss of stromal components is
commonly found in patients after cytotoxic chemothera-
py [Ishida et al., 1994]. Therefore, there is a need to recon-
stitute the bone marrow stroma to augment the recovery
of hematopoietic function [Lazarus et al., 1995]. Koc et al.
[2000] intravenously (i.v.) coinfused human culture-ex-
panded autologous MSCs and hematopoietic stem cells
into the advanced breast cancer patients after chemother-
apy and found rapid hematopoietic recovery.
Since bone marrow-derived MSCs can be isolated
from marrow aspirates, expanded in vitro with many pas-
sages without loss of osteogenic potential [Caplan, 1991;
Haynesworth et al., 1992; Bruder et al., 1997], MSCs have
the potential of being gene therapy vehicles following
transduction with viral vectors that contain specific genes
[Allay et al., 1997]. Transduced MSCs have been deliv-
ered systemically in vivo and their secretory products
were found in different organs months later [Piersma et
al., 1983; Li et al., 1995; Allay et al., 1997; Chuah et al.,
1998; Pereira et al., 1998; Horwitz et al., 1999; Hou et al.,
1999; Oyama et al., 1999]. In a recent study [Xu et al.,
1999], MSCs transduced with a retroviral vector encoding
green fluorescent protein and neomycin resistance (neo)
genes were infused i.v. into syngeneic rats. Engrafted
MSCs were detected in the marrow of the recipient rat by
detecting the neo gene by PCR. However, no significant
differences in bone density or in the fibroblastic colony-
forming cell number were detected between the MSCs
and phosphate buffer saline infusion groups. These results
may indicate that insufficient numbers of infused MSCs
homed to the marrow or acquired a normal physiological
function.
Systemic MSC applications, including treatment of
musculoskeletal diseases, hematopoiesis support and gene
delivery, require an understanding of the distribution
dynamics of infused MSCs. The present study investi-
gated the dynamics of the infused MSCs via different por-
tals and traced MSCs homing in different organs using
isotope labeling, whole body scanning and real-time mon-
itoring. The aim of this study was to determine the fate of
the infused MSCs and to maximize systemic distribution
through the use of alternative delivery portals and the use
of vasoactive agents.
Materials and Methods
Rat bone marrow-derived MSCs were culture-expanded, labeled
with
111
In-oxine, and infused via various transferal portals to the syn-
geneic rats. In some cases, vasodilator was administered prior to the
cell infusion. The dynamic distribution of the infused MSCs was
monitored by the real-time imaging. The concentration of radioactiv-
ity in different excised organs was detected by a well counter. These
separate assays are outlined in figure 1.
Cell Culture
Cell isolation and culture procedures for MSCs in the authors’
laboratory have been established and published previously [Dennis
et al., 1992; Lennon et al., 1995]. Briefly, tibiae and femurs were
aseptically harvested from 3- to 4-month-old Fisher F-344 rats and
the adherent soft tissue was removed. The proximal end of the femur
and the distal end of the tibia were excised at a level just into the
beginning of marrow cavity. After the epiphyses were removed,
whole marrow plugs were obtained by flushing the bone marrow cav-
ity with a syringe (18-gauge needle) filled with Dulbecco’s modified
Eagle’s medium-low glucose (DMEM-LG; Sigma) supplemented
with antibiotic-antimycotic solution (penicillin G sodium: 100 U/ml,
streptomycin sulfate: 100 Ìg/ml, amphotericin B: 0.25 Ìg/ml; Gibco/
BRL) and 10% fetal bovine serum (FBS) of selected lots (Germini)
[Lennon et al., 1995]. The marrow plugs were dispersed to obtain a
single cell suspension by sequentially passing the dispersions through
18- and 22-gauge needles. The cells were centrifuged and resus-
pended with DMEM-LG containing 10% FBS. After counting in a
hemocytometer following an acetic acid disruption of red blood cells,
nucleated cells were plated at a density of 5.0!10
7
/100-mm culture
14
Cells Tissues Organs 2001;169:12–20
Gao/Dennis/Muzic/Lundberg/Caplan
Fig. 1.
Diagram showing the experimental
protocol including isolation and culture ex-
pansion of MSCs,
111
In-oxine labeling, cell
infusion and whole body imaging.
dish and incubated at 37
°
C in a humidified atmosphere of 95% air
and 5% CO
2
. The first medium change was after 4 days and twice a
week thereafter. When these primary MSCs reached 80–90% of con-
fluence, they were trypsinized, counted and passaged at a density of
0.5–0.7!10
6
/100-mm culture dish and are referred to as the 1st pas-
sage MSCs. As these first passage MSCs reached 80% of confluence,
they were labeled with
111
In-oxine by Mallincrodt, Cleveland, Ohio,
as described below. First passage MSCs were used for all of the exper-
iments in this study.
Indium Labeling of the Cells
MSCs in complete culture medium were stored on ice for transfer
to Mallincrodt approximately 5 h prior to infusion. Cells were
digested by 0.25% trypsin-EDTA (Gibco) for 10 min and centrifuged
at 320 g for 5 min. The cell pellet was resuspended in serum-free
medium and the cell number was determined. The cell suspension
was mixed with
111
In-oxine at 50–60 ÌCi/10
6
cells and incubated for
10 min. The
111
In-oxine radiolabeling efficiency of MSCs is about
70–80%, resulting in a specific activity of approximately 40 ÌCi/10
6
cells. The radiolabeled MSCs were aliquoted at 1–1.3!10
6
cells/ml.
Preliminary experiments showed that the viability and growth of
these labeled MSCs were not adversely affected by this labeling pro-
cedure (data not shown); the level of radioisotope was sufficient to
produce high quality images taken with a gamma camera. These
labeled MSCs were then transported to the Nuclear Medicine De-
partment of University Hospital at Case Western Reserve Universi-
ty, and infusion of these cells was done within 1 h followed by whole
body scanning and real-time imaging.
Infusion of
111
In-Labeled MSCs via Different Portals and
Dynamic Imaging
The infusion portals included femoral vein (i.v.), femoral artery
(i.a.) and intraperitoneal cavity (i.p.). Blood vessel cannulation of
these syngeneic Fisher F-344 rats was done by the commercial sup-
plier (Charles River, Wilmington, Mass.) prior to transport to our
site. The animals were anesthetized by intraperitoneal injection of
ketamine (800 mg/kg) and xylazine (10 mg/kg). A 1-ml cell suspen-
sion was infused via a cannulated tube that was then flushed with 0.2
to 0.3 ml of normal saline in order to infuse cells remaining in the
tubing. Radioactivity in the syringe was measured before and after
injection to determine the infused counts. Results from pilot studies
demonstrated that most radioactivity was localized in the lungs via
i.a. or i.v. injections even after 48 h of infusion. We postulated that
the reason for this distribution may be due to the size of the MSCs
(average of about 20–30 Ìm, unpubl. data) relative to the pulmonary
capillaries that are 14 Ìm in diameter on average [Richardson, 1976],
which may have impeded the infused MSCs from passing through
the lung circulation. Therefore, in some cases, the vasodilator, sodi-
um nitroprusside, was administered at the dosage of 0.001 g/kg body
weight prior to the infusion of cell suspension. In order to test the
effectiveness and duration of sodium nitroprusside administration,
arterial blood pressure measurements were performed before and
after sodium nitroprusside injections. Within 1 min postinjection,
blood pressure dropped by 50% and recovered to 90% of preinjection
values in 6–7 min (data not shown). For intraperitoneal cavity infu-
sion, 1 ml of labeled cell suspension was directly injected into the
cavity with a 23-gauge needle. Planar whole body images were
acquired with Siemens E.Cam scanner (Hoffman Estates, Ill.) using a
medium energy collimator. Scanning was performed for 15 min at
one frame per minute (fig. 2). Whole body scanning was repeated
48 h after infusion.
