Вы находитесь на странице: 1из 17

Cell Transplantation, Vol. 24, pp. 1981–1997, 2015 0963-6897/15 $90.00 + .

00
Printed in the USA. All rights reserved. DOI: http://dx.doi.org/10.3727/096368914X685302
Copyright Ó 2015 Cognizant Comm. Corp. E-ISSN 1555-3892
www.cognizantcommunication.com

Comparison of Magnetic Intensities for Mesenchymal Stem Cell Targeting


Therapy on Ischemic Myocardial Repair: High Magnetic Intensity Improves
Cell Retention but Has no Additional Functional Benefit
Yunli Shen,*1 Xuebo Liu,*1 Zheyong Huang,† Ning Pei,‡ Jianfeng Xu,† Zheng Li,§ Yunkai Wang,*
Juying Qian,† and Junbo Ge†

*Department of Cardiology, Shanghai East Hospital, Tongji University, Shanghai, China


†Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
‡College of Science, Shanghai University, Shanghai, China
§Biomedical Research Center, Zhongshan Hospital, Fudan University, Shanghai, China

Magnetic targeting has the potential to enhance the therapeutic effects of stem cells through increasing reten-
tion of transplanted cells. To investigate the effects of magnetic targeting intensities on cell transplantation, we
performed different magnetic intensities for mesenchymal stem cell (MSC)-targeting therapy in a rat model of
ischemia/reperfusion. Rat MSCs labeled with superparamagnetic oxide nanoparticles (SPIOs) were injected
into the left ventricular (LV) cavity of rats during a brief aorta and pulmonary artery occlusion. The 0.15
Tesla (T), 0.3 T, and 0.6 T magnets were placed 0~1 mm above the injured myocardium during and after
the injection of 1 × 106 MSCs. Fluorescence imaging and quantitative PCR revealed that magnetic targeting
enhanced cell retention in the heart at 24 h in a magnetic field strength-dependent manner. Compared with the
0 T group, three magnetic targeting groups enhanced varying cell engraftment at 3 weeks, at which time LV
remodeling was maximally attenuated, and the therapeutic benefit (LV ejection fraction) was also highest in
the 0.3 T groups. Interestingly, due to the low MSC engraftment resulting from microvascular embolisms, the
0.6 T group failed to translate into additional therapeutic outcomes, though it had the highest cell retention.
Magnetic targeting enhances cell retention in a magnetic field strength-dependent manner. However, too high
of a magnetic intensity may result in microembolization and consequently undermine the functional benefits
of cell transplantation.

Key words: Mesenchymal stem cells; Magnetic targeting intensity; Microembolization; Myocardial
infarction; Rat model

INTRODUCTION delivery of cardiosphere-derived cells (CDCs) (6) or


Cellular therapies are an attractive option as adjunct endothelial progenitor cells (EPCs) (4) in a rat model of
therapies for acute or chronic ischemic heart diseases (26). acute ischemia/reperfusion (I/R). In Cheng et al.’s study
Intracoronary cell injection is the more clinically relevant (6), the actual working intensity was theoretically esti-
and less invasive setting of an intravascular approach mated to be less than 0.3 Tesla (T) for a 1.3 T magnet
than intramyocardial injection. However, the therapeutic that was placed ~1 cm above the heart, and the magnet
effect of intracoronary cell transplantation is extremely group exhibited greater than a fourfold greater cell reten-
restricted by low cell retention and engraftment in injured tion. In Chaudeurge et al.’s study (4), a 0.1 T magnet
myocardium (10,24). was implanted into the subcutaneous layer, leading to
Magnetic targeting is an emerging approach to augment an actual working intensity that was far less than 0.1 T,
cellular therapies by guiding cells to sites of injury using and magnetic targeting failed to increase cell retention
externally generated magnetic fields and field gradients using RT-PCR quantification. Our previous in vitro study
(3). Using different magnetic forces, two animal studies demonstrated that cell capture efficiency was positively
recently tested the magnetically enhanced intracoronary related to the magnetic flux density between 10 and

Received July 24, 2013; final acceptance September 20, 2014. Online prepub date: November 5, 2014.
1
These authors provided equal contribution to this work.
Address correspondence to Juying Qian, M.D., Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Feng Lin Road,
Shanghai 200032, China. Tel: +86-21-64041990, x10564; Fax: +86-21-64223006; E-mail: juyingqian@126.com or Zheyong Huang, M.D., Shanghai
Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Feng Lin Road, Shanghai 200032, China. Tel: +86-21-64041990, x12596;
Fax: +86-21-64223006; E-mail: zheyonghuang@126.com
1981
1982 SHEN ET AL.

640 mT, and it reached 89.3% with a magnetic flux den- (potassium ferrocyanide and HCl solution from Jueshen
sity of 640 mT, a magnetic intensity gradient of 38.4 T/m, Bio-Technology Co. Ltd., Shanghai, China) and electron
and a flow velocity of 0.8 mm/s (14). microscopy (Phillips CM120, Philips FEI, Eindhoven,
Based on the above data, it seems that cell retention Netherlands) were undertaken to indicate the presence and
and engraftment can continuously increase with the localization of intracellular iron particles, and DiR stain-
enhancement of magnetic force, subsequently translat- ing was observed by fluorescence microscopy using the
ing into additional cardiac functional and structural ben- XF112-Omega optical filter after cell nuclei were coun-
efits. This notion also seems to be evidenced by some terstained with 4¢,6-diamidino-2-phenylindole (DAPI;
early studies without magnetic targeting, in which the Sigma-Aldrich, St. Louis, MO, USA) (16).
relationship between injected cell number and func-
tional effect was a positive linear correlation regardless Effects of SPIO Labeling on MSC
of intramyocardial injection or intracoronary delivery Differentiation Capacity
(17,22,23,27). However, intracoronary delivery of mes- Our previous study demonstrated that SPIO labeling
enchymal stem cells (MSCs) can induce microvascular had no significant effect on the proliferation and viabil-
obstruction as has been reported previously (11,29,30), ity of MSCs (16). Here we further determined the dif-
and little is known about whether microembolisms would ferentiation capacity of SPIOs-MSCs. The MSCs and
be aggravated with the escalating magnetic intensity and SPIOs-MSCs were, respectively, cultured in adipogenic
thereby have an influence on therapeutic outcome of (SR811D250, Amsbio, Abingdon, UK) or osteogenic
transplanted cells. Here, we investigated the effects of (SR417D250, Amsbio) media for 21 days with media
magnetic targeting intensities on intraventricular MSC replenishment every 3 days. To demonstrate adipogenic
delivery in a rat I/R model. differentiation, the cells were stained with 0.3% Oil red O
(O0625; Sigma-Aldrich) in isopropanol for 30 min and
MATERIALS AND METHODS
rinsed with PBS. For osteogenic differentiation, the cells
Magnet Cylinder were stained with 1% Alizarin red (500-4; RiccaChem,
Three cylindrical NdFeB magnets (Shanghai Yahao Arlington, TX, USA) for 15 min (2).
Instrument Equipment Co., Shanghai, China) with 0.15 T, To determine the myocardium-like cell differentiation,
0.3 T, and 0.6 T (diameter 8 mm) were used in this study. the MSCs and SPIOs-MSCs were cultured in culture
The magnetic flux density (B) of the magnet surface is medium-added 5-aza-2¢-deoxycytidine (5-azaC; Sigma-
measured using a model 51,662 Leybold Tesla meter Aldrich) for 24 h. Incubation then continued in complete
(Beijing Zhuo Sheng Jia Magnetic Technology Co. Ltd., medium lacking 5-azaC, with medium changes every 3 days.
Beijing, China). The distribution of the magnetic flux Undifferentiated cells and normal mature rat cardiomyo-
density was calculated by a finite element analysis. cytes were, respectively, used as negative and positive
controls. Total RNA was extracted from cultured cells
Rat MSCs, SPIOs, and DiR Labeling using TRIzol reagent (Invitrogen, Carlsbad, CA, USA)
Animal experiments were approved by the Animal Care according to the manufacturer’s protocol. RT-PCR prim-
and Use Committee of Fudan University and were in com- ers (synthesized by Sangon, Shanghai, China) were as fol-
pliance with the Guide for the Care and Use of Laboratory lows: GATA-4, 5¢-CTG TCA TCT CAC TAT GGG CA-3¢
Animals published by the National Academy Press (1). and 5¢-CCA AGT CCG AGC AGG AAT T T-3¢; a-actin,
Femurs of 4-week-old male Sprague–Dawley (SD) rats, (F) 5¢-TCC TTT ATC GGT ATG GAG TCT G-3¢ and
obtained from Shanghai animal administration center, (R) 5¢-TGA TCT TGA TCT TCA TGG TGC T-3¢; cTnT,
were excised under sterile conditions. MSCs were cul- 5¢-AGA GGA CTC CAA ACC CAA GC-3¢ and 5¢-ATT
tured from bone marrow plugs explanted from 4-week-old GCG AAT ACG CTG CTG TT-3¢; Cx43, 5¢-TCC TTG
male SD rats, as previously described (34). All used cells GTG TCT CTC GCT TT-3¢ and 5¢-GAG CAG CCA TTG
were harvested by 0.25% trypsin (Gibco, Grand Island, AAG TAG G C-3¢; GAPDH, 5¢-GAC ATG CCG CCT
NY, USA) and 0.038% EDTA (Gibco) when they reached GGA GAA AC-3¢ and 5¢-AGC CCA GGA TGC CCT
80–90% confluence at passage 4. MSCs were labeled with TTA GT-3¢. Transcriptional expressions of GATA-4, cTnT,
superparamagnetic oxide nanoparticles (SPIOs; 100 mg/ a-actin, and Cx43 genes from the MSCs and SPIOs-MSCs
ml; Schering, Berlin, Germany), as previously described were determined by real-time PCR according to the manu-
(14). For cell tracking in vivo, cells were labeled with 1,1- facturer’s instructions. Transcript levels were standardized
dioctadecyl-3,3,3,3-tetramethyl indotricarbocyanine iodide to the corresponding rat GAPDH level.
(DiR; AAT Bioquest, Inc., Sunnyvale, CA, USA) accord-
ing to the manufacturer’s protocol. Labeled cells are here- In Vitro Magnetic Capture of Flowing SPIOs-MSCs
after called SPIOs-DiR-MSCs or DiR-MSCs for brevity. To test the magnetic responsiveness of MSCs labeled
To evaluate the labeling efficacy, Prussian blue staining with SPIOs (SPIOs-MSCs), the accumulation of SPIOs-
MAGNETIC INTENSITY FOR STEM CELL TARGETING 1983

