Almeida and Wu JC 2011

Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Am J Physiol Heart Circ Physiol 301: H663–H671, 2011.

First published June 10, 2011; doi:10.1152/ajpheart.00337.2011. Review

In vivo bioluminescence for tracking cell fate and function


Patricia E. de Almeida,1,2 Juliaan R. M. van Rappard,4 and Joseph C. Wu1,2,3
Departments of 1Medicine and 2Radiology and 3Institute for Stem Cell Biology and Regenerative Medicine, Stanford
University School of Medicine, Stanford, California; and 4Leiden University School of Medicine, Leiden, The Netherlands
Submitted 4 April 2011; accepted in final form 1 June 2011

de Almeida PE, van Rappard JR, Wu JC. In vivo bioluminescence for


tracking cell fate and function. Am J Physiol Heart Circ Physiol 301: H663–H671,
2011. First published June 10, 2011; doi:10.1152/ajpheart.00337.2011.—Tracking
the fate and function of cells in vivo is paramount for the development of rational
therapies for cardiac injury. Bioluminescence imaging (BLI) provides a means for
monitoring physiological processes in real time, ranging from cell survival to gene
expression to complex molecular processes. In mice and rats, BLI provides
unmatched sensitivity because of the absence of endogenous luciferase expression
in mammalian cells and the low background luminescence emanating from animals.
In the field of stem cell therapy, BLI provides an unprecedented means to monitor
the biology of these cells in vivo, giving researchers a greater understanding of their
survival, migration, immunogenicity, and potential tumorigenicity in a living
animal. In addition to longitudinal monitoring of cell survival, BLI is a useful tool
for semiquantitative measurements of gene expression in vivo, allowing a better
optimization of drug and gene therapies. Overall, this technology not only enables
rapid, reproducible, and quantitative monitoring of physiological processes in vivo
but also can measure the influences of therapeutic interventions on the outcome of
cardiac injuries.
bioluminescence imaging; heart; gene therapy; stem cell therapy; in vivo cell
tracking

THIS ARTICLE is part of a collection on Assessing Cardiovas- Histopathological examination has been the main approach
cular Function in Mice: New Developments and Methods. for ex vivo evaluation of the distribution, the engraftment, and
Other articles appearing in this collection, as well as a full the differentiation of injected cells. However, histopathology
archive of all collections, can be found online at http://ajpheart. precludes the evaluation and monitoring of these parameters in
physiology.org/. real time and in vivo. Molecular and cellular imaging has
Cell-based therapies have rapidly emerged as a potential provided various techniques for identifying and tracking trans-
therapeutic approach for heart disease. After the initial work to planted cells in cardiovascular research (15). Magnetic reso-
characterize putative endothelial progenitor cells (1) and their nance imaging (MRI), computed tomography (CT), positron
potential to promote cardiac neovascularization and to attenu- emission tomography (PET), and single photon emission com-
ate ischemic injury, a decade of intense research has examined puted tomography (SPECT) offer deep tissue penetration and
several novel approaches to promote cardiac repair in adult high spatial resolution (64, 70, 94a). However, in small animal
life. A variety of adult stem and progenitor cells from different studies, these techniques are more costly and time consuming
sources have been examined for their potential to promote to implement compared with optical imaging. Among the
cardiac repair and regeneration: bone marrow-derived cells optical imaging tools, bioluminescence imaging (BLI) is a
(55, 72), circulating and mobilized CD133⫹ and CD34⫹ stem promising technique that is especially useful in small animal
and progenitor cells (36), mesenchymal stem cells (56, 65, 83), models and does not require the use of radionuclides with their
cardiac resident stem cells (43, 62), and skeletal myoblasts (54, associated hazards. BLI is a high throughput technique that can
97). However, many questions such as the optimal type and provide unmatched sensitivity because of the absence of en-
dogenous luciferase expression in mammalian cells and the
number of progenitor cells to be administered, the route of
low background luminescence emanating from animals. BLI
administration, and the best time to administer cells after injury
can be very useful in evaluating the delivery efficiency of
remain unresolved. This is particularly true in vivo where it is
therapeutic genes and their expression levels in vivo. By the
difficult to assess cellular activity in the heart in real time.
use of different cellular promoters to drive the expression of a
Therefore, it would be helpful to have practical tools to luminescent reporter gene, BLI allows monitoring of the trans-
accurately track cell location and their functional status over planted cells’ differentiation status as well as their location and
time in vivo. functional characteristics in vivo (39, 51, 72, 78, 94).
Overall, BLI of reporters cloned into promoter/enhancer
Address for reprint requests and other correspondence: J. C. Wu, Stanford
sequences or engineered into fusion proteins has demonstrated
Univ. School of Medicine, Lokey Stem Cell Research Bldg., 265 Campus Dr., the modality’s ability to monitor fundamental processes such
Rm. G1120B, Stanford, CA, 94305-5454 (e-mail: [email protected]). as transcriptional regulation, signal transduction cascades, pro-
http://www.ajpheart.org 0363-6135/11 Copyright © 2011 the American Physiological Society H663
Review
H664 IN VIVO BIOLUMINESCENCE FOR TRACKING CELL FATE AND FUNCTION

Fig. 1. Diagram illustrating steps required for biolumines-


cence imaging (BLI). A: therapeutic cells are transduced
with a luciferase (Luc) reporter construct and injected into
an animal. B: alternatively, luciferase-expressing cells can
be isolated from a luciferase transgenic animal and injected
to the experimental subject. C: after systemic substrate
injection, a charge-coupled device camera can be used to
localize the luciferase photon signals in vivo. Pseudo-col-
ored images that represent signal intensity are overlaid with
grayscale reference images of the animal to facilitate local-
ization of the signal. Cell survival, expansion, and homing,
as well as the expression of therapeutic genes, can be
monitored using this strategy.

tein-protein interactions, protein degradation, oncogenic trans- BLI is based on the detection of light emitted by cells that
formation, cell trafficking, and targeted drug action under in express light-generating enzymes such as luciferase. In biolu-
vivo spatial registration. In this review, we will discuss recent minescent reactions, luciferase generates visible light through
advances and applications of BLI, specifically as they apply to the oxidation of enzyme-specific substrates such as D-luciferin
cardiovascular research. for terrestrial organisms (29, 30) and coelenterazine for marine
organisms (31, 53). Luciferases from different organisms can
Principles of Bioluminescence be distinguished by their abbreviations: lux (bacterial), luc
In nature, numerous luminous species exist in more than 700 (firefly), and lcf (dinoflagellate). To track cells in vivo by BLI,
genera, of which 80% are marine species (93). Luciferase the cells of interest need to be genetically modified to express
enzymes have been cloned from both marine (e.g., Renilla luciferase. The animal recipient of the cells receives the lu-
luciferase) and terrestrial (e.g., firefly and click beetle lu- ciferase substrate either intraperitoneally or intravenously and
ciferase) eukaryotic organisms and are commonly used as is placed in a light-tight dark box where luminescence is
reporters for in vitro and in vivo studies. They emit long detected (Fig. 1). In a bioluminescent reaction, the generation
wavelengths of bioluminescence (⬎600 nm) in the red and of light depends on several factors. Firefly luciferase requires
near-infrared regions of the spectrum and are efficiently trans- ATP, Mg2⫹, and oxygen to catalyze the oxidation of its
mitted through mammalian tissues (20, 86). These wavelengths substrate D-luciferin and generates CO2, AMP, inorganic py-
can avoid absorbing and scattering environment of mammalian rophosphate, oxyluciferin, and a yellow-green light at a wave-
tissues (69), thus can be efficiently detected outside a small length that peaks at 562 nm (Fig. 2). By comparison, Renilla
animal’s body using BLI. luciferase catalyzes the oxidative decarboxylation of coelen-

Fig. 2. Representation of the bioluminescence reaction.