111
In Distribution by Organ
At 48 h postinjection, immediately following the imaging, ani-
mals were euthanized by overdose anesthesia, and organs including
liver, lungs, spleen, kidneys and long bones were harvested. These
organs were weighed and assayed for radioactivity using a 1282
Compugamma (LKB Wallac, Gaithersburg, Md.). Unlike the other
organs, the liver was too large to assay in toto. Consequently, a piece
of it was sampled and its radioactivity concentration was deter-
mined. The concentration was then scaled based on the mass of the
i.a.
Imaging of Infused MSCs
Cells Tissues Organs 2001;169:12–20
15
Fig. 2.
Planar whole body images were ac-
quired with Siemens E.Cam scanner using a
medium energy collimator. Scanning was
performed for 15 min at one frame per min-
ute and the composite image was finally
reconstructed (white-marked area at bottom
right).
Table 1.
Distribution of experimental animals into different groups
Infusion portals No vasodilator Use of vasodilator Total
5510
i.v. 6 6 12
i.p. 0 6 6
Total 11 18 28
entire liver in order to estimate the liver radioactivity. Bone tissue
was harvested from tibiae and femurs of both hind limbs. Tibias were
dislocated from the ankle joint and isolated from the proximal tibial
growth plate. The femur was harvested by dislocation from the hip
joint and from the distal femoral growth plate. All soft tissue and
periosteum were cleaned from these bones which were then pooled
together for gamma counting. Because it was difficult to quantitative-
ly harvest all bone marrow tissue from the long bone, no attempt was
made to distinguish between the radioactivity in the marrow itself
and that in the native bone.
Data Analysis. Totally, 28 rats were used for this study. These
animals were distributed into different group as summarized in
table 1. Radioactivity in each organ was expressed in two ways: by
counts per unit mass and as a percentage of injected dose. In all cases,
radioactive decay of
111
In was taken into account by decay corrected
to the time of injection. Differences of the radioactivity among the
measured organs were determined using analysis of variances
(ANOVA) at a threshold of p = 0.05 to indicate statistical signifi-
cance.
Results
Viability and Proliferation of
111
In-Oxine-Labeled
MSCs
A small aliquot of MSCs was sampled from the cell
preparation labeled by
111
In-oxine. The cell viability was
determined by exclusion of trypan blue. The average via-
bility of
111
In-oxine-labeled cells was approximately 90%
without significant differences between different cell
preparations. Labeled MSCs were plated at a density of
about 0.6!10
6
/100-mm culture dish. Cell attachment
was observed at about 3–4 h. After 2 days, cells had a typi-
cal fibroblast-like morphology and were evenly distribut-
ed on the plate (fig. 3a, b). Typically, about 80–90% of
16
Cells Tissues Organs 2001;169:12–20
Gao/Dennis/Muzic/Lundberg/Caplan
Fig. 3.
111
In-oxine-labeled MSCs prolifer-
ate in the culture plate at 2 days. These cells
had a typical fibroblast-like morphology
and were evenly distributed on the plate.
Original magnification: !4 (
a
) and !10
(
b
).
Fig. 4.
Composite image of whole body scanning immediately (
a
) and at 48 h after i.v. cell infusion (
b
), with (left) and
without (right) the use of vasodilator. Lu = Lungs; Li = liver; S = spleen; K = kidneys.
confluence was reached by day 6–7. These growth pat-
terns were similar to those of normal rat bone marrow-
derived MSCs [Lennon et al., 1995], which may indicate
that
111
In-oxine labeling did not affect the cell viability
and proliferation.
Whole Body Scanning and Dynamic Imaging
The distribution of radioactivity after infusion of the
labeled MSCs was imaged at 1-min intervals for 15 min.
Although not quantitative, this imaging provides an im-
mediate indication of where a majority of the signal
resides. A single composite image representing the sum of
all 15 images was reconstructed (fig. 2). After both i.a. and
i.v. infusion, the radioactivity was first observed to accu-
mulate in the lungs, and gradually, the radioactivity was
observed in the liver. When vasodilator was administered
prior to the cell infusion, although considerable radioac-
tivity is still observed in the lungs (fig. 4a), it was observed
Imaging of Infused MSCs
Cells Tissues Organs 2001;169:12–20
17
Fig. 5.
Composite image of whole body scanning after i.p. infusion:
radioactivity was identified in the liver, lungs, spleen and testicles.
Fig. 6.
Radioactivity concentration in different organs was scaled
based on the organ mass and the total infused dose after normaliza-
tion to the half-life of the
111
In. Most of the radioactivity accumu-
lated in the lungs and liver. When vasodilator was used, the radioac-
tivity in the lungs was decreased by about 15%; in contrast, radioac-
tivity in the liver was increased by a similar percentage (
a
), and
radioactivity in the long bone increased by about 50% (
b
).
directly from the monitor of the scanner that radioactivity
accumulated in the liver faster than in infusions without
using vasodilator. At 48 h after cell infusion, whole body
scanning was repeated. The radioactivity was observed
primarily in the liver for i.a., i.v. and i.p. infusion with
considerable amounts detected in the lungs and kidneys
after both i.a. and i.v. infusion. Vasodilator-treated rats
showed greater intensity of radioactivity in the liver than
the lungs compared to rats receiving no vasodilator. Ra-
dioactivity was also identified in the bone of the rat that
received vasodilator (fig. 4b, arrow). After i.p. infusion,
radioactivity was observed in kidneys, spleen, liver and
lungs in small amounts (fig. 5).
Organ Distribution of the Radioactivity
In order to quantify the distribution of
111
In, the spe-
cific radioactivity of each organ was calculated as a per-
centage of the total infused radioactivity related to the
particular organ mass. After both i.v. and i.a. infusion, the
radioactivity in the lungs and liver comprised about 50%
of the infused radioactivity. The use of vasodilator in-
creased the counts in the liver by about 10% and de-
creased that in the lungs by 15% (fig. 6a). Although the
percentage of radioactivity in bone was low when com-
pared to larger organs such as lungs and liver, the radioac-
tivity in the bone was increased by almost 50% with the
use of vasodilator (p = 0.008) (fig. 6b). No significant dif-
ference in gamma counts in the different organs was
observed when comparing i.a. and i.v. infusion.
Discussion
In the present study, infused MSCs lodged in the lungs
and only a small percentage of the radioactivity gradually
accumulated in the liver and spleen after infusion via
either intravenous or intra-artery. Kuppen et al. [1992]
found that
111
In-oxine-labeled lymphokine-activated kill-
5
6
18
Cells Tissues Organs 2001;169:12–20
Gao/Dennis/Muzic/Lundberg/Caplan
er cells infused via jugular vein were all detected in the
lungs up to 2 h after injection, but, after 8 h, radioactivity
was only observed in the liver and the spleen. In contrast,
even as long as 48 h after injection of MSCs in the present
study, considerable amount of radioactivity was still de-
tected in the lungs. The primary factors attributed to the
lodging of MSCs within lungs are probably cellular diame-
ter and cellular attachment potential. The average size of
rat MSCs at the 1st passage of culture is between 20 and
24 Ìm in diameter (data not shown), while the capillaries
of the lungs are about 10–15 Ìm in diameter. For compar-
ison, detached human MSCs are 2–3 times larger than
neutrophils in cytospin preparation [Koc et al., 2000].