MSCs was observed, while cells flowed through a tube time (5,6). The 217 survivors were randomized into six
served as a model of blood vessels in a magnetic field. A groups according to the strength of the magnetic field
total of 20 ml SPIOs-MSCs suspension, at a concentra- (n = 37 for 0.3 T group, n = 36 for other groups): 0 T group,
tion of 5 × 104 cells/ml, was placed in a 50-ml syringe 0.15 T group, 0.3 T group, 0.6 T group, MSC group, and
(Shanghai Boguang Biotechnology Co. Ltd., Shanghai, PBS group. All magnets were close to the target site, that
China) and flowed through a quartz tube (ID 2.3 mm, is, within 0–1 mm, during and after infusion in the three
OD 4.3 mm, length 20 cm) (Shanghai Puya Quartz Glass magnetic targeting groups. Except for the PBS group,
Factory, Shanghai, China). Cell suspension circulated which was intraventricularly injected with vehicle (PBS)
at flows of 5 mm/s to mimic circulatory conditions in only, the other treatment groups were intraventricularly
animals and human beings. The aforementioned 0.15-T injected with 1 × 106 SPIOs-DiR-MSCs (0 T, 0.15 T, 0.3 T,
magnet cylinder was placed tightly at the midsegment of and 0.6 T group) or DiR-MSCs (MSC group). All animals
the tube. The cell suspension flowed through the mag- received humane care in compliance with the Guide for
net field was collected in a centrifuge tube (Shanghai the Care and Use of Laboratory Animals.
Boguang Biotechnology Co. Ltd.) 1#. The remaining
cells within the quartz tube were washed using PBS and Fluorescence Imaging
collected in centrifuge tube 2#. The numbers of SPIOs- After assessment of cardiac function using trans-
MSCs in centrifuge tube 1# and 2# were counted using a thoracic echocardiography, six animals were randomly
counting plate and referred to quantity of captured cells selected from each treatment group and were eutha-
(Q1) and quantity of uncaptured cells (Q2). The capture nized 24 h after cell injection for fluorescence imaging.
Q1 Extensive PBS washing was performed to remove any
efficiency (CE) was calculated by CE = × 100%.
Q1 + Q2 cells adherent to the surface of organs. Hearts, lungs, livers,
Then, the effects of the 0.3-T and 0.6-T magnets on the spleens, and kidneys were placed in a Carestream In-Vivo
accumulation of SPIOs-MSCs in turn were observed. Multispectral Imaging System FX PRO (Carestream
The magnetic responsiveness of MSCs unlabeled with Health, Toronto, Canada) to detect DiR fluorescence under
SPIOs was tested according to the method mentioned 748 nm of excitation and 780 nm of emission. Exposure
above. All experiments were performed in six copies for time was set at 3 s and kept the same during the entire
each condition. imaging session. Organs from the PBS group (i.e., the
animals receiving normal saline) were also imaged as
Animal Model and Magnetic Targeting controls for background noise. Fluorescence signals
To make our experiment more relevant to reperfused (photon/s) from a fixed region of interest (ROI) were
acute myocardial infarction (AMI), a rat I/R model was measured as described (36).
developed in female SD rats (150–200 g). Rats were
anesthetized with ketamine (100 mg/kg, IP) (Beijing Quantification of Engraftment by Real-Time PCR
Zizhu Pharmaceutical Co. Ltd., Beijing, China) and Quantitative PCR was performed 24 h (after fluo-
xylazine (10 mg/kg, IP) (Shanghai Ruicong Laboratory rescence imaging and assessment of cardiac function),
Equipment Co. Ltd., Shanghai, China) and were venti- 10 days, and 3 weeks (after fluorescence imaging and
lated with a rodent ventilator using room air at 80 breaths assessment of cardiac function) after cell injection in six
per minute. Female SD rats (n = 236 total), obtained from animals from each cell-injected group to quantify cell
Shanghai animal administration center, underwent a left retention/engraftment. We injected MSCs from male donor
thoracotomy in the fourth intercostal space under gen- SD rats into the left ventricle of female recipients, uti-
eral anesthesia. The heart was exposed, and myocardial lizing the sex-determining region Y (SRY) gene located
infarction was produced by a 90-min ligation of the left on the Y chromosome as a detection target (28). The
anterior descending coronary artery (LAD) using a 7-0 whole heart was harvested, weighed, and homogenized.
silk suture (Unik Surgical Sutures Mfg. Co. Ltd., Suzhou, Genomic DNA was isolated from aliquots of the homoge-
China). The suture was then released to allow coronary nate corresponding to 12.5 mg of myocardial tissue using
reperfusion. Twenty minutes later, cells were injected into commercial kits (DNA Easy minikit, Qiagen, Hilden,
the left ventricle cavity during a 5-s temporary aorta and Germany). The TaqMan® assay (Applied Biosystems,
main pulmonary artery occlusion, as previously described Carlsbad, CA, USA) was used to quantify the number of
(18,19). Cell delivery was confirmed by the temporary transplanted cells with the rat SRY gene as a template
observation of bradycardia and epicardial blanching. [forward primer: 5¢-GAG CTT TGG GAG CAG TGA
For magnetic targeting, three cylindrical NdFeB mag- C-3¢ (TM 55.1), reverse primer 5¢- ATG AGG CTG ATA
nets with 0.15 T, 0.3 T, and 0.6 T were separately placed TTT ATA GTT TGG-3¢ (TM 51.5), Taq Man probe: 6
above the injured myocardium during and after cell injec- FAM CAA CAG AAT CCC AGC ATG CAG AAT TCA
tion for 10 min, which was determined to be the optimal G TAMRA; Applied Biosystems]. A standard curve was
1984 SHEN ET AL.