A cell expressing the firefly luciferase reporter gene
produces the luciferase enzyme that in the presence of
oxygen, ATP, and Mg2⫹ catalyzes D-luciferin into oxy-
luciferin, CO2, inorganic pyrophosphate (PPi), and light.
Light can then be detected, collected, and quantified by
a charge-coupled device camera.

AJP-Heart Circ Physiol • VOL 301 • SEPTEMBER 2011 • www.ajpheart.org


Review
IN VIVO BIOLUMINESCENCE FOR TRACKING CELL FATE AND FUNCTION H665
terazine in the presence of dissolved oxygen to yield oxylu- emitted. A mutation of a single amino acid has been shown to
ciferin, CO2, and blue light that peaks at 480 nm (49). Biolu- cause red-shifted luminescence and improve in vivo perfor-
minescence generated by the luciferase reporter reaction is mance. Recently, a red-shifted mutant of luciferase from Pho-
captured by a cooled charge-coupled device camera that can tinus pyralis was created and described to have an emission
detect very low levels of visible light emitted from internal maximum of 612 nm at pH 7.0, a narrow emission bandwidth,
organs (19). Charge-coupled device camera-imaged biolumi- and to be thermostable (with a half-life 8.8 h at 37°C vs. 0.26
nescence can then be superimposed on photographic images of h for the wild-type luciferase). This Photinus pyrallis mutant,
the mouse to detect quantitatively and repetitively the biolu- called Ppy RE-TS reporter, has been successfully used in small
minescent signal from a given location (95). Imaging can be animals to visualize cancer progr ession and shown to have
conducted 10 to 15 min after intraperitoneal injection of superior in vivo imaging performance compared with the
D-luciferin (reporter probe), with relatively stable light emis- wild-type photinus pyralis luciferase (6, 7).
sion levels for 30 to 60 min, depending on the experimental The choice of reporter gene reporter probe pair to be used
conditions. The sensitivity of detection depends on the wave- should ultimately be based on the specific biological process to
length of light emission, expression levels of the enzyme in the be monitored, the duration and intensity of the signal needed,
target cells, the location of the source of bioluminescence in and the tissue to be imaged (21, 100) (Fig. 3). As longer
the animal, the efficiency of the collection optics, and the wavelengths of light penetrate mammalian tissues with less
sensitivity of the detector (95). absorbance, luciferase from Photinus pyralis (⬎600 nm) is
Reporter genes commonly used for BLI. To track cells in more readily detected (17). Moreover, its substrate D-luciferin
vivo, reporter genes and reporter probes must be able to reveal remains in circulation longer than other substrates because it is
cellular and molecular processes throughout an entire study poorly catalyzed by mammalian tissues (101). However, Pho-
period, be highly sensitive to small changes in cell function and tinus pyralis luciferase has the disadvantage of having a longer
distribution over time, and must not alter the labeled biological coding sequence (1,653 bp) compared with marine luciferases
process itself. A variety of reporter genes have been introduced such as Renilla and Gaussia. Renilla and Gaussia luciferases
and validated for different imaging modalities, including var- have coding sequences of 936 and 558 bp, respectively, mak-
ious luciferases for BLI (5, 67, 74). Luciferase (Luc) is the only ing them more suitable for studies that require compact trans-
one that produces light without requiring an external excitation gene sequences, such as gene transfer and gene expression
source, and they offer inherently low background signals studies. Another advantage of Renilla and Gaussia luciferases
because animal tissues do not emit significant amounts of light.
over Photinus pyralis is the fact that they do not require ATP
Since the first report of the cDNA encoding Luc in 1985 (23),
as a cofactor during bioluminescent reaction and thus can be
DNA sequences have been optimized so that the Luc gene can
used for imaging cells independent of their metabolic state (5,
be expressed at high levels and its product localized in the
66). One particular characteristic of Gaussia luciferase is that
cytoplasm of the cells. For all reporter systems, the intensity of
it is naturally secreted (96) and therefore can be used as a
light is proportional to the amount of luciferase expressed in
each individual cell or the number of cells in which a gene has reporter for quantitative assessment of cells in vivo by mea-
been transferred. suring its concentration in blood (61). However, Renilla and
Of the many available luciferase enzymes, only a subset Gaussia luciferases produce shorter wavelengths of light
have been developed and used as reporter genes. Luciferase (peaking at 480 nm), which are not transmitted through tissues
from the North American firefly Photinus pyralis is the most as effectively as Photinus pyralis (100). Overall, the utility of
common choice, but other luciferases such as the sea pansy Renilla and Gaussia for in vivo BLI can benefit from improve-
Renilla reniformis, the click beetle Pyrophorus plagiophala- ments in sensitivity (46 – 48).
mus, and the copepod Gaussia princeps have also been inves- Codon-optimized humanized Gaussia (2, 79) has improved
tigated (48, 79). Gaussia and Renilla luciferase enzymes emit its sensitivity in mammalian cells, but the pharmacokinetics of
in the blue/green region of the UV visible spectrum, where its reporter probe coelenterazine are somewhat limiting in vivo.
light is strongly absorbed and scattered by tissues. Conse- Coelenterazine is prone to quick inactivation, including degra-
quently, the imaging performance suffers from poor sensitivity dation through autoxidation. This substrate is also costly with
and spatial resolution. On the contrary, Photinus pyralis and low solubility. Furthermore, coelenterazine binds to serum
click beetle luciferase emit ⬃60% of their light at ⬎600 nm, proteins, is cleared rapidly from the bloodstream (101), and
which enables great tissue penetration. Photinus pyralis has decays rapidly with time (5). Therefore, when imaging in vivo
emission at 620 nm when collected at 37°C, making it among with coelenterazine, the signal needs to be acquired immedi-
the longest emitting luciferases at mammalian body tempera- ately after substrate administration. On the other hand, this
tures and the most sensitive for in vivo applications (100). short half-life can be advantageous in cases in which sequential
Bacterial luciferases (Lux) such as from Photorhabdus lu- imaging of two luciferases, such as Gaussia princeps and
minescens emit blue light that has been used for BLI of Photinus pyralis, is required to monitor two biological pro-
bacterial infections. They are unique in that their lux operon cesses in tandem in the same animal (5).
cassette codes for the luciferase and also for enzymes that In summary, the development of reporter gene variants with
produce the substrate required for luminescence reaction, better emission spectra, brightness, and stability can ultimately
thereby eliminating the need for exogenous substrate. Trans- improve sensitivity and the overall performance of luciferases
ferring this operon to mammalian cells would be advantageous in BLI imaging studies. Moreover, cloning new luciferases
for in vivo imaging; however, limited research has been done from different organisms, such as those from Luciola italica
to determine whether this is possible (16). Mutations in lu- and Cratomorphus distinctus, can certainly improve current
ciferases can change the wavelength of the luminescence applications and make novel ones possible (8, 88).