Bone marrow-derived MSCs are attachment-competent
cells. MSCs attach in a fibronectin-facilitated manner in
in vitro culture conditions [Dennis et al., 1992] and may
specifically attach to fibronectin-rich endothelia domains
in vivo. Therefore, when the infused
111
In-oxine-labeled
MSCs circulated to the lungs, the larger MSCs may not be
able to pass through the lung capillaries or the MSCs
attach to the endothelial cells in a receptor-mediated pro-
cess. In this regard, respiratory failure after bone marrow
transplantation is one of the most common complications
in the clinical practice and may be due to abundant cells
lodging in the lung [Crawford and Petersen, 1992; Bojko
and Notterman, 1999; Leneveu et al., 1999]. When sodi-
um nitroprusside was used, as reported here, the radioac-
tivity in the lungs was decreased by 15% and that in the
liver, kidneys and bone increased by 10–50%. Sodium
nitroprusside is a strong vasodilator and also is an inhibi-
tor of aggregation and adhesion of platelets [Tinker and
Michenfelder, 1976; Krossnes et al., 1996, 1998]. There-
fore, the administration of sodium nitroprusside may
have influenced the distribution of infused MSCs by
enlarging the diameter of the pulmonary capillaries and
preventing cell adhesion and aggregation. Homing of ex
vivo expanded marrow-adherent cells has been described
after intravenous infusion in mice [Pereira et al., 1995,
1998]. About 5% of the infused cells were found in the
lungs as identified by PCR assays for a marker collagen I
gene at 5 months after infusion. Moreover, the pulmonary
vascular bed has been tested as a site for implantation of
isolated liver cells [Selden, 1984], in which rat hepato-
cytes were expanded ex vivo and infused via jugular vein.
Infused hepatocytes were identified at 24 h to 56 days
after infusion within the vascular space or capillaries of
the lungs. All these data suggest that the lung may be an
ectopic binding site for different types of cells or that cul-
tured cells lodge there due to their size or lack of deforma-
tion properties compared to the blood cells. Specific
receptor was not identified on the epithelial cells of the
lungs and the effect of these lodged cells on the function of
lungs has not yet been critically evaluated.
In the present study,
111
In-oxine was chosen as the
labeling agent, which has been widely used for labeling of
many cell types.
111
In-oxine is a lipophilic agent with
67.5 h half-life and emits gamma rays suitable for imag-
ing. These physical characteristics of
111
In have permitted
computer-assisted imaging with a scintillation camera for
the study of kinetics as well as the organ distribution of
labeled cells [Thakur, 1983; Abreo et al., 1985]. Further,
the viability of labeled cells was higher using
111
In-oxine
(90%) compared with
18
-F-FDG (80%) and
99m
Tc (85%)
[Botti et al., 1997]. In a pilot study, we observed that
the viability of
111
In-oxine-labeled MSCs at the dose of
40 ÌCi/10
6
MSCs was about 90% using the trypan blue
exclusion method. These labeled cells grew well after they
were plated and cultured under the standard conditions.
Similar results of cell proliferation and biological func-
tions were reported for
111
In-oxine-labeled lymphocytes
[Zakhireh et al., 1979; Kuyama et al., 1997]. These data
confirmed that the
111
In-oxine is a safe, non-cell-toxic and
effective labeling agent.
Bone marrow transplantation is a common procedure
for the recovery of hematopoietic function following se-
vere damage by conventional high-dosage cytotoxic che-
motherapy or by radiation exposure and for the cure of
hematopoietic disease such as leukemia. In the present
study, only a small percentage of infused MSCs lodged in
the marrow, even when vasodilator was administered.
Pretransplantation chemo- or radioablation of the bone
marrow may increase the homing of infused MSCs. Piers-
ma et al. [1983] infused chromosome-marked bone mar-
row cells to a mouse that had been given lethal total body
irradiation. At 1 day after the transplantation, 72% of
donor fibroblastic colony-forming cells (CFU-F) had
reached the recipient bone marrow, indicating a highly
specific lodging of CFU-F within marrow in these ani-
mals. Even 3 months after transplantation, donor CFU-F
was still detectable and comprised about half of the femo-
ral CFU-F population. Vascular cell adhesion molecule-1
(VCAM-1) is expressed by the marrow stromal cells [Ja-
cobsen et al., 1996] and may play an important role in the
interchange of cells between bone marrow and blood.
After gamma-irradiation to deplete the marrow, the label-
ing intensity of VCAM-1 on reticular and endothelial cells
was increased [Jacobsen et al., 1996]. Irradiation of mar-
row may also modify the marrow-blood barrier to facili-
tate the lodging of the circulating infused cells [Tavassoli,
1979; Tavassoli and Hardy, 1990]. Marrow depletion
Imaging of Infused MSCs
Cells Tissues Organs 2001;169:12–20
19
may, therefore, be another way to improve the homing of
infused MSCs.
The distribution of MSCs to different organs after
infusion by different portals may indicate that these
MSCs may circulate in the blood or lymphatic flow and
eventually home in different organs. These results are
supported by other studies where the systemic infusion or
local injection of ex vivo expanded MSCs have been
reported [Pereira et al., 1995; Oyama et al., 1999]. Since
culture-expanded MSCs can be transfected using retrovi-
ruses and express high-level marker gene in vitro and in
vivo [Allay et al., 1997; Oyama et al., 1999], the technique
of MSCs in vitro expansion and infusion may hold prom-
ise as part of gene therapy. The multiorgan homing of the
infused MSCs may facilitate their use in gene therapy to
different organs if we will be able to guide these gene-
modified MSCs to the location of particular interest. Last-
ly, if the lodging efficiency of MSCs to bone marrow could
be enhanced, systemic administration of osteoprogenitor
cells, MSCs, may provide all bony sites with osteogenic
cells capable of combating age-related osteoporosis.
Acknowledgement
The authors thank Dr. Donald Lennon for MSC preparation,
Debbie Fein-Krantz for animal care, Pat Devlin and Nicole Marcan-
tonio for assistance with the whole body scanning, and Dr. N.R.
Prabhakar and Dr. D.D. Kline for assistance with the blood pressure
measurement. This study was supported by grants from National
Institutes of Health. M.L. was supported by grants from The Swedish
Medical Society and The Fulbright Commission.
References
Abreo, K., A.K. Lieberman, A.V. Moorthy (1985)
Distribution studies of
111
In-oxine-labeled
peritoneal mononuclear cells in tumor-bearing
rats. Int J Nucl Med Biol 12: 53−55.
Allay, J.A., J.E. Dennis, S.E. Haynesworth, M.K.
Majumdar, D.W. Clapp, L.D. Shultz, A.I.
Caplan, S.L. Gerson (1997) LacZ and interleu-
kin-3 expression in vivo after retroviral trans-
plantation of marrow-derived human osteo-
genic mesenchymal progenitors. Hum Gene
Ther 8: 1417–1427.
Bojko, T., D.A. Notterman (1999) Reversal of for-
tune? Respiratory failure after bone marrow
transplantation. Crit Care Med 27: 1061–
1062.
Botti, C., D.R.M. Negri, E. Seregni, et al (1997)
Comparison of three different methods for ra-
diolabeling human activated T lymphocytes.
Eur J Nucl Med 24: 497–504.
Bruder, S.P., N. Jaiswal, S.E. Haynesworth (1997)
Growth kinetics, self-renewal, and the osteo-
genic potential of purified human mesenchy-
mal stem cells during extensive subcultivation
and following cryopreservation. J Cell Biochem
64: 278–294.
Bruder, S.P., N. Jaiswal, N.S. Ricalton, J.D. Mosca,
K.H. Kraus, S. Kadiyala (1998a) Mesenchymal
stem cells in osteobiology and applied bone
regeneration. Clin Orthop 355S: S247–256.
Bruder, S.P., K.H. Kraus, V.M. Goldberg, S. Ka-
diyala (1998b) The effect of implants loaded
with autologous mesenchymal stem cells on the
healing of canine segmental bone defects. J
Bone Joint Surg Am 80: 985–996.