generated with multiple dilutions of genomic DNA iso- echocardiography of the rat. Cryostat sections (8 mm
lated from male hearts to quantify the absolute gene copy thickness) were obtained for immunofluorescence stain-
numbers. All samples were spiked with equal amounts ing to evaluate cardiomyocyte or endothelial differentia-
of female genomic DNA as a control. The copy number tion of injected MSCs. Potential transdifferentiation of
of the SRY gene at each point of the standard curve was myocardium-like cells from implanted MSCs was veri-
calculated with the amount of DNA in each sample and fied by antibody immunostaining for troponin T (cTnT).
the mass of the rat genome per cell. For each reaction, Briefly, frozen tissue sections were fixed in acetone at
50 ng of template DNA was used. Real-time PCR was 4°C for 10 min and incubated separately with a mouse
performed using an Applied Biosystems 7900 HT Fast anti-rat cTnT (TnT, cardiac isoform Ab-1; Labora-
real-time PCR System. All experiments were performed tory Vision Corp, Chicago, IL, USA) diluted 1:100 for
in triplicate. Cell numbers per milligram of heart tissue 60 min at room temperature. After washing with PBS
and percentages of retained cells of the total injected cells solution, sections were incubated with a goat anti-mouse-
(1 × 106) were calculated. conjugated FITC IgG (Molecular Probes, Inc., Eugene,
OR, USA) diluted 1:200 for cTnT. Goat anti-rat CD31
Assessment of Cardiac Function (BD Biosciences, San Diego, CA, USA) was diluted
Postinfarct cardiac function was evaluated by transtho- 1:100 to check for evidence of angiogenesis. Secondary
racic echocardiography using a Vevo 770 high-resolution antibody of donkey anti-goat IgG conjugated with FITC
imaging system (Visual Sonics, Toronto, Canada) with a (Molecular Probes) diluted 1:200 was used. Cell nuclei
17.5-MHz probe 24 h and 3 weeks after I/R and treat- were counterstained with DAPI. Capillary density was
ment (n = 13 for the 0.3 T group, n = 12 for other groups). also assessed by staining with anti-CD31 antibody as pre-
After the induction of light general anesthesia, hearts viously described (31). A total of 10 visual fields where a
were imaged in 2D and M-mode. The recordings were cross-section of capillaries was clearly visible were ran-
obtained from the parasternal long axis view at the level domly selected in the peri-infarct zone, and the number
of the greatest LV diameter. The LV internal end-diastolic of capillaries was counted under 200× magnification by a
diameter (LVIDd) and LV internal end-systolic diameter Leica Fluorescence microscope (Leica DM LB2).
(LVIDs) were measured from M-mode recordings. The For morphometric analysis, representative recipient
LV ejection fraction (LVEF) was calculated as follows: hearts in each group were harvested at 3 weeks (after
LVEF (%) = [(LVIDd)3 − (LVIDs)3]/(LVIDd)3 × 100. All cardiac function assessment) and embedded in paraffin.
echocardiographic measurements were averaged from at Masson’s trichrome staining was performed as described
least five separate cardiac cycles. by the manufacturer’s instructions [HT15 Trichrome
Staining (Masson) Kit; Sigma-Aldrich]. To determine
Immunohistochemistry and Morphometric the infarct size, the average ratio of infarct size per LV
Heart Analysis was measured for all paraffin slides after staining with
At 72 h after cell injection, six representative recipient Masson’s trichrome as previously described (21). All
hearts from each group embedded in paraffin were cut analyses were performed in a blinded manner.
into 5-µm slices, which were stained with Prussian blue
(Shanghai HuaYi Bio Technology Co. Ltd., Shanghai, Statistical Analysis
China). The representative hearts were harvested and fro- Animal numbers used in each experiment are shown
zen in OCT compound (Leica JUNG, Wetzlar, Germany) in Table 1. All data are shown as mean ± standard devia-
72 h after transplantation and 3 weeks subsequent to tion (SD). The difference between mean values was
Table 1. Study Protocol and Rat Numbers Used in Each Experiment
24 Hours 72 Hours 10 Days 21 Days
Heart
Fluorescence/ Prussian Cell PCR Masson Staining/
PCR for SRY Blue Staining Fluorescence for SRY Immunofluorescence PCR for SRY
PBS group 6 6 6 6 6 6
MSC group 6 6 6 6 6 6
0 T group 6 6 6 6 6 6
0.15 T group 6 6 6 6 6 6
0.3 T group 6 6 6 6 7 6
0.6 T group 6 6 6 6 6 6
Twelve animals from each group (except for 13 animals in 0.3 T group) underwent echocardiography at 24 h and 21 days after injection, respectively.
SRY, sex-determining region Y gene; PBS, phosphate-buffered saline (control); MSC, mesenchymal stem cell; T, Tesla (magnetic field strength).
MAGNETIC INTENSITY FOR STEM CELL TARGETING 1985

determined with one-way analysis of variance (ANOVA) 21 days in media that promote differentiation into adi-
and Bonferroni post hoc test. The correlation analysis was pogenic and osteogenic lineages. As shown in Figure 2A
based on the Pearson correlation coefficient. Statistical and B, both MSCs and SPIOs-MSCs differentiated into
significance was set at p < 0.05. the two lineages, as demonstrated by staining for Oil
red O and Alizarin red, respectively. Following myo-
RESULTS cardial differentiation, real-time PCR showed that myo-
Labeling of Rat MSCs With SPIOs and DiR cardial genes GATA-4, cTnT, a-actin, and Cx43 were
The fourth passage of MSCs was characterized by both detected in differentiated MSCs and SPIOs-MSCs
fusiform or spindle-shaped morphology (Fig. 1A). The (all p > 0.05) (Fig. 2C). The expression of these myocar-
SPIOs-DiR-MSCs had a strong red fluorescence sig- dial genes was comparable in differentiated MSCs and
nal (Fig. 1B). The Prussian blue staining of the MSCs SPIOs-MSCs (Fig. 2D). The results from RT-PCR sug-
showed intracytoplasmic iron inclusions as dense blue- gested that low concentration of SPIO labeling did not
stained vesicles, and the magnetic nanoparticles were affect MSC differentiation.
distributed evenly around the nucleus of the MSCs as
In Vitro Theoretical Analysis of the Magnetic Targeting
a spherical shell (Fig. 1C). The labeling efficiency was
approximately 100% reproducible using a very low con- The magnetic field lines around the magnet were
centration of iron oxide (50 µg/ml). Transmission elec- mapped by point-by-point magnetic field measurements
tron microscopy of labeled cells indicated the presence using a model 51662 Leybold Tesla meter. The center
of anionic magnetic nanoparticles exclusively in polydis- magnetic flux densities of three cylindrical magnet poles
perse vesicles in the cytoplasm, and no obvious change in were 0.15 T, 0.3 T, and 0.6 T, and the peripheral strengths
the ultramicrostructure was observed (Fig. 1D). of the corresponding magnetic poles were 0.40 T, 0.56 T,
and 0.93 T, respectively. As shown in Figure 3A–C, the
Characterization of SPIOs-MSCs magnetic field strength gradually declines with the short-
To determine the adipogenic and osteogenic differ- ened distance from periphery to center of the magnetic
entiation, the MSCs and SPIOs-MSCs were cultured for pole in the horizontal direction. Line 1 and line 2,