AJP-Heart Circ Physiol • VOL 301 • SEPTEMBER 2011 • www.ajpheart.org


Review
H666 IN VIVO BIOLUMINESCENCE FOR TRACKING CELL FATE AND FUNCTION

Fig. 3. Emission wavelengths for the most commonly used reporter luciferase enzymes in BLI and their advantages and disadvantages. V, violet; B, blue; Y,
yellow; O, orange; R, red.

Vector-mediated expression of reporter genes. A fundamen- over time via the detection of reporter gene expression by BLI
tal requirement for BLI is the expression of a reporter gene (27, 35).
(e.g., luciferase) by the cells or tissues to be imaged, which
requires the introduction of genetically encoded imaging re- Applications of BLI in Cardiovascular Research
porters into cells cultured in vitro (50). Alternatively, lu-
ciferase-expressing cells can be obtained from luciferase trans- Because physiological processes are dynamic in time and
genic mice (Fig. 1). This can be achieved by using a reporter space, end-point assays do not always provide a comprehen-
vector that incorporates a luciferase gene driven by a promoter sive understanding of biology in vivo. In cardiovascular re-
that allows luciferase to be constitutively expressed by all cells search and many other scientific areas, BLI has been used for
in the animal’s body (11, 83). noninvasive visualization of a variety of biological processes in
Delivery of bioluminescent reporter genes to cells has been real time. In this section, we will review the many applications
achieved through several means, but lentiviral-based gene of BLI in cardiovascular research.
transfer has been the method of choice because of its effective Monitoring expression of therapeutic genes in the heart.
gene delivery and high expression levels of transgenes in Gene therapy is a rapidly evolving field in cardiovascular
mammalian cells in culture as well as in vivo (22). Lentiviruses medicine. This technology allows for the correction of func-
have the capability to deliver target genes to both dividing and tional gene loss and enables the expression of a therapeutic
nondividing cells and are capable of inserting genetic informa- gene in a target tissue (52). Nevertheless, gene therapy studies
tion into the host genome, ensuring prolonged gene expression continue to be plagued by suboptimal delivery of genes, poor
with a more limited host immune response (80). The safety of survival of cells carrying the therapeutic gene(s), and the
the lentiviral vectors has been further improved with the inability to evaluate the levels of gene expression in vivo (28).
generation of self-inactivating vectors and the use of minimal Thus the information gathered from BLI studies in small
packaging systems. The efficiency of gene expression has been animals can be used to design solid clinical trials that will help
improved by the introduction of a relatively strong internal improve gene therapy for cardiac repair. In cardiovascular
promoter such as cytomegalovirus. This promoter drives the research, BLI has been used to address a variety of questions
expression of the reporter gene and guarantees that the reporter that range from determining the effects of transgene expression
expression is always “on” under all conditions, in all tissue (e.g., BCL2) on cardiomyoblast survival and cardiac repair
types. Moreover, lentiviral vectors can be used to simultane- (40) to testing the efficiency of anti-inflammatory drugs on the
ously induce the expression of multiple genes in a cell. Cou- expression of specific genes (e.g., inducible nitric oxide syn-
pling the expression of a gene with a luciferase reporter gene thase) (99). Additionally, BLI has been used to optimize vector
provides a simple yet effective mechanism for studying the systems (33) and treatment regimens (71) for delivery of
regulation of gene expression and monitoring it by BLI. This therapeutic genes (e.g., hypoxia-inducible factor-1␣) to the
provides exciting opportunities for transcriptional targeting, heart.
double reporter labeling for BLI monitoring, as well as gene The applications of BLI in research are constantly expanding
therapy. For example, by linking the expression of luciferase to with the development of new therapeutic modalities. For in-
the cardiac-specific promoter myosin light chain 2v, it is stance, BLI enabled tracking the activity of a short hairpin
possible to monitor cells undergoing cardiac differentiation RNA plasmid in knocking down inhibitory factors of angio-

AJP-Heart Circ Physiol • VOL 301 • SEPTEMBER 2011 • www.ajpheart.org


Review
IN VIVO BIOLUMINESCENCE FOR TRACKING CELL FATE AND FUNCTION H667
genic genes in the heart (32). The advances in reporter gene BLI has demonstrated the poor survival of bone marrow
technology and dual reporter labeling have enabled a simulta- mononuclear cells (72), cardiac resident stem cells (43), mes-
neous monitoring of distinct physiological events via BLI. enchymal stem cells (83), adipose stromal cells (82), ESC-
Dual-reporter labeling can address questions such as how the derived cardiac cells (10), and ESC-derived endothelial cells
expression of a gene affects the survival or differentiation of a (45) by 8 wk after cell injection. Conversely, the tumorigenic
specific cell type or how it alters the expression of another potential of ESCs can also be uncovered by BLI. Lee et al. (42)
gene. However, challenges in designing dual-reporter systems, demonstrated that a minimum of 100,000 human ESCs are
such as finding adequate means for separating the two biolu- necessary to form teratoma in the heart. Overall, BLI studies
minescent signals while retaining sensitivity, remain (60, 87). have provided significant insights into the biology of stem cells
Monitoring survival and homing of therapeutic cells. Stem in vivo and how issues such as poor survival and potential
cell-based therapies are expected to have an enormous impact tumorigenicity must be overcome before translation into the
in the treatment and cure of various diseases and disorders in clinic.
the future. These cells, given the right conditions, have the Monitoring immune rejection of heart transplant and cell
ability to differentiate into the constituent cells of various grafts. Allogeneic heart transplantation is the most commonly
organs. When stem cells are used in therapies, it would be ideal used therapy for end-stage heart disease. Transplant loss due to
to track their migration or differentiation process in vivo. The immune rejection remains a significant problem in clinical
therapeutic use of bone marrow stem and progenitor cells heart transplantation despite current immunosuppressive ther-
initially was popular and has been evaluated furthest in the apies (73). Acute rejection of heart transplants is an immune
clinic setting. More recently, circulating stem and progenitor response mediated by the coordinated infiltration and function
cells, resident cardiac stem cells, and mesenchymal stem cells of host alloantigen-specific T cells in the allograft (57). In this
have also been used in translational studies for clinical appli-
context, BLI has allowed monitoring of cardiac allograft over
cations (15). Although small animal studies have provided
time and revealed its rejection by day 12 after transplantation.
promising results, significant questions regarding the biology
A decrease in the intensity of bioluminescence signals was
of transplanted cells and their ability to promote cardiac repair
remain (92) (Table 1). detected as early as day 4, suggesting acute graft rejection (78).
BLI is an important tool that can answer some of these The correlation of bioluminescence intensity to other measures
pressing questions by uncovering the dynamics of cell expan- of heart function such as beating score, fractional shortening,
sion, migration, and survival upon transplantation into the heart and lymphocyte infiltration has provided additional means to
(90, 102). Several studies have indicated that cardiomyocytes assess function and to determine the possible mechanisms
purified from embryonic stem cells (ESCs) can benefit cardiac responsible for rejection.
function following myocardial ischemia (10, 12, 13, 41, 59) Acute rejection can also be a major issue in human ESC-
with a low risk of tumor formation. However, one of the central based therapy (25, 84). BLI has provided significant informa-
issues in cell therapy is the lack of long-term survival of these tion for the development of therapeutic strategies to prevent the
therapeutic cells in vivo. BLI has shown that more than 90% of rejection of stem cells. An immunosuppressant cocktail con-
transplanted adult stem cells die within the first 3 wk of sisting of tacrolimus and sirolimus has been shown by BLI to
delivery (43, 45, 83). This could be the reason for the short mitigate the rejection of human ESCs (77). Recently, Pearl et
duration of improvement of cardiac function observed in mul- al. (63) used BLI to demonstrate that a cocktail of costimula-
tiple studies focused on cell therapy for cardiac repair. Thus the tory blockade agents (anti-CD40 ligand, cytotoxic T-lympho-
problem of donor cell death continues to be troublesome and cyte antigen 4-immunoglobulin, and anti-lymphocyte function-
may limit the overall efficacy of stem cell-based therapy, associated antigen-1) induced long-term allogeneic and xeno-
making further studies to improve cell fate monitoring via geneic human ESCs and induced pluripotent stem cell
imaging technologies essential. engraftment. Overall, these studies elucidate the importance of
Failure of long-term survival of therapeutic cells has been in vivo imaging for the development of therapies that may one
demonstrated for a variety of cell types (43, 45, 83) (Fig. 4). day enable stem cell therapy to become feasible.