Caplan, A.I. (1991) Mesenchymal stem cells. J Or-
thop Res 9: 641–650.
Chuah, M.K.L., H. Brems, V. Vanslembrouck, D.
Collen, T. Vandendriessche (1998) Bone mar-
row stromal cells as targets for gene therapy of
hemophilia A. Hum Gene Ther 9: 353–365.
Crawford, S.W., F.B. Petersen (1992) Long-term
survival from respiratory failure after marrow
transplantation for malignancy. Am Rev Re-
spir Dis 145: 510–514.
Dennis, J.E., S.E. Haynesworth, R.G. Young, A.I.
Caplan (1992) Osteogenesis in marrow-derived
mesenchymal cell porous ceramic composites
transplanted subcutaneously: Effect of fibro-
nectin and laminin on cell retention and rate of
osteogenic expression. Cell Transplant 1: 23–
32.
Dexter, T.M., T.D. Allen, L.G. Lajtha (1977) Con-
ditions controlling the proliferation of hemo-
poietic stem cells in vitro. J Cell Physiol 91:
335–344.
Gordon, M.Y., D. Clarke, J. Atkinson, M.F.
Greaves (1990) Hemopoietic progenitor cell
binding to the stromal microenvironment in
vitro. Exp Hematol 18: 837–842.
Haynesworth, S.E., M.A. Baber, A.I. Caplan (1992)
Characterization of cells with osteogenic poten-
tial from human marrow. Bone 13: 81–88.
Haynesworth, S.E., M.A. Baber, A.I. Caplan (1996)
Cytokine expression by human marrow-de-
rived mesenchymal progenitor cells in vitro:
Effects of dexamethasone and IL-1 alpha. J
Cell Physiol 166: 585–592.
Horwitz, E.M., D.J. Prockop, L.A. Fitzpatrick,
W.W.K. Koo, P.L. Gordon. M. Neel, M. Suss-
man, P. Orchard, J.C. Marx, R.E. Pyeritz,
M.K. Brenner (1999) Transplantability and
therapeutic effects of bone marrow-derived
mesenchymal cells in children with osteogene-
sis imperfecta. Nat Med 5: 309–313.
Hou, Z., Q. Nguyen, B. Frenkel, S.K. Nilsson, M.
Milne, A.J. van Wijnen, J.L. Stein, P. Quesen-
berry, J.B. Lian, G.S. Stein (1999) Osteoblast-
specific gene expression after transplantation
of marrow cells: Implications for skeletal thera-
py. Proc Natl Acad Sci USA 96: 7294–7299.
Ishida, T., M. Inaba, H. Hisha, et al (1994) Re-
quirement of donor-derived stromal cells in the
bone marrow for successful allogeneic bone
marrow transplantation. J Immunol 152:
3319–3127.
Jacobsen, K., J. Kravitz, P.W. Kincade, D.G. Os-
mond (1996) Adhesion receptors on bone mar-
row stromal cells: In vivo expression of vascu-
lar cell adhesion molecule-1 by reticular cells
and sinusoidal endothelium in normal and Á-
irradiated mice. Blood 87: 73–82.
Jilka, R.L., R.S. Weinstein, K. Takahashi, A.M.
Parfitt, S.C. Manolagas (1996) Linkage of de-
creased bone mass with impaired osteoblasto-
genesis in a murine model of accelerated senes-
cence. J Clin Invest 97: 1732–1740.
Johnstone, B., T.H. Hering, V.M. Goldberg, J.U.
Yoo, A.I. Caplan (1998) In vitro chondrogene-
sis of bone marrow-derived mesenchymal pro-
genitor cells. Exp Cell Res 238: 265–272.
Johnstone, B., J.U. Yoo (1999) Autologous mesen-
chymal progenitor cells in articular cartilage
repair. Clin Orthop 367S: 156–162.
Koc, O.N., S.L. Gerson, B.W. Cooper, S.M. Dy-
house, S.E. Haynesworth, A.I. Caplan, N.T.
Tainer, M.L. Lazarus (2000) Rapid hemato-
poietic recovery after co-infusion of autologous
blood stem cells and culture-expanded marrow
mesenchymal stem cells in advanced breast
cancer patients receiving high dose chemother-
apy. J Clin Oncol 18: 307–316.
Krossnes, B.K., S. Geisler, O. Mella (1998) Intrave-
nous infusion of glucose and the vasodilator
sodium nitroprusside in combination with lo-
cal hyperthermia: Effect on tumor pH and
tumor response in the BT4An rat tumor model.
Int J Hyperthermia 14: 403–416.
20
Cells Tissues Organs 2001;169:12–20
Gao/Dennis/Muzic/Lundberg/Caplan
Krossnes, B.K., O. Mella, O. Dahl (1996) Use of the
vasodilator sodium nitroprusside during local
hyperthermia: Effects on tumor temperature
and tumor response in a rat tumor model. Int J
Radiat Oncol Biol Phys 36: 403–415.
Kuppen, P.J., A. Marinelli, J.A. Camps, E.K. Pau-
wels, C.J. van de Velde, G.J. Fleuren, A.M.
Eggermont (1992) Distribution of lymphokine-
activated killer (LAK) cells in Wag rats after
hepatic-artery or jugular vein infusion. Int J
Cancer 9: 266–270.
Kuyama, J., A. McCormack, A.J. George, B.T.
Heelan, S. Osman, J.R. Batchelor, A.M. Peters
(1997) Indium-111 labeled lymphocytes: Iso-
tope distribution and cell division. Eur J Nucl
Med 24: 488–496.
Lazarus, H.K., S.E. Haynesworth, S.L. Gerson,
N.S. Rosenthal, A.I. Caplan (1995) Ex vivo
expansion and subsequent infusion of human
bone marrow-derived stromal progenitor cells
(mesenchymal progenitor cells): Implications
for therapeutic use. Bone Marrow Transplant
16: 557–564.
Leneveu, H., F. Bremont, H. Rubie, et al (1999)
Respiratory function in children undergoing
bone marrow transplantation. Pediatr Pulmo-
nol 28: 31–38.
Lennon, D.P., S.E. Haynesworth, R.G. Young, J.E.
Dennis, A.I. Caplan (1995) A chemically de-
fined medium supports in vitro proliferation
and maintains the osteochondral potential of
rat marrow-derived mesenchymal stem cells.
Exp Cell Res 219: 211–222.
Li, K.J., M.S. Dilber, M.R. Abedi, B. Björkstrand,
C.I.E. Smith, H. Caroff, G. Gahrton, K.G.
Xanthopoulos (1995) Retroviral-mediated
gene transfer into human bone marrow stromal
cells: Studies of efficiency and in vivo survival
in SCID mice. Eur J Haematol 55: 302–306.
Oyama, M., A. Tatlock, S. Fukuta, K. Kavalkovich,
B. Johnstone, P.D. Robbins, C.H. Evans, C.
Niyibizi (1999) Retrovirally transduced bone
marrow stromal cells isolated from a mouse
model of human osteogenesis imperfecta (oim)
persist in bone and retain the ability to form
cartilage and bone after extended passaging.
Gene Ther 6: 321–329.
Parfitt, A.M., A.R. Vilanueva, J. Foldes, D.S. Rao
(1995) Relations between histologic indices of
bone formation: Implications for the pathogen-
esis of spinal osteoporosis. J Bone Miner Res
10: 466–473.
Pereira, R.F., K.W. Halford, W.D. O’Hara, D.B.
Leeper, B.P. Sokolov, M.D. Pollard, O. Bagas-
ra, D.J. Prockop (1995) Cultured adherent cells
from marrow can serve as long-lasting precur-
sor cells for bone, cartilage, and lung in irra-
diated mice. Proc Natl Acad Sci USA 92:
4857–4861.