Figure 1. SPIOs and DiR labeling of rat MSCs. (A) The fourth passage of MSCs used in the present study. (B) MSCs were labeled
with DiR, and cell nuclei were counterstained with DAPI. (C) Prussian blue staining of MSCs labeled with SPIOs on cover glass.
(D) Transmission electron microscopy demonstrated the presence of numerous iron-containing vesicles in the cytoplasm of MSCs.
SPIOs, superparamagnetic oxide nanoparticles; DiR, 1,1-dioctadecyl-3,3,3,3-tetramethyl indotricarbocyanine iodide; MSCs, mesen-
chymal stem cells; DAPI, 4¢,6-diamidino-2-phenylindole.
1986 SHEN ET AL.

Figure 2. Differentiation capacity of SPIOs-MSCs. (A) Oil red O staining for adipogenic differentiation. (B) Alizarin red staining for
osteogenic differentiation. (C) Real-time PCR for myocardial gene GATA-4, cTnT, a-actin, and Cx43. (D) Quantitative gene expres-
sion analysis showed no difference between MSCs and SPIOs-MSCs, suggesting low concentration of SPIO labeling did not affect
MSCs differentiation. NC, negative control; PC, positive control.

respectively, represent magnetic field lines from the magnetic attenuation but also to make magnetic field
center and periphery of the magnetic pole in the vertical lines cover the target site for administering a specific
direction (Fig. 3D). According to the principle of mag- strength of magnetic targeting. We controlled the move-
netic attenuation, when the magnetic pole radius is 4 mm, ment ranges of magnets with the heartbeat to within
the density of magnetic flux is attenuated by 47%, 43%, 1 mm to maximally overcome magnetic attenuation. The
and 40% at 2 mm from the magnetic poles with 0.15 T, actual measured working strengths of the magnets with
0.3 T, and 0.6 T and is further decreased by 83%, 80%, 0.15 T, 0.3 T, and 0.6 T were at least 0.12 T, 0.23 T, and
and 76% at 6 mm from the three magnets, respectively 0.49 T, respectively. All magnets used here were 8 mm in
(Fig. 3E–G). Thus, it is critical not only to overcome the diameter and approximately the size of a MI in rats. We
MAGNETIC INTENSITY FOR STEM CELL TARGETING

Figure 3. Theoretical analysis of magnetic fields. (A–C) Distribution of magnetic field lines from three magnets. In the horizontal direction, the magnetic field strength gradually
declines along the radius from the periphery to the center. (D–G) Schematic diagram of magnetic attenuation in the vertical direction. Lines 1 and 2 represent magnetic decay curves
of central and peripheral of magnetisms, respectively, in the vertical direction.
1987
1988 SHEN ET AL.

managed to cover the infarct area and infarct border zone of magnetic fields only showed minimal accumulation
with the strongest area of the magnetic field, thus ensur- (0.22% ± 0.04%, 0.21% ± 0.02%, 0.21% ± 0.02% and
ing target homing of magnetic-responsive cells. 0.22% ± 0.03% for 0 T, 0.15 T, 0.3 T, and 0.6 T, respec-
tively; all p > 0.05 among groups), indicating that MSCs
Magnetic Capture of Flowing SPIOs-MSCs without SPIOs were not magnetically responsive.
The flowing SPIOs-MSCs were substantially attracted
to the site where the magnetic pole was positioned. Magnetic Strength Influences Cell Distribution
The flowing SPIOs-MSCs in the absence of an applied After Retention
magnetic field demonstrated minimal accumulation Because vascular-delivered cells reportedly translocate
(0.26% ± 0.04%), while the CEs reached 42.92% ± 4.76%, into the parenchyma after 48–72 h (29), a subpopula-
70.53% ± 6.53%, and 80.2% ± 4.79% when the magnetic tion of animals was histologically examined at 72 h. Rep-
intensities were 0.15 T, 0.3 T, and 0.6 T, respectively resentative hearts were cryosectioned and analyzed for
(all p < 0.001 among groups) (Fig. 4A–C). The capture covisualization of CD31+ blood vessels (green) and DiR-
efficiency of cells increased as the magnetic intensity labeled MSCs (red). Cells were found residing in the
enhanced, indicating that the flowing SPIOs-MSCs infarct area and peri-infarct border zone in the groups
can be significantly magnetically attracted in a magnetic treated with SPIOs-DiR-MSCs or DiR-MSCs. Not sur-
flux density-dependent manner. In contrast, the flow- prisingly, the numbers of DiR-MSCs were indistinguish-
ing unlabeled MSCs exposed to the varying intensities able in the 0 T and MSC groups (p > 0.05) (Fig. 5A, B).

Figure 4. Magnetic concentration of the flowing SPIOs-MSCs. (A) A schematic illustration of the microfluidic system. The SPIOs-
MSCs were injected into the tube, and the flow rate was controlled using a syringe pump. Cells were captured by use of three NdFeB
magnets located beside the tube. (B) Representative images of cell accumulation in the segment close to the magnet at velocities of
5 mm/s. (C) The efficiency of magnetically capturing flowing SPIOs-MSCs with different magnetic intensities. (#p < 0.001 vs. the 0 T
group; Dp < 0.001 vs. the 0.15 T group; ☆p < 0.01 vs. the 0.3 T group).
MAGNETIC INTENSITY FOR STEM CELL TARGETING 1989

Three magnetic targeting groups exhibited more SPIOs- has been reported previously (13,30). More importantly,
DiR-MSCs compared to either the 0 T group or the MSC the percentage of blocked vessels was 4.3% ± 0.98%,
group (all p < 0.001) (Fig. 5A–E). DiR fluorescence from 8.24% ± 1.12%, and 15.12% ± 2.09% when the magnetic
MSCs suggested that the 0.15 T, 0.3 T, and 0.6 T groups intensities were 0.15 T, 0.3 T, and 0.6 T, respectively,
demonstrated 2.18-, 3.95-, and 2.31-fold cell distribution, indicating that the external magnetic field may exacer-
respectively, outside vascular structures relative to the 0 T bate magnetically responsive MSCs obstructed in micro-
group (all p < 0.001, respectively) (Fig. 5F). Interestingly, vascular, and the microembolisms further worsened with
comparable results were found in the 0.6 T and 0.15 T the escalating magnetic intensity (Fig. 6C).
groups (p > 0.05).
Meanwhile, microemboli were detected in the groups The Relationship Between Magnetic Strength and
treated with MSCs (Fig. 6A, B). The percentage of Short-Term Cell Retention
blocked vessels was comparable in the MSC and 0 T Fluorescence imaging of hearts excised 24 h after cell
groups (2.93 ± 1.12% vs. 3.16 ± 1.36%, p > 0.05), indicat- infusion revealed more fluorescence signals in hearts
ing that MSCs can induce microvascular obstruction as from the three magnetic targeting groups than the 0 T

Figure 5. Microembolization at 72 h after intraventricular infusion (n = 6 for each group). (A–E) Representative fluorescence image
of heart sections from MSC group (A), 0 T group (B), 0.15 T group (C), 0.3 T group (D), and 0.6 T group (E). CD31 antibody (green)
stained for detecting blood vessels. MSCs were visualized by red fluorescence, and cell nuclei were counterstained with DAPI.
(F) MSC numbers from 10 randomly selected high-power fields (five from the infarct area and five from the border zone) in each group
were quantified by fluorescence microscopy. Data are mean ± SD. (#p < 0.001 vs. the MSC group or the 0 T group; Dp < 0.001 vs. the
0.15 T group; ☆p < 0.001 vs. the 0.3 T group). HPF, high power field.
1990 SHEN ET AL.