Table 1. Cells types that have been investigated for their potential to promote cardiac repair using BLI
Cell Type Information Gained from BLI References

Bone marrow mononuclear cells Comparison of different cell types for treatment of MI 83
Timing of cell delivery on acute vs. chronic MI 76
Systemic homing to injured heart 72
Adipose tissue-derived stem cells (ASCs) Long-term survival of cells in injured heart 3
Homing and survival of fresh versus cultured cells to injured heart 4
Mesenchymal stem cells (MSCs) Comparison of ASCs vs. MSCs for treatment of MI 82
Growth factor-treated MSCs for treatment of MI 24
Human CD34⫹ cells Cell fate in the heart using a MI model 89
Rat cardiomyoblasts Improvement of engraftment and survival with collagen matrix 38, 39
Embryonic stem cells Cell survival, proliferation, and migration 9, 42, 77
Skeletal myoblasts Cell fate in MI model 83
ESC-derived endothelial cells (ESC-EC) Cell fate in MI model 44, 45
Effects of nicotine on the therapeutic effects ESC-ECs 98
Resident cardiac stem cells Cell fate and function in MI model 43
BLI, bioluminescence imaging; MI, myocardial infarction.

AJP-Heart Circ Physiol • VOL 301 • SEPTEMBER 2011 • www.ajpheart.org


Review
H668 IN VIVO BIOLUMINESCENCE FOR TRACKING CELL FATE AND FUNCTION

Fig. 4. Monitoring survival and homing of cells via BLI. A: skeletal myoblasts (SkMb), bone marrow-derived mononuclear cells (MN), mesenchymal stem cells
(MSC), and fibroblasts (Fibro) were injected intramyocardially after myocardial infarction. All cell types demonstrated a decrease in bioluminescence signal
intensity starting at 4 wk postinjection. Yellow arrows indicate homing of MN to femur, spleen, and liver. Red arrows indicate cells retained in the heart and
lungs. Values in y-axis are in photons·s⫺1·cm⫺2·sr⫺1. Abbreviations in x-axis: d, day; w, week. Reprinted with permission (83). B: BLI demonstrating preferential
homing of bone marrow mononuclear cells to the ischemic myocardium in a model of ischemic reperfusion injury (I/R). Bioluminescence was also detected in
the spleen and bone marrow (yellow and red arrows, respectively). A progressive decrease in bioluminescence signal was observed starting at day 14
postinjection. Reprinted with permission (72). C: cardiac resident stem cells were injected in the heart following myocardial infarction. Robust bioluminescence
activity was detected on day 2 but mostly disappeared by 8 wk postinjection. Values in x-axis represent days post-cell injection. Reprinted with permission (43).

Limitations to help define the requirements for imaging instrumentation.


However, the overall low spatial resolution (3–5 mm range)
BLI has been successfully used to obtain semiquantitative and limited tissue penetrance restrict the use of BLI to small
measurements of biological processes because of a strong animal studies (69).
correlation between the number of cells and the biolumines- Changes in tissue oxygenation can also alter biolumines-
cence signal detected both in vitro (75) and in vivo (68). cence signal. In rat gliosarcoma for instance, bioluminescence
However, a simple quantification of light emission may not signal has been shown to decrease by ⬃50% at 0.2% oxygen
provide a true representation of biological processes. This is (58). Thus, for reliable BLI measurements, it is important to
because the firefly luciferase reaction is a complex interaction understand the effects of local niche in which the luciferase-
of a variety of molecules (e.g., ATP, Mg2⫹, oxygen, and expressing cells of interest reside. This is especially the case in
luciferin) and because the intensity of the bioluminescence cardiovascular studies involving hypoxia (e.g., myocardial in-
signal depends on multiple factors. In particular, the number of farction). Similarly, BLI quantification has to be carefully
metabolically active luciferase-transfected cells, the concentra- interpreted in studies that involve surgical procedures. Changes
tion of luciferin, ATP and oxygen levels, the spectral emission in tissue thickness because of the presence of inflammation,
of bioluminescence probes, and the depth and optical proper- edema, sutures, and animal growth can alter light absorption
ties of tissues are known to alter the intensity of biolumines- and scattering as well as the bioluminescence signal.
cence signal (69). Another issue that should be considered
during quantitative BLI is the limited and wavelength-depen- Conclusions and Future Directions
dent transmission of light through animal tissues. Light sources
closer to the surface of the animal appear brighter compared Many different organisms, ranging from bacteria and fungi
with deeper sources because of tissue attenuation properties to fireflies and fish, are endowed with the ability to emit light.
(91). It is estimated that for every centimeter of depth, there is The discovery of new luminescence reporters and the use of
a 10-fold decrease in bioluminescence signal intensity (18). genetically modified reporters may further strengthen reporter
Mathematical models can be used to predict in vivo imaging gene expression in mammalian cells, thus improving the
signal levels and spatial resolution as a function of depth and sensitivity and expanding the applications of this imaging