Pereira, R.F., M.D. O’Hara, A.V. Laptev, K.W.
Halford, M.D. Pollard, R. Class, D. Simon, K.
Livenzey, D.J. Prockop (1998) Marrow stromal
cells as a source of progenitor cells for nonhe-
matopoietic tissues in transgenic mice with a
phenotype of osteogenesis inperfecta. Proc
Natl Acad Sci USA 95: 1142–1147.
Piersma, A.H., R.E. Ploemacher, G.M. Brockbank
(1983) Transplantation of bone marrow fibro-
blastoid stromal cells in mice via the intrave-
nous route. Br J Haematol 54: 285–290.
Pittenger, M.F., A.M. Mackay, S.C. Beck, et al
(1999) Multilineage potential of adult human
mesenchymal stem cells. Science 248: 143–
147.
Richardson, D.R. (1976) The microcirculation and
regulation of blood flow; in Richardson, D.R.
(ed): Basic Circulator Physiology. Boston, Lit-
tle, Brown, pp 101–136.
Selden, C. (1984) The pulmonary vascular bed as a
site for implantation of isolated liver cells in
inbred rats. Transplantation 38: 81–83.
Tavassoli, M. (1979) The marrow-blood barrier. Br
J Haematol 41: 297–302.
Tavassoli, M., C.L. Hardy (1990) Molecular basis
of homing of intravenously transplanted stem
cells to the marrow. Blood 76: 1059–1070.
Thakur, M.L. (1983) Radioisotopic labeling of
platelets: A historical perspective. Semin
Thromb Hemost 9: 79–85.
Tinker, J.H., J.D. Michenfelder (1976) Sodium ni-
troprusside: Pharmacology, toxicology and
therapeutics. Anesthesiology 45: 340–354.
Weimar, I.S., N. Miranda, E.J. Muller, A. Hekman,
J.M. Kerst, G.C. de Gast, W.R. Gerritsen
(1998) Hepatocyte growth factor/scatter factor
(HGF/SF) is produced by human bone marrow
stromal cells and promotes proliferation, adhe-
sion and survival of human hematopoietic pro-
genitor cells (CD34+). Exp Hematol 26: 885–
894.
Xu, T., L. Chuang, M. Yagami, J. Gasser, M.
Thiede (1999) Engraftment potential of trans-
planted mesenchymal stem cells in non-condi-
tioned rats. Am Soc Bone Miner Res SU168
S437.
Young, R.G., D.L. Butler, W. Weber, A.I. Caplan,
S.L. Gordon, D.J. Fink (1998) Use of mesen-
chymal stem cells in a collagen matrix for
Achilles tendon repair. J Orthop Res 16: 406–
413.
Zakhireh, B., M.L. Thakur, H.L. Malech, M.S. Co-
hen, A. Gottschalk, R.K. Root (1979) Indium-
111-labeled human polymorphonuclear leuko-
cytes: Viability, random migration, chemotax-
is, bactericidal capacity and ultrastructure. J
Nucl Med 20: 741–747.
... However, this approach has several disadvantages, including the risk of pulmonary embolism and the low percentage of injected cells that reach the target site. Instead, many cells become sequestered in the spleen, liver, kidneys, or lungs [38,39]. The most notable limitation of MPC therapy is the poor survival rate of the cells post-transplantation. ...
Article
Ovarian reserve diminishes with age, and older women experience a corresponding shift in sex hormone levels. These changes contribute to an age-dependent decrease in fertility and a decline in overall health. Furthermore, while survival rates following cancer treatment have improved for young female patients, a reduction in ovarian function due to the side effects of such treatments can be difficult to avoid. To date, no effective therapy has been recommended to preserve ovarian health in these patients. Mesenchymal progenitor cells (MPCs) are considered a promising option for cell therapy aimed at maintaining fertility and fecundity. Although MPCs derived from human adult tissues are recognized for their various protective effects against ovarian senescence, they are limited in quantity. Consequently, human pluripotent stem cell-derived MPCs (hPSC-MPCs), which exhibit high proliferative capacity and retain genetic stability during growth, have been utilized to delay reproductive aging. This review highlights the impact of hPSC-MPCs on preserving the functionality of damaged ovaries in female mouse models subjected to chemotherapy and natural aging. It also proposes their potential as a valuable cell source for fertility preservation in women with a variety of diseases.
... Another critical characteristic of MSCs linked to their retention in the lung following systemic administration. A previous study has mentioned that intravenous infusion of MSCs results in their homogeneous distribution into the lung parenchyma (Gao et al, 2001), ascribable to a well-de ned cell-trapping phenomenon happening in the lung microvasculature (Schrepfer et al, 2007). ...
Article
Aim: This study aimed to clarify the anti-fibrotic potential of bone marrow-derived mesenchymal stem cells (BM-MSCs), compared to conditioned media (CM), in amiodarone (AD)-induced lung fibrosis. Methods: A total of 64 Wistar rats were categorized into eight groups: negative control group, positive control group, 3 AD-challenged-BM-MSCs-treated groups (1, 2 and 4 months) and 3 AD- challenged-CM-treated groups (1, 2 and 4 months). Serum macrophage in􀃀 ammatory protein 2 (MIP-2) evels were measured. Gene expression levels of TGF-􀈕1, SMAD3 and S100A4, were estimated in the lung tissues. Results: Treatment with BM-MSCs/CM mediated a significant reduction in serum MIP-2 concentrations, while downregulating AD-induced up-regulation of lung TGF-􀈕1, SMAD3 and S100A4 gene expression levels. BM-MSCs transplantation revealed better effect than CM in mitigating lung fibrosis. Conclusion: BM-MSCs advance anti-fibrotic effect on lung fibrosis by targeting inflammatory response and TGF-B1 signaling.
... Терапия, получаемая пациентом, может оказывать влияние на миграционную способность МСК. Показано, что сосудорасширяющие средства и антикоагулянты, такие как гепарин, уменьшают захват МСК легкими и увеличивают количество МСК в других органах, в частности печени и красном костном мозге [18]. Однако процесс миграции МСК обусловлен, как было описано выше, профилем экспрессии специфических поверхностных молекул и их рецепторов, а не просто пассивным распространением по сосудистой сети. ...
Article
Full-text available
В последние десятилетия накапливается все больше данных о механизмах обновления и регенерации тканей, которые были бы невозможны без участия стволовых клеток. Доказано, что данные процессы во многих тканях осуществляются за счет тканеспецифичных стволовых клеток (ТСК), однако их получение, культивация и введение с терапевтической целью крайне затруднительны. Наряду с этим наибольший интерес представляют мезенхимальные стволовые клетки (МСК), которые, благодаря возможности их выделения, экспансии и мультипотентности, являются многообещающим терапевтическим агентом, уже доказавшим свою клиническую эффективность при различных нозологиях, в том числе в вопросах тканевой инженерии. Одной из особенностей МСК, введенных системно, является способность находить нишу в пораженной ткани и оставаться в ней, оказывая существенное влияние на воспаление, процессы ремоделирования ткани и ее регенеративный потенциал. Однако механизмы дифференцировки и миграции МСК, а также факторы, влияющие на эти процессы, раскрыты не полностью. В данном обзоре обобщены современные данные о механизмах миграции МСК и возможных путях ее улучшения.
... In order to overcome the pulmonary obstacle and improve targeting, pre-administration of vasodilators such as sodium nitroprusside have been used to increase blood vessel diameter to reduce cell entrapment in the lung [141,147]. Another approach is to use macrophage-derived extracellular vesicles or exosomes instead of macrophages to modify cell size. Exosomes are 40-100 nm particles, smaller than parental cells in diameter, but they inherit most of the characteristics of parental cells such as biocompatibility, targeting ability, and long-term circulation [148]. ...