Figure 6. Microembolization at 72 h after intraventricular infusion (n = 6 for each group). (A) Representative heart frozen sections
were stained for CD31 antibody (green) to detect blood vessels. SPIOs-DiR-MSCs were visualized by red fluorescence, and cell nuclei
were counterstained with DAPI. The 0.6 T groups showed blood vessel completely blocked by cell clumps, whereas the 0 T, 0.15 T,
and 0.3 T groups exhibited that blood vessels containing cells were still patent. (B) Prussian blue staining confirmed that blood vessels
were blocked by cell clumps in the 0.3 T and 0.6 T groups (arrows), but most blood vessels containing cells were still patent in the 0 T
and 0.15 T groups (arrows). (C) Quantitative analysis of blocked vessels by frozen sections stained with CD31 antibody. (#p < 0.001
vs. the MSC group or the 0 T group; Dp < 0.001 vs. the 0.15 T group; ☆p < 0.001 vs. the 0.3 T group).

and MSC groups. The enhanced fluorescence from three three magnetically targeted groups decreased with the
magnetic targeting groups was dependent on magnetic enhancement of strength of heart magnetic targeting
strength. As a negative control, excised hearts from the (Fig. 7C).
PBS group were also imaged. No signals were detectable Quantitative PCR for the male-specific SRY gene
(Fig. 7A, B). was performed 24 h after cell infusion. Consistent with
To compare off-target migration, lungs, livers, spleens, the fluorescence imaging results, magnetic targeting
and kidneys from the same animals were also harvested enhanced short-term cell retention in a magnetic strength-
to detect fluorescence signals of transplanted MSCs. dependent manner: the 0.15 T, 0.3 T, and 0.6 T groups
Not surprisingly, fluorescence signals were detectable exhibited 1.45-, 2.76-, and 3.86-fold greater cell reten-
in these organs, but less so in the organs from three mag- tion rates, respectively, than the 0 T group (all p < 0.001)
netically targeted groups than in those from the MSC (Fig. 7D). The relationship between magnetic intensity
and 0 T groups (Fig. 7C). Moreover, there is a trend and retained cell numbers is a positive linear correlation
that fluorescence signals in the off-target organs from (r = 0.983, p = 0.017).
MAGNETIC INTENSITY FOR STEM CELL TARGETING 1991

Figure 7. Short-term cell retention and long-term cell engraftment in hearts and other organs. (A) Representative fluorescence imag-
ing of hearts harvested 24 h after cell injection. (B) Quantificative analysis of fluorescence intensities. (*p < 0.001 vs. the PBS group;
#p < 0.001 vs. the MSC group or the 0 T group; Dp < 0.001 vs. the 0.15 T group; ☆p < 0.001 vs. the 0.3 T group). (C) Cell retention in
other organs by fluorescence imaging 24 h after cell injection. (#p < 0.001 vs. the MSC group or the 0 T group; Dp < 0.001 vs. the 0.15 T
group; ☆p < 0.001 vs. the 0.3 T group). (D) Cell detection 24 h, 10 days, and 3 weeks after injection in the heart by quantitative PCR
of the male-specific SRY gene (n = 6 for each group).

Different Magnetic Strengths Induce Varying Long-Term cell engraftment rates in the 0.6 T group were lower than
Cell Engraftment the 0.3 T group (p < 0.001) (Fig. 7D). Compared with the
To examine the effects of magnetic targeting on long- 0.15 T group, the 0.6 T group exhibited comparable cell
term engraftment, quantitative PCR for the male-specific engraftment rates (p > 0.05) (Fig. 7D).
SRY gene was performed 10 days and 3 weeks after cell
infusion. Consistent with previous findings (4,6,19,20), Magnetic Strength Affects Therapeutic Benefits of Cell
quantitative PCR revealed that the five groups treated Transplantation and Left Ventricular Remodeling
with MSCs experienced a substantial drop in surviving To investigate functional outcomes mediated by mag-
cells 10 days after cell infusion. The surviving cells fur- netic targeting, LVEF was assessed by echocardiography
thered reduced 3 weeks after cell infusion; however, the at baseline (24 h after I/R and treatment) and for a sub-
three magnetic targeting groups still exhibited enhanced sequent 3 weeks. The 0.6 T group had the lowest LVEF
cell engraftment relative to the 0 T group (all p < 0.001) at baseline, but LVEF did not differ between treatment
(Fig. 7D), which is consistent with previous findings groups (Fig. 8A), indicating a comparable degree of ini-
(5,6), indicating that magnetic targeting can enhance tial injury. Over the next 3 weeks, LVEF declined progres-
MSC homing and engraftment. At 3 weeks after cell sively in the PBS group, but it did not decline in the four
infusion, the 0.15 T, 0.3 T, and 0.6 T groups exhibited MSC-treated groups. Both the 0.15 T and 0.3 T groups
1.88-, 3.06-, and 1.90-fold greater cell engraftment rates, exhibited better therapeutic outcome, with LVEF superior
respectively, than the 0 T group (Fig. 7D). Interestingly, to the 0 T group (all p < 0.001) (Fig. 8A) at 3 weeks. Not
1992 SHEN ET AL.

Figure 8. Functional benefits of cell transplantation. (A) LVEF measured by echocardiography 24 h and 3 weeks after cell injection
(n = 13 for 0.3 T and n = 12 for other groups). (B) Changes in LVEF from 24 h to 3 weeks in each group. (*p < 0.001 vs. the PBS group;
#p < 0.001 vs. the MSC group or the 0 T group; Dp < 0.001 vs. the 0.15 T group; ☆p < 0.001 vs. the 0.3 T group). (C–D) Changes in
LVEF are plotted against changes in LVIDs (C) and LVIDd (D). Change reflects the difference between parameters measured 24 h
and 3 weeks after therapy. LVEF, left ventricular ejection fraction; LVIDs, left ventricular internal end-systolic diameter; LVIDd, left
ventricular internal end-diastolic diameter; DLVEF, Changes in LVEF from 24 h to 3 weeks.