AJP-Heart Circ Physiol • VOL 301 • SEPTEMBER 2011 • www.ajpheart.org


Review
IN VIVO BIOLUMINESCENCE FOR TRACKING CELL FATE AND FUNCTION H669
modality (7, 14). Additionally, alternative methods for 9. Cao F, Lin S, Xie X, Ray P, Patel M, Zhang X, Drukker M, Dylla SJ,
transduction of reporter gene may reduce the risk of anom- Connolly AJ, Chen X, Weissman IL, Gambhir SS, Wu JC. In vivo
visualization of embryonic stem cell survival, proliferation, and migra-
alous and inappropriate transcription of unwanted sequences tion after cardiac delivery. Circulation 113: 1005–1014, 2006.
in mammalian cells (37). 10. Cao F, Wagner RA, Wilson KD, Xie X, Fu JD, Drukker M, Lee A,
BLI allows real-time monitoring of survival and homing of Li RA, Gambhir SS, Weissman IL, Robbins RC, Wu JC. Transcrip-
various therapeutic cells that are currently being investigated to tional and functional profiling of human embryonic stem cell-derived
promote cardiac repair. By the use of cardiac-specific promot- cardiomyocytes. PLoS One 3: e3474, 2008.
ers linked to reporter genes, BLI can be used to monitor cell 11. Cao YA, Bachmann MH, Beilhack A, Yang Y, Tanaka M, Swijnen-
burg RJ, Reeves R, Taylor-Edwards C, Schulz S, Doyle TC, Fath-
differentiation in vivo. It also provides excellent opportunities man CG, Robbins RC, Herzenberg LA, Negrin RS, Contag CH.
to evaluate strategies designed to improve the survival of Molecular imaging using labeled donor tissues reveals patterns of en-
therapeutic cells or transplanted hearts. The ability to monitor graftment, rejection, and survival in transplantation. Transplantation 80:
physiological processes in vivo in real time is extremely 134 –139, 2005.
beneficial to evaluate the success or failure of the various 12. Caspi O, Huber I, Kehat I, Habib M, Arbel G, Gepstein A, Yankel-
son L, Aronson D, Beyar R, Gepstein L. Transplantation of human
therapies designed to promote cardiac repair. As most basic embryonic stem cell-derived cardiomyocytes improves myocardial per-
cardiovascular research is performed in rodent models, BLI formance in infarcted rat hearts. J Am Coll Cardiol 50: 1884 –1893, 2007.
can efficiently reveal problems and provide insight into solu- 13. Caspi O, Lesman A, Basevitch Y, Gepstein A, Arbel G, Habib IH,
tions for validating and optimizing novel therapies for the Gepstein L, Levenberg S. Tissue engineering of vascularized cardiac
treatment of heart disease. Overall, this technology can furnish muscle from human embryonic stem cells. Circ Res 100: 263–272, 2007.
great insights that can drive the development of better clinical 14. Caysa H, Jacob R, Muther N, Branchini B, Messerle M, Soling A. A
redshifted codon-optimized firefly luciferase is a sensitive reporter for
trials in cardiovascular medicine. bioluminescence imaging. Photochem Photobiol Sci 8: 52–56, 2009.
15. Chen IY, Wu JC. Cardiovascular molecular imaging: focus on clinical
ACKNOWLEDGMENTS translation. Circulation 123: 425–443, 2011.
16. Close DM, Patterson SS, Ripp S, Baek SJ, Sanseverino J, Sayler GS.
Because of space limitations, we were unable to cite all the important
Autonomous bioluminescent expression of the bacterial luciferase gene
papers relevant to bioluminescence imaging and cardiovascular research. We
apologize to investigators not mentioned here who have made significant cassette (lux) in a mammalian cell line. PLoS One 5: e12441, 2010.
contributions to this field. 17. Contag CH. In vivo pathology: seeing with molecular specificity and
cellular resolution in the living body. Annu Rev Pathol 2: 277–305, 2007.
18. Contag CH, Contag PR, Mullins JI, Spilman SD, Stevenson DK,
GRANTS Benaron DA. Photonic detection of bacterial pathogens in living hosts.
This work was supported in part by National Institutes of Health Grants Mol Microbiol 18: 593–603, 1995.
HL-093172, HL-099117, and EB-009689 (to J. C. Wu) and by the Interna- 19. Contag CH, Jenkins D, Contag FR, Negrin RS. Use of reporter genes
tional Society for Heart and Lung Transplantation (to P. E. de Almeida). for optical measurements of neoplastic disease in vivo. Neoplasia 2:
41–52, 2000.
20. Contag PR, Olomu IN, Stevenson DK, Contag CH. Bioluminescent
DISCLOSURES
indicators in living mammals. Nat Med 4: 245–247, 1998.
No conflicts of interest, financial or otherwise, are declared by the author(s). 21. Cui K, Xu X, Zhao H, Wong ST. A quantitative study of factors
affecting in vivo bioluminescence imaging. Luminescence 23: 292–295,
REFERENCES 2008.
22. De A, Yaghoubi SS, Gambhir SS. Applications of lentiviral vectors in
1. Asahara T, Murohara T, Sullivan A, Silver M, van der Zee R, Li T, noninvasive molecular imaging. Methods Mol Biol 433: 177–202, 2008.
Witzenbichler B, Schatteman G, Isner JM. Isolation of putative 23. de Wet JR, Wood KV, Helinski DR, DeLuca M. Cloning of firefly
progenitor endothelial cells for angiogenesis. Science 275: 964 –967, luciferase cDNA and the expression of active luciferase in Escherichia
1997. coli. Proc Natl Acad Sci USA 82: 7870 –7873, 1985.
2. Badr CE, Hewett JW, Breakefield XO, Tannous BA. A highly 24. Deuse T, Peter C, Fedak PW, Doyle T, Reichenspurner H, Zimmer-
sensitive assay for monitoring the secretory pathway and ER stress. PLoS mann WH, Eschenhagen T, Stein W, Wu JC, Robbins RC, Schrepfer
One 2: e571, 2007. S. Hepatocyte growth factor or vascular endothelial growth factor gene
3. Bai X, Yan Y, Coleman M, Wu G, Rabinovich B, Seidensticker M, transfer maximizes mesenchymal stem cell-based myocardial salvage
Alt E. Tracking long-term survival of intramyocardially delivered human after acute myocardial infarction. Circulation 120: S247–S254, 2009.
adipose tissue-derived stem cells using bioluminescence imaging. Mol 25. Drukker M, Benvenisty N. The immunogenicity of human embryonic
Imaging Biol 13: 633–645, 2011. stem-derived cells. Trends Biotechnol 22: 136 –141, 2004.
4. Bai X, Yan Y, Song YH, Seidensticker M, Rabinovich B, Metzele R, 26. Gritti A, Frolichsthal-Schoeller P, Galli R, Parati EA, Cova L,
Bankson JA, Vykoukal D, Alt E. Both cultured and freshly isolated
Pagano SF, Bjornson CR, Vescovi AL. Epidermal and fibroblast
adipose tissue-derived stem cells enhance cardiac function after acute
growth factors behave as mitogenic regulators for a single multipotent
myocardial infarction. Eur Heart J 31: 489 –501, 2010.
stem cell-like population from the subventricular region of the adult
5. Bhaumik S, Gambhir SS. Optical imaging of Renilla luciferase reporter
mouse forebrain. J Neurosci 19: 3287–3297, 1999.
gene expression in living mice. Proc Natl Acad Sci USA 99: 377–382,
2002. 27. Gruber PJ, Li Z, Li H, Worrad D, Huang B, Abdullah I, Wang W,
6. Branchini BR, Ablamsky DM, Davis AL, Southworth TL, Butler B, El-Deiry W, Ferrari VA, Zhou R. In vivo imaging of MLC2v-lu-
Fan F, Jathoul AP, Pule MA. Red-emitting luciferases for biolumines- ciferase, a cardiac-specific reporter gene expression in mice. Acad Radiol
cence reporter and imaging applications. Anal Biochem 396: 290 –297, 11: 1022–1028, 2004.
2010. 28. Gupta R, Tongers J, Losordo DW. Human studies of angiogenic gene
7. Branchini BR, Ablamsky DM, Murtiashaw MH, Uzasci L, Fraga H, therapy. Circ Res 105: 724 –736, 2009.
Southworth TL. Thermostable red and green light-producing firefly 29. Hastings JW, Gibson QH. The role of oxygen in the photoexcited
luciferase mutants for bioluminescent reporter applications. Anal luminescence of bacterial luciferase. J Biol Chem 242: 720 –726, 1967.
Biochem 361: 253–262, 2007. 30. Hastings JW, McElroy WD, Coulombre J. The effect of oxygen upon
8. Branchini BR, Southworth TL, DeAngelis JP, Roda A, Michelini E. the immobilization reaction in firefly luminescence. J Cell Physiol 42:
Luciferase from the Italian firefly Luciola italica: molecular cloning and 137–150, 1953.
expression. Comp Biochem Physiol B Biochem Mol Biol 145: 159 –167, 31. Hastings JW, Wilson T. Bioluminescence and chemiluminescence.
2006. Photochem Photobiol 23: 461–473, 1976.