Article
Full-text available
As a natural immune cell and antigen presenting cell, macrophages have been studied and engineered to treat human diseases. Macrophages are well-suited for use as drug carriers because of their biological characteristics, such as excellent biocompatibility, long circulation, intrinsic inflammatory homing and phagocytosis. Meanwhile, macrophages’ uniquely high plasticity and easy re-education polarization facilitates their use as part of efficacious therapeutics for the treatment of inflammatory diseases or tumors. Although recent studies have demonstrated promising advances in macrophage-based drug delivery, several challenges currently hinder further improvement of therapeutic effect and clinical application. This article focuses on the main challenges of utilizing macrophage-based drug delivery, from the selection of macrophage sources, drug loading, and maintenance of macrophage phenotypes, to drug migration and release at target sites. In addition, corresponding strategies and insights related to these challenges are described. Finally, we also provide perspective on shortcomings on the road to clinical translation and production.
... Therapies [2]. The limiting factors with respect to direct cell injections are the poor cell survival and localisation at the implantation site [3,4], whilst tissue engineered medicines require prolonged in vitro culture periods for the development of implantable products, which are associated with cell phenotypic drift [5,6] and extremely high manufacturing costs [7][8][9][10][11], making the therapies prohibitively expensive for healthcare providers and inaccessible to patients. ...
Article
Full-text available
Even though tissue-engineered medicines are under intense academic, clinical, and commercial investigation, only a handful of products have been commercialised, primarily due to the costs associated with their prolonged manufacturing. While macromolecular crowding has been shown to enhance and accelerate extracellular matrix deposition in eukaryotic cell culture, possibly offering a solution in this procrastinating tissue-engineered medicine development, there is still no widely accepted macromolecular crowding agent. With these in mind, we herein assessed the potential of gum Arabic, gum gellan, gum karaya, and gum xanthan as macromolecular crowding agents in WS1 skin fibroblast cultures (no macromolecular crowding and carrageenan were used as a control). Dynamic light scattering analysis revealed that all macromolecules had negative charge and were polydispersed. None of the macromolecules affected basic cellular function. At day 7 (the longest time point assessed), gel electrophoresis analysis revealed that all macromolecules significantly increased collagen type I deposition in comparison to the non-macromolecular crowding group. Also at day 7, immunofluorescence analysis revealed that carrageenan; the 50 µg/mL, 75 µg/mL, and 100 µg/mL gum gellan; and the 500 µg/mL and 1000 µg/mL gum xanthan significantly increased both collagen type I and collagen type III deposition and only carrageenan significantly increased collagen type V deposition, all in comparison to the non-macromolecular crowding group at the respective time point. This preliminary study demonstrates the potential of gums as macromolecular crowding agents, but more detailed biological studies are needed to fully exploit their potential in the development of tissue-engineered medicines.
... Although significant progress has been made in regenerative medicine, the delivery of stem cells to the target tissues is still lagging [7]. Traditional delivery methods such as intravenous injections or infusions can result in a large percentage of the cells being lost or destroyed before they reach the target tissue, limiting their efficacy [8]. Interventional radiology (IR) techniques have emerged as a promising solution to this problem [9]. ...
Article
Full-text available
Regenerative medicine is taking a step forward in treating multiple diseases. The possibility of renewing damaged tissues with stem cells has become a topic of interest in recent decades. Still a relatively new research topic, many issues in this discipline are being addressed, from cell culturing to the study of different graft materials, and, moreover, cell delivery. For instance, direct intravenous injection has a big downfall regarding its lack of precision and poorly targeted treatment. Trans-arterial and direct percutaneous infusion to the aimed tissue/organ are both considered ideal for reaching the desired region but require image guidance to be performed safely and precisely. In this context, interventional radiology becomes pivotal for providing different cell delivery possibilities in every case. In this review, we analyze different basic stem cell therapy concepts and the current and future role of interventional radiology with a focus on trans-arterial delivery.
... Among these effects, the immunomodulatory effects of MSCs are the focus of treatment strategies in regenerative medicine, including inflammatory diseases [5][6][7]. MSCs have the ability to migrate to the site of injury, but only 0.1-1% of transplanted MSCs have been found at the target site [8][9][10]. However, even if MSCs do not migrate to the site of injury, they can be exposed to inflammatory cytokines and secrete various paracrine factors, such as indoleamine 2,3-dioxygenase (IDO), prostaglandin E2, TSG-6, nitric oxide (NO), interleukin (IL)-6, IL-10, and IL-1 receptor antagonist (IL-1ra), to modulate the activities of various immune cells [11,12]. ...
Article
Full-text available
Mesenchymal stem cells (MSCs) regulate immune cell activity by expressing tumor necrosis factor-α (TNF-α)-stimulated gene 6 (TSG-6) in inflammatory environments; however, whether anti-inflammatory responses affect TSG-6 expression in MSCs is not well understood. Therefore, we investigated whether transforming growth factor-β (TGF-β) regulates TSG-6 expression in adipose tissue-derived stem cells (ASCs) and whether effective immunosuppression can be achieved using ASCs and TGF-β signaling inhibitor A83-01. TGF-β significantly decreased TSG-6 expression in ASCs, but A83-01 and the p38 inhibitor SB202190 significantly increased it. However, in septic C57BL/6 mice, A83-01 further reduced the survival rate of the lipopolysaccharide (LPS)-treated group and ASC transplantation did not improve the severity induced by LPS. ASC transplantation alleviated the severity of sepsis induced by LPS+A83-01. In co-culture of macrophages and ASCs, A83-01 decreased TSG-6 expression whereas A83-01 and SB202190 reduced Cox-2 and IDO-2 expression in ASCs. These results suggest that TSG-6 expression in ASCs can be regulated by high concentrations of pro-inflammatory cytokines in vitro and in vivo, and that A83-01 and SB202190 can reduce the expression of immunomodulators in ASCs. Therefore, our data suggest that co-treatment of ASCs with TGF-β or p38 inhibitors is not adequate to modulate inflammation.
Article
Full-text available
Wernicke’s encephalopathy (WE) is a major central nervous system disorder resulting from thiamine deficiency (TD) in which a number of brain regions can develop serious damage including the thalamus and inferior colliculus. Despite decades of research into the pathophysiology of TD and potential therapeutic interventions, little progress has been made regarding effective treatment following the development of brain lesions and its associated cognitive issues. Recent developments in our understanding of stem cells suggest they are capable of repairing damage and improving function in different maladys. This article puts forward the case for the potential use of stem cell treatment as a therapeutic strategy in WE by first examining the effects of TD on brain functional integrity and its consequences. The second half of the paper will address the future benefits of treating TD with these cells by focusing on their nature and their potential to effectively treat neurodegenerative diseases that share some overlapping pathophysiological features with TD. At the same time, some of the obstacles these cells will have to overcome in order to become a viable therapeutic strategy for treating this potentially life-threatening illness in humans will be highlighted.
Chapter
Full-text available
Cellular and cell-free therapies have provided novel therapeutic opportunities for treating various incurable diseases. This is because the mesenchymal stromal/stem cells (MSCs) produce a plethora of bioactive molecules able to target different biological pathways through extracellular vesicle-mediated paracrine mechanisms simultaneously. However, to share transcriptomic signatures with their origin tissue, it is expected that each MSC population has a unique molecular profiler. In this sense, to analyze the transcriptome of therapeutic cells, it is crucial to identify the molecular profiler of these cells to predict the potential clinical benefits promoted by these cells. Herein, we discuss the application of high-throughput RNA-sequencing (RNA-Seq) in the Pharma Intelligence Era, discussing and exemplifying how the combination of molecular biology with Analytics can revolutionize the Research, Development, & Innovation (RD&I) of advanced medicinal products.