surprisingly, the 0.3 T group had a higher LVEF than the (Fig. 8D) percentage of change indicated that appropri-
0.15 T group (p < 0.001) (Fig. 8A). Notably, LVEF was ate intensity of magnetic targeting mediating cell therapy
comparable among the 0.6 T, 0 T, and MSC groups (all extended the therapeutic benefit of cell therapy for car-
p > 0.05) (Fig. 8A). To facilitate comparisons, we calcu- diac function and structure.
lated the treatment effect, that is, the change in LVEF at Morphometry at 3 weeks showed more infarct size in
3 weeks relative to baseline, in each group. the PBS group than the five MSC-treated groups. The
The PBS group had a negative treatment effect, with 0.15 T and 0.3 T groups showed significantly smaller
LVEF decreasing over time. In contrast, the 0.15 T and infarct size than the 0 T group and the MSC group. Fur-
0.3 T groups exhibited a sizable positive treatment effect thermore, the infarct size was 4.7% lower in the 0.3 T
that was greater than the 0 T and 0.6 T groups (Fig. 8B). group than the 0.15 T group (p < 0.001) (Fig. 9A, B).
The similar changes were also observed in LVIDd and Again, LV infarct size was indistinguishable among the
LVIDs. Compared with the MSC, 0 T, and 0.6 T groups, 0.6 T, 0 T, and MSC groups (p > 0.05) (Fig. 9A, B).
the 0.15 T group exhibited the greater reduction in LVIDd
and LVIDs (0.21 mm and 0.15 mm, 0.19 mm and 0.13 mm, Different Intensities of Magnetically Targeted Cell
0.21 mm and 0.14 mm vs. 0.59 mm and 0.40 mm, respec- Delivery Induce Varying Angiogenesis
tively, all p < 0.001). More importantly, the 0.3 T group To determine the mechanisms underlying the ben-
exhibited the greatest reduction in LVIDs (1.4 mm) and eficial effects of cell transplantation, we investigated the
LVIDd (0.90 mm) among three magnetic targeting groups effects of cell transplantation on angiogenesis in the post-
(all p < 0.001). The percentage of change in the LVEF at I/R hearts. At 3 weeks after cell transplantation, immu-
3 weeks, as a function of the LVIDs (Fig. 8C) or LVIDd nofluorescence staining for CD31 antibody indicated
MAGNETIC INTENSITY FOR STEM CELL TARGETING 1993

Figure 9. Left ventricular remodeling. (A) Representative Masson’s trichrome stained myocardial sections from a subgroup of ani-
mals 3 weeks after treatment (n = 7 for 0.3 T and n = 6 for other group). (B) Quantitative analysis of fibrosis size. (*p < 0.001 vs. the PBS
group; #p < 0.001 vs. the MSC group or the 0 T group; Dp < 0.01 vs. the 0.15 T group; ☆p < 0.001 vs. the 0.3 T group).

significant angiogenesis in MSC-treated hearts, with contributing to the observed cellular therapy-associated
more CD31-expressing capillaries being present in the beneficial effects.
peri-infarct region of MSC-treated groups compared with
the PBS group (all p < 0.05) (Fig. 10A, B). Quantitative DISCUSSION
evaluation of CD31-positive capillary numbers indicated To our knowledge, this is the first study that compares
that the PBS group exhibited the lowest capillary den- the impact of different strengths of magnetic targeting
sity. In contrast, both the 0.15 T and 0.3 T groups had on the therapeutic outcomes of intraventricular deliv-
more capillary density than the 0 T and 0.6 T groups (all ery of MSCs in a rat model of ischemia/reperfusion. We
p < 0.01) (Fig. 10A, B). Not surprisingly, the 0.3 T group found that magnetic targeting enhanced cell retention in
had the most capillary density. Also, no significant differ- a magnetic intensity-dependent manner, but “too high”
ence was found among the 0.6 T, 0 T, and MSC groups of a strength can worsen embolic injury, thus undermin-
(p > 0.05) (Fig. 10B). However, few cells stained positive ing the therapeutic effects of cell transplantation. A num-
for both the endothelial cell marker CD31 or the cardio- ber of animal studies and clinical trials have confirmed
myocyte marker TnT and DiR in the five MSC-treated that transplantation effect was positively related to cell
groups. These data suggest that transplanted MSCs dose. Pouzet et al. reported that a fivefold difference
may promote angiogenesis through a paracrine effect, in the number of transplanted skeletal myoblasts (SM,
1994 SHEN ET AL.

Figure 10. Angiogenesis 3 weeks after cell infusion. (A) Frozen sections of representative hearts were stained for CD31 antibody
(green) to detect capillary densities in the peri-infarct region 3 weeks after MSC transplantation. (B) Quantitative analysis of CD31-
containing capillaries in the peri-infarct region (n = 7 for 0.3 T and n = 6 for other group). (*p < 0.05 vs. the PBS group; #p < 0.05 vs. the
MSC group or the 0-T group; Dp < 0.01 vs. the 0.15-T group; ☆p < 0.001 vs. the 0.3-T group).

1 ´ 106 vs. 5 ´ 106) caused a twofold difference in LVEF delivery routes and different heterogeneity evaluations,
(22). Tambara et al. found that a large number of trans- these results demonstrated that, in patients with AMI, the
planted neonatal SM can replace the infarcted myocar- effects of cell transplantation are dependent on the num-
dium with reverse LV remodeling in infracted rat hearts ber of cells. This finding is the theoretical basis of cell
(27). Clinical trials (17,23,32) and meta analysis (7) also transplantation guided by magnetic targeting. Previous
demonstrated that heart function in patients with AMI studies have found that magnetic targeting can enhance
was related to intracoronary transplanted cell dose. Our cell homing and improve therapeutic effects irrespective
previous study found that repeated bone marrow mono- of delivery route (5,6).
nuclear cell administration can significantly improve LV We found that the 0.15 T, 0.3 T, and 0.6 T groups
function, compared with a single infusion in patients exhibited 1.45-, 2.76-, and 3.86-fold greater cell retention
with large AMI (35). Despite different cell types and rates, respectively, than the 0 T group. Cheng et al. (6)
MAGNETIC INTENSITY FOR STEM CELL TARGETING 1995

reported the magnet group exhibited more than fourfold of MSCs is related to cell number (13). Cheng et al. (6)
greater cell retention rates than the cardiosphere-derived found that the intracoronary delivery of 1.0 × 106 CDCs
cell (CDC) group. Chaudeurge et al. (4) implanted a 0.1 T with or without magnetic targeting may result in microem-
magnet (7 mm diameter, 5 mm thickness) subcutane- bolization. Nakamae et al. (20) have found that an external
ously, leading to cell retention between groups failing to magnetic force can enhance adhesion of MSCs, which may
be statistically significant using RT-PCR quantification. partly account for microembolization. How, then, can mag-
We speculate that the different results may be attributed netically targeted intracoronary cell delivery resulting in
to the difference in magnetic intensities and cell types. microvascular obstruction be improved? We speculate that
To our knowledge, there are several factors affecting the smaller cells may have the advantage of reducing embo-
magnetic attraction of transplanted cells. First, the mag- lization. For example, very small embryonic-like stem
netic strength and the magnetic exposure time must be cells (~2–4 µm) found in recent years have been proven to
taken into consideration. Notably, the magnet intensity is attenuate LV remodeling and improve LV systolic function
not equal to the actual working strength, which is limited in small animal studies (8,33). In addition, previous study
by magnetic attenuation. To overcome magnetic attenua- found that sodium nitroprusside could influence the distri-
tion and obtain maximal working magnetic strength, the bution of MSCs by reducing the number of MSCs trapped
distance from the magnetic pole to the target site should in the lungs (12). Vasodilator applications might represent
be as short as possible. Second, it is critical that the dis- another promising method to reduce embolism.
tribution of magnetic field lines must cover the target The magnetic targeting is an emerging and promising
site. Third, the iron content and the magnetic responsive- strategy in stem cell therapeutics. Before translation into
ness of the transplanted cells may also be an important possible clinical application, it is warranted to elucidate
factor. In the present study, the distance from magnet to the optimized magnetic strength, exposure time, cell type,
target lesion was 0~1 mm to maximally overcome mag- cell number, and administration route. Because the infarct
netic attenuation; moreover, although the three magnets size is larger and the ventricular wall is thicker in human
we used had different strengths, their surface areas were beings or large animals than those in rats, we speculate
close to the size of a rat MI, ensuring the magnetic target- that stronger magnetic strength is needed, and the vascu-
ing effect. lar embolism may become a more serious complication.
Surprisingly, we found the 0.6 T group had the highest
cell retention, but at 3 weeks cell engraftment was indis- Study Limitations
tinguishable relative to the 0.15 T group; moreover, thera- Although it describes a new and promising method, the
peutic benefits had no significant difference compared to present study has a number of limitations. The optimized
the 0 T group. What led to the paradoxical phenomenon magnet strength obtained from the small animal stud-
between cell retention and therapeutic effects? We found ies may not be directly extrapolatable to large animals.
more microembolization in the 0.6 T group at 72 h than Additionally, we did not include a magnet targeting group
the 0.15 T and 0.3 T groups. We speculate that the para- without cell infusion and hence did not explore the effects
doxical result may be associated with severer microem- of magnet targeting on a rat model of acute ischemia/
bolization (15). Owing to more serious microembolisms, reperfusion. Furthermore, instead of optimizing basal cell
although the 0.6 T group had the highest cell retention, the dosage and magnetic exposure time, we chose cell dose
retained cells did not represent effectively homing cells. and duration consistent with previous studies that showed
Consequently, the result did not translate into enhanced functional benefits in small animals (5,6,9,18,19,25).
cell engraftment. Moreover, these microembolisms may Further dosage and exposure time optimization would be
cause new myocardial injury or deteriorate original injury, valuable, to inform future clinical studies. Finally, we did
partly offsetting the therapeutic benefit of homing cells not investigate whether the SPIO movement guided by
and eventually resulting in comparable functional effects the magnetic field damaged the MSCs’ micro- and ultra-
in the 0.6 T and 0 T groups. Basically, the cell transplanta- structures. Future studies will be designed to capture any
tion effect is dependent on the effective homing cell num- changes caused by magnetic targeting in transplanted cell
ber rather than retained cell number. micro- and ultrastructures.
The intracoronary injection of MSCs has been proposed
as a potential therapeutic option to repair an ischemic- or CONCLUSION
infarct-damaged heart. However, because MSCs are large We conclude that cell retention can be continuously
(rounded up cells are ~22 to 25 µm in diameter), an important increased with the enhancement of magnetic targeting
consideration of this approach is the potential of these cells intensity. Magnetic targeting can enhance cell engraft-
to induce myocardial damage via microvascular obstruc- ment and improve therapeutic benefits through increasing
tion, as previously reported (30). In addition, microvascu- the effectively homing MSCs in a magnetic intensity-
lar embolism caused by the intracoronary administration dependent manner. However, when the magnetic field
1996 SHEN ET AL.