AJP-Heart Circ Physiol • VOL 301 • SEPTEMBER 2011 • www.ajpheart.org


Review
H670 IN VIVO BIOLUMINESCENCE FOR TRACKING CELL FATE AND FUNCTION

32. Huang M, Chan DA, Jia F, Xie X, Li Z, Hoyt G, Robbins RC, Chen 52. Markkanen JE, Rissanen TT, Kivela A, Yla-Herttuala S. Growth
X, Giaccia AJ, Wu JC. Short hairpin RNA interference therapy for factor-induced therapeutic angiogenesis and arteriogenesis in the heart—
ischemic heart disease. Circulation 118: S226 –S233, 2008. gene therapy. Cardiovasc Res 65: 656 –664, 2005.
33. Huang M, Chen Z, Hu S, Jia F, Li Z, Hoyt G, Robbins RC, Kay MA, 53. Mccapra F, Hart R. The origins of marine bioluminescence. Nature
Wu JC. Novel minicircle vector for gene therapy in murine myocardial 286: 660 –661, 1980.
infarction. Circulation 120: S230 –S237, 2009. 54. Menasche P, Alfieri O, Janssens S, McKenna W, Reichenspurner H,
35. Kammili RK, Taylor DG, Xia J, Osuala K, Thompson K, Menick Trinquart L, Vilquin JT, Marolleau JP, Seymour B, Larghero J,
DR, Ebert SN. Generation of novel reporter stem cells and their Lake S, Chatellier G, Solomon S, Desnos M, Hagege AA. The
application for molecular imaging of cardiac-differentiated stem cells in myoblast autologous grafting in ischemic cardiomyopathy (MAGIC)
vivo. Stem Cells Dev 19: 1437–1448, 2010. trial: first randomized placebo-controlled study of myoblast transplanta-
36. Kawamoto A, Iwasaki H, Kusano K, Murayama T, Oyamada A, tion. Circulation 117: 1189 –1200, 2008.
Silver M, Hulbert C, Gavin M, Hanley A, Ma H, Kearney M, Zak V, 55. Meyer GP, Wollert KC, Lotz J, Pirr J, Rager U, Lippolt P, Hahn A,
Asahara T, Losordo DW. CD34-positive cells exhibit increased potency Fichtner S, Schaefer A, Arseniev L, Ganser A, Drexler H. Intracoro-
and safety for therapeutic neovascularization after myocardial infarction nary bone marrow cell transfer after myocardial infarction: 5-year fol-
compared with total mononuclear cells. Circulation 114: 2163–2169, low-up from the randomized-controlled BOOST trial. Eur Heart J 30:
2006. 2978 –2984, 2009.
37. Keravala A, Calos MP. Site-specific chromosomal integration mediated 56. Mias C, Lairez O, Trouche E, Roncalli J, Calise D, Seguelas MH,
by phiC31 integrase. Methods Mol Biol 435: 165–173, 2008. Ordener C, Piercecchi-Marti MD, Auge N, Salvayre AN, Bourin P,
38. Kutschka I, Chen IY, Kofidis T, Arai T, von Degenfeld G, Sheikh Parini A, Cussac D. Mesenchymal stem cells promote matrix metallo-
AY, Hendry SL, Pearl J, Hoyt G, Sista R, Yang PC, Blau HM, proteinase secretion by cardiac fibroblasts and reduce cardiac ventricular
Gambhir SS, Robbins RC. Collagen matrices enhance survival of fibrosis after myocardial infarction. Stem Cells 27: 2734 –2743, 2009.
transplanted cardiomyoblasts and contribute to functional improvement 57. Miura M, Morita K, Kobayashi H, Hamilton TA, Burdick MD,
of ischemic rat hearts. Circulation 114: I167–I173, 2006. Strieter RM, Fairchild RL. Monokine induced by IFN-gamma is a
39. Kutschka I, Chen IY, Kofidis T, von Degenfeld G, Sheikh AY, dominant factor directing T cells into murine cardiac allografts during
Hendry SL, Hoyt G, Pearl J, Blau HM, Gambhir SS, Robbins RC. In acute rejection. J Immunol 167: 3494 –3504, 2001.
vivo optical bioluminescence imaging of collagen-supported cardiac cell 58. Moriyama EH, Niedre MJ, Jarvi MT, Mocanu JD, Moriyama Y,
grafts. J Heart Lung Transplant 26: 273–280, 2007. Subarsky P, Li B, Lilge LD, Wilson BC. The influence of hypoxia on
40. Kutschka I, Kofidis T, Chen IY, von Degenfeld G, Zwierzchoniewska bioluminescence in luciferase-transfected gliosarcoma tumor cells in
M, Hoyt G, Arai T, Lebl DR, Hendry SL, Sheikh AY, Cooke DT, vitro. Photochem Photobiol Sci 7: 675–680, 2008.
Connolly A, Blau HM, Gambhir SS, Robbins RC. Adenoviral human 59. Mummery CL, van Laake LW, Passier R, den Ouden K, Schreurs C,
BCL-2 transgene expression attenuates early donor cell death after Monshouwer-Kloots J, Ward-van Oostwaard D, van Echteld CJ,
cardiomyoblast transplantation into ischemic rat hearts. Circulation 114: Doevendans PA. Improvement of mouse cardiac function by hESC-
I174 –I180, 2006. derived cardiomyocytes correlates with vascularity but not graft size.
41. Laflamme MA, Chen KY, Naumova AV, Muskheli V, Fugate JA, Stem Cell Res 3: 106 –112, 2009.
Dupras SK, Reinecke H, Xu C, Hassanipour M, Police S, O’Sullivan 60. Na IK, Markley JC, Tsai JJ, Yim NL, Beattie BJ, Klose AD, Holland