Article
Introduction: Autologous cell-based therapies (CBT) to treat Chronic Kidney Disease (CKD) with diabetes are novel and can potentially preserve renal function and decelerate disease progression. CBT dosing schedules are in early development and may benefit from individual bilateral organ dosing and kidney-dependent function to improve efficacy and durability. The objective of this open-label, phase 2 randomized controlled trial (RCT) is to evaluate participants' response to rilparencel (Renal Autologous Cell Therapy-REACT®) following bilateral percutaneous kidney injections into the kidney cortex with a prescribed dosing schedule versus re-dosing based on biomarker triggers. Methods: Eligible participants with type 1 or 2 diabetes and CKD, eGFR 20-50 ml/min/1.73 m2, UACR 30-5000 mg/g, Hemoglobin > 10 g/dL and glycated hemoglobin A1c < 10% were enrolled. After a percutaneous kidney biopsy and bioprocessing ex vivo expansion of Selected Renal Cells, participants were randomized 1:1 into two cohorts determined by the dosing scheme. Cohort 1 receives two cell injections, one in each kidney three months apart, and Cohort 2 receives one injection and the second dose only if there is a sustained eGFR decline of ≥20 ml/min/1.73 m2 and/or UACR increase of ≥ 30% confirmed by re-testing. Conclusion: This multicenter phase 2 RCT is designed to investigate the efficacy and safety of rilparencel with bilateral kidney dosing and comparing two injection schedules with the potential of preserving or improving kidney function and delaying kidney disease progression among patients with Stages 3a-4 CKD with diabetes.
Article
Cell adhesion molecules (CAMs) play a key role in interactions between stromal and hematopoietic cells in bone marrow (BM) and in cell traffic through vascular endothelium. To examine the identity of CAMs involved in these processes in mouse BM, we have investigated the in vivo expression of vascular cell adhesion molecule-1 (VCAM-1) and its counter-receptor, very late antigen-4 (VLA-4). Radioiodinated monoclonal antibodies (MoAbs) detecting VLA-4 and VCAM-1 were injected intravenously. Antibody binding was detected in BM by light and electron microscope radioautography. VCAM-1 labeling was restricted to stromal reticular cells and endothelial cells lining BM sinusoids. VCAM- 1+ reticular cells formed patchy concentrations, especially in subosteal regions, associated with lymphoid, granulocytic, and erythroid cells. After gamma-irradiation to deplete hematopoietic cells, reticular cells and endothelial cells all showed VCAM-1 labeling in apparently increased intensity. VLA-4 labeling was shown by undifferentiated blast cells and lymphohematopoietic cells both in BM cell suspensions and in vivo, especially at reticular cell contact points. The results demonstrate that VCAM-1 is expressed in vivo by certain BM reticular cells, suggesting that the molecule mediates adhesion to multiple lineages of lymphohematopoietic cells. The finding that VCAM-1 is also expressed constitutively by BM sinusoidal endothelium, unlike its inductive expression by endothelia elsewhere, suggests that VCAM-1 and VLA-4 may be involved in regulating the normal cell traffic between BM and the blood stream.
Article
Since lymphocytes continue to proliferate and divide in vivo, it is important to determine the fate of a radionulide following lymphocyte labelling. Using the mixed lymphocyte reaction (MLR), we induced indium-111 labelled lymphocytes from a specific in-bred rat strain (AS) to divide and then observed the subsequent 111In distribution between cells and supernatant. L10 and L12.4 cells, which are allospecific CD4+ T lymphocytes from the AS rat, were stimulated in the MLR by antigen-presenting cells from the August rat, a different strain. We labelled L10 or L12.4 lymphocytes on day 0, the first day of the stimulation cycle, and continued to culture the lymphocytes in vitro. The proliferation of the cells was estimated according to their increase in number. The distribution of 111In between cell and supernatant fractions and between viable and dead (but intact) cells was measured in the cell suspension each day after labelling. The metabolic activity of 111In-labelled lymphocytes was compared with control cells by measuring their uptake of fluorine-18 fluorodeoxyglucose ([18F]FDG). 111In-labelled lymphocytes showed a poor proliferative response compared with control cells 24-48&#114h after labelling but increased in number after this time. From 24 to 72&#114h, about 70% of 111In was in the supernatant but only about 5%-10% was associated with intact dead cells. These dead cells tended to retain their 111In, losing less than 30% per day, suggesting that 111In in the supernatant was the result of active elimination from viable cells. Moreover, 24&#114h after culture, considerably more 111In was associated with viable than with dead lymphocytes, although over the next few days this distribution reversed. 111In-labelled lymphocytes took up more [18F]FDG than control cells at 24&#114h but not at 0 or 72-96&#114h; the maximum [18F]FDG uptake coincided with the greatest reduction in cell number. Furthermore, [18F]FDG uptake correlated with the initial 111In burden in lymphocytes labelled with 111In 24&#114h previously. The results are consistent with active elimination of 111In by 111In-labelled lymphocytes. The energy requirements for this are diverted away from cell division, thereby increasing the probability of cell death. As lymphocytes become 111In deplete, they recover their capacity to proliferate and their risk of death decreases. These findings have important implications for 111In-labelled lymphocyte scintigraphy, suggesting that cells remaining viable immediately after labelling will either subsequently die or alternatively eliminate the label.
Article
Bone marrow contains a population of rare progenitor cells capable of differentiating into bone, cartilage, muscle, tendon, and other connective tissues. These cells, referred to as MSCs, can be purified and culture expanded from animals and humans. This review summarizes recent experimentation focused on characterizing the cellular aspects of osteogenic differentiation, and exploration of the potential for using autologous stem cell therapy to augment bone repair and regeneration. The authors have completed an array of preclinical studies showing the feasibility and efficacy of MSC based implants to heal large osseous defects. After confirming that syngeneic rat MSCs could heal a critical size segmental defect in the femur, it was established that human MSCs form bone of considerable mechanical integrity when implanted in an osseous defect in an immunocompromised animal. Furthermore, bone repair studies in do gs verify that the technology is transferable to large animals, and that the application of this technology to patients at geographically remote sites is feasible. These studies suggest that by combining MSCs with an appropriate delivery vehicle, it may be possible to offer patients new therapeutic options.
Article
Recent studies have demonstrated the existence of a subset of cells in human bone marrow capable of differentiating along multiple mesenchymal lineages. Not only do these mesenchymal stem cells (MSCs) possess multilineage developmental potential, but they may be cultured ex vivo for many passages without overt expression of a differentiated phenotype. The goals of the current study were to determine the growth kinetics, self-renewing capacity, and the osteogenic potential of purified MSCs during extensive subcultivation and following cryopreservation. Primary cultures of MSCs were established from normal iliac crest bone marrow aspirates, an aliquot was cryopreserved and thawed, and then both frozen and unfrozen populations were subcultivated in parallel for as many as 15 passages. Cells derived from each passage were assayed for their kinetics of growth and their osteogenic potential in response to an osteoinductive medium containing dexamethasone. Spindle-shaped human MSCs in primary culture exhibit a lag phase of growth, followed by a log phase, finally resulting in a growth plateau state. Passaged cultures proceed through the same stages, however, the rate of growth in log phase and the final number of cells after a fixed period in culture diminishes as a function of continued passaging. The average number of population doublings for marrow-derived adult human MSCs was determined to be 38 ± 4, at which time the cells finally became very broad and flattened before degenerating. The osteogenic potential of cells was conserved throughout every passage as evidenced by the significant increase in APase activity and formation of mineralized nodular aggregates. Furthermore, the process of cryopreserving and thawing the cells had no effect on either their growth or osteogenic differentiation. Importantly, these studies demonstrate that replicative senescence of MSCs is not a state of terminal differentiation since these cells remain capable of progressing through the osteogenic lineage. The use of population doubling potential as a measure of biological age suggests that MSCs are intermediately between embryonic and adult tissues, and as such, may provide an in situ source for mesenchymal progenitor cells throughout an adult's lifetime. J. Cell. Biochem. 64:278–294. © 1997 Wiley-Liss, Inc.