strength is elevated to a certain extent, microemboliza- Li, W.; Gaebel, R.; Li, R. K.; Vollmar, B.; Steinhoff, G.;
tion is increased, thus reducing long-term cell engraft- Ma, N. Is the intravascular administration of mesenchymal
stem cells safe? Mesenchymal stem cells and intravital
ment and further undermining therapeutic benefit.
microscopy. Microvasc. Res. 77:370–376; 2009.
ACKNOWLEDGMENTS: The authors gratefully acknowl- 12. Gao, J.; Dennis, J. E.; Muzic, R. F.; Lundberg, M.;
edge the financial support from the National Natural Science Caplan, A. I. The dynamic in vivo distribution of bone
Foundation of China (81370003, 81000043, 11304194, and marrow-derived mesenchymal stem cells after infusion.
81100145) and the Science and Technology Commission of Cells Tissues Organs 169:12–20; 2001.
Shanghai Municipality (13JC1401703). The authors declare no 13. Grieve, S. M.; Bhindi, R.; Seow, J.; Doyle, A.; Turner,
conflicts of interest. A. J.; Tomka, J.; Lay, W.; Gill, A.; Hunyor, S. N.; Figtree,
G. A. Microvascular obstruction by intracoronary delivery
REFERENCES of mesenchymal stem cells and quantification of resulting
1. Bayne, K. Revised Guide for the Care and Use of myocardial infarction by cardiac magnetic resonance. Circ.
Laboratory Animals available. American Physiological Heart Fail. 3:e5–e6; 2010.
Society. Physiologist 39:199, 208–211; 1996. 14. Huang, Z.; Pei, N.; Wang, Y.; Xie, X.; Sun, A.; Shen, L.;
2. Boopathy, A. V.; Pendergrass, K. D.; Che, P. L.; Yoon, Y. S.; Zhang, S.; Liu, X.; Zou, Y.; Qian, J.; Ge, J. Deep magnetic
Davis, M. E. Oxidative stress-induced Notch1 signaling pro- capture of magnetically loaded cells for spatially targeted
motes cardiogenic gene expression in mesenchymal stem therapeutics. Biomaterials 31:2130–2140; 2010.
cells. Stem Cell Res. Ther. 4:43; 2013. 15. Huang, Z.; Shen, Y.; Pei, N.; Sun, A.; Xu, J.; Song, Y.;
3. Castro, E.; Mano, J. F. Magnetic force-based tissue engi- Huang, G.; Sun, X.; Zhang, S.; Qin, Q.; Zhu, H.; Yang, S.;
neering and regenerative medicine. J. Biomed. Nanotechnol. Yang, X.; Zou, Y.; Qian, J.; Ge, J. The effect of nonuniform
9:1129–1136; 2013. magnetic targeting of intracoronary-delivering mesenchy-
4. Chaudeurge, A.; Wilhelm, C.; Chen-Tournoux, A.; Farahmand, mal stem cells on coronary embolisation. Biomaterials
P.; Bellamy, V.; Autret, G.; Menager, C.; Hagege, A.; 34:9905–9916; 2013.
Larghero, J.; Gazeau, F.; Clement, O.; Menasche, P. Can 16. Huang, Z.; Shen, Y.; Sun, A.; Huang, G.; Zhu, H.; Huang, B.;
magnetic targeting of magnetically labeled circulating cells Xu, J.; Song, Y.; Pei, N.; Ma, J.; Yang, X.; Zou, Y.; Qian, J.;
optimize intramyocardial cell retention? Cell Transplant. Ge, J. Magnetic targeting enhances retrograde cell reten-
21:679–691; 2012. tion in a rat model of myocardial infarction. Stem Cell Res.
5. Cheng, K.; Li, T. S.; Malliaras, K.; Davis, D. R.; Zhang, Ther. 4:149; 2013.
Y.; Marban, E. Magnetic targeting enhances engraftment 17. Meluzin, J.; Mayer, J.; Groch, L.; Janousek, S.; Hornacek, I.;
and functional benefit of iron-labeled cardiosphere-derived Hlinomaz, O.; Kala, P.; Panovsky, R.; Prasek, J.; Kaminek, M.;
cells in myocardial infarction. Circ. Res. 106:1570–1581; Stanicek, J.; Klabusay, M.; Koristek, Z.; Navratil, M.; Dusek,
2010. L.; Vinklarkova, J. Autologous transplantation of mononuclear
6. Cheng, K.; Malliaras, K.; Li, T. S.; Sun, B.; Houde, C.; bone marrow cells in patients with acute myocardial infarc-
Galang, G.; Smith, J.; Matsushita, N.; Marban, E. Magnetic tion: The effect of the dose of transplanted cells on myocardial
enhancement of cell retention, engraftment, and functional function. Am. Heart J. 152:975.e9–975.e15; 2006.
benefit after intracoronary delivery of cardiac-derived stem 18. Molina, E. J.; Palma, J.; Gupta, D.; Gaughan, J. P.; Houser,
cells in a rat model of ischemia/reperfusion. Cell Transplant. S.; Macha, M. Right ventricular effects of intracoronary
21:1121–1135; 2012. delivery of mesenchymal stem cells (MSC) in an ani-
7. Clifford, D. M.; Fisher, S. A.; Brunskill, S. J.; Doree, C.; mal model of pressure overload heart failure. Biomed.
Mathur, A.; Clarke, M. J.; Watt, S. M.; Martin-Rendon, E. Pharmacother. 63:767–772; 2009.
Long-term effects of autologous bone marrow stem cell 19. Molina, E. J.; Palma, J.; Gupta, D.; Torres, D.; Gaughan,
treatment in acute myocardial infarction: Factors that may J. P.; Houser, S.; Macha, M. Improvement in hemodynamic
influence outcomes. PLoS One 7:e37373; 2012. performance, exercise capacity, inflammatory profile, and
8. Dawn, B.; Tiwari, S.; Kucia, M. J.; Zuba-Surma, E. K.; left ventricular reverse remodeling after intracoronary deliv-
Guo, Y.; Sanganalmath, S. K.; Abdel-Latif, A.; Hunt, G.; ery of mesenchymal stem cells in an experimental model
Vincent, R. J.; Taher, H.; Reed, N. J.; Ratajczak, M. Z.; of pressure overload hypertrophy. J. Thorac. Cardiovasc.
Bolli, R. Transplantation of bone marrow-derived very Surg. 135:292–299; 2008.
small embryonic-like stem cells attenuates left ventricular 20. Nakamae, T.; Adachi, N.; Kobayashi, T.; Nagata, Y.;
dysfunction and remodeling after myocardial infarction. Nakasa, T.; Tanaka, N.; Ochi, M. The effect of an exter-
Stem Cells 26:1646–1655; 2008. nal magnetic force on cell adhesion and proliferation of
9. Erwin, G. S.; Crisostomo, P. R.; Wang, Y.; Wang, M.; magnetically labeled mesenchymal stem cells. Sports Med.
Markel, T. A.; Guzman, M.; Sando, I. C.; Sharma, R.; Arthrosc. Rehabil. Ther. Technol. 2:5; 2010.
Meldrum, D. R. Estradiol-treated mesenchymal stem cells 21. Pfeffer, M. A.; Pfeffer, J. M.; Fishbein, M. C.; Fletcher,
improve myocardial recovery after ischemia. J. Surg. Res. P. J.; Spadaro, J.; Kloner, R. A.; Braunwald, E. Myocardial
152:319–324; 2009. infarct size and ventricular function in rats. Circ. Res.
10. Fukushima, S.; Varela-Carver, A.; Coppen, S. R.; Yamahara, 44:503–512; 1979.
K.; Felkin, L. E.; Lee, J.; Barton, P. J.; Terracciano, C. M.; 22. Pouzet, B.; Vilquin, J. T.; Hagege, A. A.; Scorsin, M.;
Yacoub, M. H.; Suzuki, K. Direct intramyocardial but Messas, E.; Fiszman, M.; Schwartz, K.; Menasche, P.
not intracoronary injection of bone marrow cells induces Factors affecting functional outcome after autologous skel-
ventricular arrhythmias in a rat chronic ischemic heart fail- etal myoblast transplantation. Ann. Thorac. Surg. 71:844–
ure model. Circulation 115:2254–2261; 2007. 850; discussion 850–851; 2001.
11. Furlani, D.; Ugurlucan, M.; Ong, L.; Bieback, K.; 23. Quyyumi, A. A.; Waller, E. K.; Murrow, J.; Esteves, F.; Galt,
Pittermann, E.; Westien, I.; Wang, W.; Yerebakan, C.; J.; Oshinski, J.; Lerakis, S.; Sher, S.; Vaughan, D.; Perin, E.;
MAGNETIC INTENSITY FOR STEM CELL TARGETING 1997