C, Collins L, Chen Y, Minami E, Gill EA, Ueno S, Yuan C, Gold J, AM, Ghosh A, Rao UK, Stephan MT, Serganova I, Santos EB,
Murry CE. Cardiomyocytes derived from human embryonic stem cells Brentjens RJ, Blasberg RG, Sadelain M, van den Brink MR. Con-
in pro-survival factors enhance function of infarcted rat hearts. Nat current visualization of trafficking, expansion, and activation of T lym-
Biotechnol 25: 1015–1024, 2007. phocytes and T-cell precursors in vivo. Blood 116: e18 –e25, 2010.
42. Lee AS, Tang C, Cao F, Xie X, van der Bogt K, Hwang A, Connolly 61. Niers JM, Kerami M, Pike L, Lewandrowski G, Tannous BA.
AJ, Robbins RC, Wu JC. Effects of cell number on teratoma formation Multimodal in vivo imaging and blood monitoring of intrinsic and
by human embryonic stem cells. Cell Cycle 8: 2608 –2612, 2009. extrinsic apoptosis. Mol Ther 19: 1090 –1096, 2011.
43. Li Z, Lee A, Huang M, Chun H, Chung J, Chu P, Hoyt G, Yang P, 62. Oh H, Bradfute SB, Gallardo TD, Nakamura T, Gaussin V, Mishina
Rosenberg J, Robbins RC, Wu JC. Imaging survival and function of Y, Pocius J, Michael LH, Behringer RR, Garry DJ, Entman ML,
transplanted cardiac resident stem cells. J Am Coll Cardiol 53: 1229 – Schneider MD. Cardiac progenitor cells from adult myocardium: hom-
1240, 2009. ing, differentiation, and fusion after infarction. Proc Natl Acad Sci USA
44. Li Z, Wilson KD, Smith B, Kraft DL, Jia F, Huang M, Xie X, 100: 12313–12318, 2003.
Robbins RC, Gambhir SS, Weissman IL, Wu JC. Functional and 63. Pearl JI, Lee AS, Leveson-Gower DB, Sun N, Ghosh Z, Lan F,
transcriptional characterization of human embryonic stem cell-derived Ransohoff J, Negrin RS, Davis MM, Wu JC. Short-term immunosup-
endothelial cells for treatment of myocardial infarction. PLoS One 4: pression promotes engraftment of embryonic and induced pluripotent
e8443, 2009. stem cells. Cell Stem Cell 8: 309 –317, 2011.
45. Li Z, Wu JC, Sheikh AY, Kraft D, Cao F, Xie X, Patel M, Gambhir 64. Pennell DJ. Cardiovascular magnetic resonance. Circulation 121: 692–
SS, Robbins RC, Cooke JP. Differentiation, survival, and function of 705, 2010.
embryonic stem cell derived endothelial cells for ischemic heart disease. 65. Quevedo HC, Hatzistergos KE, Oskouei BN, Feigenbaum GS, Ro-
Circulation 116: I46 –I54, 2007. driguez JE, Valdes D, Pattany PM, Zambrano JP, Hu Q, McNiece I,
46. Loening AM, Dragulescu-Andrasi A, Gambhir SS. A red-shifted Heldman AW, Hare JM. Allogeneic mesenchymal stem cells restore
Renilla luciferase for transient reporter-gene expression. Nat Methods 7: cardiac function in chronic ischemic cardiomyopathy via trilineage dif-
5–6, 2010. ferentiating capacity. Proc Natl Acad Sci USA 106: 14022–14027, 2009.
47. Loening AM, Fenn TD, Wu AM, Gambhir SS. Consensus guided 66. Raty JK, Liimatainen T, Unelma Kaikkonen M, Grohn O, Airenne
mutagenesis of Renilla luciferase yields enhanced stability and light KJ, Yla-Herttuala S. Non-invasive imaging in gene therapy. Mol Ther
output. Protein Eng Des Sel 19: 391–400, 2006. 15: 1579 –1586, 2007.
48. Loening AM, Wu AM, Gambhir SS. Red-shifted Renilla reniformis 67. Ray P, Bauer E, Iyer M, Barrio JR, Satyamurthy N, Phelps ME,
luciferase variants for imaging in living subjects. Nat Methods 4: 641– Herschman HR, Gambhir SS. Monitoring gene therapy with reporter
643, 2007. gene imaging. Semin Nucl Med 31: 312–320, 2001.
49. Lorenz WW, Mccann RO, Longiaru M, Cormier MJ. Isolation and 68. Rehemtulla A, Stegman LD, Cardozo SJ, Gupta S, Hall DE, Contag
Expression of a Cdna-Encoding Renilla-Reniformis Luciferase. Proc CH, Ross BD. Rapid and quantitative assessment of cancer treatment
Natl Acad Sci USA 88: 4438 –4442, 1991. response using in vivo bioluminescence imaging. Neoplasia 2: 491–495,
50. Luker GD, Pica CM, Song J, Luker KE, Piwnica-Worms D. Imaging 2000.
26S proteasome activity and inhibition in living mice. Nat Med 9: 69. Rice BW, Cable MD, Nelson MB. In vivo imaging of light-emitting
969 –973, 2003. probes. J Biomed Opt 6: 432–440, 2001.
51. Ma L, Xiang Z, Sherrill TP, Wang L, Blackwell TS, Williams P, 70. Rubinshtein R, Halon DA, Gaspar T, Jaffe R, Karkabi B, Flugelman
Chong A, Chari R, Yin DP. Bioluminescence imaging visualizes MY, Kogan A, Shapira R, Peled N, Lewis BS. Usefulness of 64-slice
activation of nuclear factor-kappaB in mouse cardiac transplantation. cardiac computed tomographic angiography for diagnosing acute coro-
Transplantation 85: 903–910, 2008. nary syndromes and predicting clinical outcome in emergency depart-