Article
One approach in the treatment of ovarian cancer MOv18/anti-CD3 (biMAb OC/TR), which recognizes a 38-kDa glycoprotein expressed on ovarian carcinomas and the CD3 T cell receptor. However, little is known about the in vivo biodistribution of injected activated lymphocytes, information that could be obtained by scintigraphic imaging of radiolabelled T cells in order to visualize the migratory pattern. We compared the efficiency, stability and toxicity of technetium-99m hexamethylpropylene amine oxime (HMPAO),indium-111 oxine and fluorine-18 2-fluoro-2-deoxy-d-glucose (FDG) in radiolabelling activated lymphocytes targeted with biMAb OC/TR. The mean labelling efficiencies of111In-oxine and18F-FDG using 2.5×108 lymphocytes (68% and 64%, respectively) were more than twice that of99mTc-HMPAO (31%). Retention of the radionuclide in the cell was highest in the case of111In-oxine labelling (less than 25% of the initial cell-bound activity released after 240 min, as compared with 44% of the99mTc label in the same period and 45% of18F radionuclide released after 150 min). None of the three radiolabelling reagents induced any significant alteration in cell viability or immunophenotype. However, both111In-oxine and18F-FDG induced a loss of cytotoxic activity of lymphocytes against the ovarian carcinoma cell line IGROV1, and all three radiolabelling reagents caused a significant reduction in the proliferative ability of labelled lymphocytes compared to controls, with cell death occurring after 8–9 days. Radiolabelling with the more stable111In-oxine reagent using a higher number of lymphocytes (1.4x109) but the same total activity (around 55.5 MBq) resulted in improved labelled T cell viability and proliferative ability, although the mean labelling efficiency decreased (35.8%). Together the data suggest that111In-oxine at low activity per cell is the most appropriate reagent for radiolabelling activated retargeted T lymphocytes useful for in vivo biodistribution studies.
Article
A culture system that facilitates the chondrogenic differentiation of rabbit bone marrow-derived mesenchymal progenitor cells has been developed. Cells obtained in bone marrow aspirates were first isolated by monolayer culture and then transferred into tubes and allowed to form three-dimensional aggregates in a chemically defined medium. The inclusion of 10−7M dexamethasone in the medium induced chondrogenic differentiation of cells within the aggregate as evidenced by the appearance of toluidine blue metachromasia and the immunohistochemical detection of type II collagen as early as 7 days after beginning three-dimensional culture. After 21 days, the matrix of the entire aggregate contained type II collagen. By 14 days of culture, there was also evidence for type X collagen present in the matrix and the cells morphologically resembled hypertrophic chondrocytes. However, chondrogenic differentiation was achieved in only approximately 25% of the marrow cell preparations used. In contrast, with the addition of transforming growth factor-β1 (TGF-β1), chondrogenesis was induced in all marrow cell preparations, with or without the presence of 10−7M dexamethasone. The induction of chondrogenesis was accompanied by an increase in the alkaline phosphatase activity of the aggregated cells. The results of RT-PCR experiments indicated that both type IIA and IIB collagen mRNAs were detected by 7 days postaggregation as was mRNA for type X collagen. Conversely, the expression of the type I collagen mRNA was detected in the preaggregate cells but was no longer detectable at 7 days after aggregation. These results provide histological, immunohistochemical, and molecular evidence for thein vitrochondrogenic differentiation of adult mammalian progenitor cells derived from bone marrow.
Article
Among the stromal elements in mammalian and avian bone marrow there exists a pluripotent subset of cells which we refer to as mesenchymal stem cells (MSCs). These cells can be isolated and will proliferate in culture. When such subcultured cells are introduced into porous tricalcium phosphate-hydroxyapatite ceramic cubes and implanted subcutaneously into syngeneic or immunocompromised hosts, the passaged MSCs are observed to differentiate into bone and cartilage. Heretofore, those assays have been conducted with MSCs which had been maintained in vitro in serum-containing medium. A serum-free medium (RDM-F), which consists of insulin, 5 μg/ml, linoleic acid-bovine serum albumin, 0.1%, platelet-derived growth factor-BB, 10 ng/ml, and basic fibroblast growth factor, 1 ng/ml in a base medium of 60% Dulbecco's modified Eagle's medium with low glucose and 40% MCDB-201, has been developed for rat marrow-derived MSCs. Proliferation rates of MSCs maintained in RDM-F equal those of cells maintained in serum-containing medium through Day 4 following subculturing and continue at up to 80% of the rate of the latter through Day 8 of subculture. When tested in the in vivo ceramic cube assay, MSCs cultured in RDM-F retain their osteochondral potential and differentiate into bone and cartilage in a manner indistinguishable from those cultivated in serum-containing medium. Utilization of this serum-free medium will facilitate analysis of the effects of other growth factors and cytokines on the proliferation and differentiation of MSCs, without the complexity of exogenous serum.
Article
Attempts to develop an ex vivo gene therapy strategy for hemophilia A, using either primary T cells or bone marrow (BM) stem/progenitor cells have been unsuccessful, due to the inability of these cell types to express coagulation factor VIII (FVIII). As an alternative, we evaluated the potential of BM-derived stromal cells which can be readily obtained and expanded in vitro. Human and murine BM stromal cells were transduced with an intron-based Moloney murine leukemia virus (MoMLV) retroviral vector expressing a B-domain-deleted human factor VIII cDNA (designated as MFG-FVIIIdeltaB). Transduction efficiencies were increased 10- to 15-fold by phosphate depletion and centrifugation, which obviated the need for selective enrichment of the transduced BM stromal cells. This resulted in high FVIII expression levels in transduced human (180 +/- 4 ng FVIII/10[6] cells per 24 hr) and mouse (900 +/- 130 ng FVIII/10[6] cells per 24 hr) BM stromal cells. Pseudotyping of the MFG-FVIIIdeltaB retroviral vectors with the gibbon ape leukemia virus envelope (GALV-env) resulted in significantly higher transduction efficiencies (100 +/- 20%) and FVIII expression levels (390 +/- 10 ng FVIII/10[6] cells per 24 hr) in transduced human BM stromal cells than with standard amphotropic vectors. This difference in transduction efficiency correlated with the higher titer of the GALV-env pseudotyped viral vectors and with the higher GALV receptor (GLVR-1) versus amphotropic receptor (GLVR-2) mRNA expression levels in human BM stromal cells. These findings demonstrate the potential of BM stromal cells for gene therapy in general and hemophilia A in particular.
Article
Human polymorphonuclear leukocytes (PMNs) were labeled with indium-111 oxine in ethanol, and the effects of the labeling procedure, radioactivity, and concentrations of oxine and ethanol on PMN function and structure were studied in vitro. The standard labeling procedure did not alter the viability, random migration, chemotaxis, bactericidal capacity, or the ultrastructure of PMNs. Exposure to higher doses of radioactivity, or to higher concentrations of ethanol, had no appreciable effects on random migration and chemotaxis of PMNs. A dose-dependent reduction in their random migration and chemotaxis was observed when higher concentrations of oxine were used. These results indicate that In-111-labeled PMNs are structurally intact and have normal in vitro locomotion and bactericidal activity. Indium-111-labeled PMNs should be suitable for studying the kinetics and distribution of these cells in health and disease.