Willerson, J.; Kereiakes, D.; Gersh, B. J.; Gregory, D.; 31. Wang, X.; Jameel, M. N.; Li, Q.; Mansoor, A.; Qiang, X.;
Werner, A.; Moss, T.; Chan, W. S.; Preti, R.; Pecora, A. L. Swingen, C.; Panetta, C.; Zhang, J. Stem cells for myocar-
CD34(+) cell infusion after ST elevation myocardial infarc- dial repair with use of a transarterial catheter. Circulation
tion is associated with improved perfusion and is dose 120:S238–S246; 2009.
dependent. Am. Heart J. 161:98–105; 2011. 32. Wohrle, J.; von Scheidt, F.; Schauwecker, P.; Wiesneth, M.;
24. Robey, T. E.; Saiget, M. K.; Reinecke, H.; Murry, C. E. Markovic, S.; Schrezenmeier, H.; Hombach, V.; Rottbauer,
Systems approaches to preventing transplanted cell death W.; Bernhardt, P. Impact of cell number and microvascular
in cardiac repair. J. Mol. Cell. Cardiol. 45:567–581; 2008. obstruction in patients with bone-marrow derived cell ther-
25. Saito, T.; Kuang, J. Q.; Lin, C. C.; Chiu, R. C. Transcoronary apy: Final results from the randomized, double-blind, pla-
implantation of bone marrow stromal cells ameliorates cebo controlled intracoronary stem cell therapy in patients
cardiac function after myocardial infarction. J. Thorac. with acute myocardial infarction (SCAMI) trial. Clin. Res.
Cardiovasc. Surg. 126:114–123; 2003. Cardiol. 102:765–770; 2013.
26. Sanganalmath, S. K.; Bolli, R. Cell therapy for heart fail- 33. Wu, J. H.; Wang, H. J.; Tan, Y. Z.; Li, Z. H. Characterization
ure: A comprehensive overview of experimental and clini- of rat very small embryonic-like stem cells and cardiac
cal studies, current challenges, and future directions. Circ. repair after cell transplantation for myocardial infarction.
Res. 113:810–834; 2013. Stem Cells Dev. 21:1367–1379; 2012.
27. Tambara, K.; Sakakibara, Y.; Sakaguchi, G.; Lu, F.; 34. Xu, J.; Qian, J.; Xie, X.; Lin, L.; Zou, Y.; Fu, M.; Huang, Z.;
Premaratne, G. U.; Lin, X.; Nishimura, K.; Komeda, M. Zhang, G.; Su, Y.; Ge, J. High density lipoprotein protects
Transplanted skeletal myoblasts can fully replace the mesenchymal stem cells from oxidative stress-induced
infarcted myocardium when they survive in the host in large apoptosis via activation of the PI3K/Akt pathway and
numbers. Circulation 108(Suppl 1):II259–II263; 2003. suppression of reactive oxygen species. Int. J. Mol. Sci.
28. Terrovitis, J. V.; Smith, R. R.; Marban, E. Assessment and 13:17104–17120; 2012.
optimization of cell engraftment after transplantation into 35. Yao, K.; Huang, R.; Sun, A.; Qian, J.; Liu, X.; Ge, L.;
the heart. Circ. Res. 106:479–494; 2010. Zhang, Y.; Zhang, S.; Niu, Y.; Wang, Q.; Zou, Y.; Ge, J.
29. Toma, C.; Wagner, W. R.; Bowry, S.; Schwartz, A.; Repeated autologous bone marrow mononuclear cell ther-
Villanueva, F. Fate of culture-expanded mesenchymal stem apy in patients with large myocardial infarction. Eur. J.
cells in the microvasculature: In vivo observations of cell Heart Fail. 11:691–698; 2009.
kinetics. Circ. Res. 104:398–402; 2009. 36. Zhang, C.; Liu, T.; Su, Y.; Luo, S.; Zhu, Y.; Tan, X.; Fan,
30. Vulliet, P. R.; Greeley, M.; Halloran, S. M.; MacDonald, S.; Zhang, L.; Zhou, Y.; Cheng, T.; Shi, C. A near-infrared
K. A.; Kittleson, M. D. Intra-coronary arterial injection of fluorescent heptamethine indocyanine dye with preferen-
mesenchymal stromal cells and microinfarction in dogs. tial tumor accumulation for in vivo imaging. Biomaterials
Lancet 363:783–784; 2004. 31:6612–6617; 2010.

Вам также может понравиться