AJP-Heart Circ Physiol • VOL 301 • SEPTEMBER 2011 • www.ajpheart.org


Review
IN VIVO BIOLUMINESCENCE FOR TRACKING CELL FATE AND FUNCTION H671
ment patients with chest pain of uncertain origin. Circulation 115: labelling for non-invasive bioluminescence imaging of mesenchymal
1762–1768, 2007. stromal cell chondrogenic differentiation in demineralized bone matrix
71. Sato T, Ramsubir S, Higuchi K, Yanagisawa T, Medin JA. Vascular scaffolds. Biomaterials 30: 4986 –4995, 2009.
endothelial growth factor broadens lentivector distribution in the heart 88. Viviani VR, Arnoldi FG, Brochetto-Braga M, Ohmiya Y. Cloning and
after neonatal injection. J Cardiol 54: 245–254, 2009. characterization of the cDNA for the Brazilian Cratomorphus distinctus
72. Sheikh AY, Lin SA, Cao F, Cao Y, van der Bogt KE, Chu P, Chang larval firefly luciferase: similarities with European Lampyris noctiluca
CP, Contag CH, Robbins RC, Wu JC. Molecular imaging of bone and Asiatic Pyrocoelia luciferases. Comp Biochem Physiol B Biochem
marrow mononuclear cell homing and engraftment in ischemic myocar- Mol Biol 139: 151–156, 2004.
dium. Stem Cells 25: 2677–2684, 2007. 89. Wang J, Zhang S, Rabinovich B, Bidaut L, Soghomonyan S, Alaud-
73. Stehlik J, Edwards LB, Kucheryavaya AY, Aurora P, Christie JD, din MM, Bankson JA, Shpall E, Willerson JT, Gelovani JG, Yeh ET.
Kirk R, Dobbels F, Rahmel AO, Hertz MI. The registry of the Human CD34⫹ cells in experimental myocardial infarction: long-term
international society for heart and lung transplantation: twenty-seventh survival, sustained functional improvement, and mechanism of action.
official adult heart transplant report—2010. J Heart Lung Transplant 29: Circ Res 106: 1904 –1911, 2010.
1089 –1103, 2010.
90. Wang XL, Rosol M, Ge SD, Peterson D, McNamara G, Pollack H,
74. Sun X, Annala AJ, Yaghoubi SS, Barrio JR, Nguyen KN, Toyokuni
Kohn DB, Nelson MD, Crooks GM. Dynamic tracking of human
T, Satyamurthy N, Namavari M, Phelps ME, Herschman HR,
hematopoietic stem cell engraftment using in vivo bioluminescence
Gambhir SS. Quantitative imaging of gene induction in living animals.
Gene Ther 8: 1572–1579, 2001. imaging. Blood 102: 3478 –3482, 2003.
75. Sweeney TJ, Mailander V, Tucker AA, Olomu AB, Zhang W, Cao Y, 91. Weissleder R. A clearer vision for in vivo imaging. Nat Biotechnol 19:
Negrin RS, Contag CH. Visualizing the kinetics of tumor-cell clearance 316 –317, 2001.
in living animals. Proc Natl Acad Sci USA 96: 12044 –12049, 1999. 92. Welt FG, Losordo DW. Cell therapy for acute myocardial infarction:
76. Swijnenburg RJ, Govaert JA, van der Bogt KE, Pearl JI, Huang M, curb your enthusiasm? Circulation 113: 1272–1274, 2006.
Stein W, Hoyt G, Vogel H, Contag CH, Robbins RC, Wu JC. Timing 93. Widder EA. Bioluminescence in the ocean: origins of biological, chem-
of bone marrow cell delivery has minimal effects on cell viability and ical, and ecological diversity. Science 328: 704 –708, 2010.
cardiac recovery after myocardial infarction. Circ Cardiovasc Imag 3: 94. Wu JC, Chen IY, Sundaresan G, Min JJ, De A, Qiao JH, Fishbein
77–85, 2010. MC, Gambhir SS. Molecular imaging of cardiac cell transplantation in
77. Swijnenburg RJ, Schrepfer S, Govaert JA, Cao F, Ransohoff K, living animals using optical bioluminescence and positron emission
Sheikh AY, Haddad M, Connolly AJ, Davis MM, Robbins RC, Wu tomography. Circulation 108: 1302–1305, 2003.
JC. Immunosuppressive therapy mitigates immunological rejection of 94a.Wu JC, Inubushi M, Sundaresan G, Schelbert HR, Gambhir SS.
human embryonic stem cell xenografts. Proc Natl Acad Sci USA 105: Positron emission tomography imaging of cardiac reporter gene expres-
12991–12996, 2008. sion in living rats. Circulation 106: 180 –183, 2002.
78. Tanaka M, Swijnenburg RJ, Gunawan F, Cao YA, Yang Y, Caf- 95. Wu JC, Sundaresan G, Iyer M, Gambhir SS. Noninvasive optical
farelli AD, de Bruin JL, Contag CH, Robbins RC. In vivo visualiza- imaging of firefly luciferase reporter gene expression in skeletal muscles
tion of cardiac allograft rejection and trafficking passenger leukocytes of living mice. Mol Ther 4: 297–306, 2001.
using bioluminescence imaging. Circulation 112: I105–I110, 2005. 96. Wurdinger T, Badr C, Pike L, de Kleine R, Weissleder R, Breake-
79. Tannous BA, Kim DE, Fernandez JL, Weissleder R, Breakefield XO. field XO, Tannous BA. A secreted luciferase for ex vivo monitoring of
Codon-optimized Gaussia luciferase cDNA for mammalian gene expres- in vivo processes. Nat Methods 5: 171–173, 2008.
sion in culture and in vivo. Mol Ther 11: 435–443, 2005. 97. Ye L, Haider H, Tan R, Toh W, Law PK, Tan W, Su L, Zhang W,
80. Thomas CE, Ehrhardt A, Kay MA. Progress and problems with the use Ge R, Zhang Y, Lim Y, Sim EK. Transplantation of nanoparticle
of viral vectors for gene therapy. Nat Rev Genet 4: 346 –358, 2003. transfected skeletal myoblasts overexpressing vascular endothelial
81. Tromberg BJ, Shah N, Lanning R, Cerussi A, Espinoza J, Pham T, growth factor-165 for cardiac repair. Circulation 116: I113–I120, 2007.
Svaasand L, Butler J. Non-invasive in vivo characterization of breast 98. Yu J, Huang NF, Wilson KD, Velotta JB, Huang M, Li Z, Lee A,
tumors using photon migration spectroscopy. Neoplasia 2: 26 –40, 2000.
Robbins RC, Cooke JP, Wu JC. nAChRs mediate human embryonic
82. van der Bogt KE, Schrepfer S, Yu J, Sheikh AY, Hoyt G, Govaert
stem cell-derived endothelial cells: proliferation, apoptosis, and angio-
JA, Velotta JB, Contag CH, Robbins RC, Wu JC. Comparison of
genesis. PLoS One 4: e7040, 2009.
transplantation of adipose tissue- and bone marrow-derived mesenchy-
mal stem cells in the infarcted heart. Transplantation 87: 642–652, 2009. 99. Zhang N, Weber A, Li B, Lyons R, Contag PR, Purchio AF, West
83. van der Bogt KE, Sheikh AY, Schrepfer S, Hoyt G, Cao F, Ransohoff DB. An inducible nitric oxide synthase-luciferase reporter system for in
KJ, Swijnenburg RJ, Pearl J, Lee A, Fischbein M, Contag CH, vivo testing of anti-inflammatory compounds in transgenic mice. J
Robbins RC, Wu JC. Comparison of different adult stem cell types for Immunol 170: 6307–6319, 2003.
treatment of myocardial ischemia. Circulation 118: S121–S129, 2008. 100. Zhao H, Doyle TC, Coquoz O, Kalish F, Rice BW, Contag CH.
84. van der Bogt KE, Swijnenburg RJ, Cao F, Wu JC. Molecular imaging Emission spectra of bioluminescent reporters and interaction with mam-
of human embryonic stem cells: keeping an eye on differentiation, malian tissue determine the sensitivity of detection in vivo. J Biomed Opt
tumorigenicity and immunogenicity. Cell Cycle 5: 2748 –2752, 2006. 10: 41210, 2005.
86. Verkhusha VV, Otsuna H, Awasaki T, Oda H, Tsukita S, Ito K. An 101. Zhao H, Doyle TC, Wong RJ, Cao Y, Stevenson DK, Piwnica-Worms
enhanced mutant of red fluorescent protein DsRed for double labeling D, Contag CH. Characterization of coelenterazine analogs for measure-
and developmental timer of neural fiber bundle formation. J Biol Chem ments of Renilla luciferase activity in live cells and living animals. Mol
276: 29621–29624, 2001. Imaging 3: 43–54, 2004.
87. Vilalta M, Jorgensen C, Degano IR, Chernajovsky Y, Gould D, Noel 102. Zhou R, Acton PD, Ferrari VA. Imaging stem cells implanted in
D, Andrades JA, Becerra J, Rubio N, Blanco J. Dual luciferase infarcted myocardium. J Am Coll Cardiol 48: 2094 –2106, 2006.

AJP-Heart Circ Physiol • VOL 301 • SEPTEMBER 2011 • www.ajpheart.org

You might also like