ArticlePDF AvailableLiterature Review

The Study of Glioma by Xenotransplantation in Zebrafish Early Life Stages

Authors:

Abstract and Figures

Zebrafish (Danio rerio) and their transparent embryos are becoming an increasingly popular tool for studying processes involved in tumor progression and in the search for novel tumor treatment approaches. The xenotransplantation of fluorescently labeled mammalian cancer cells into zebrafish embryos is an approach enabling relatively high-throughput in vivo analyses. The small size of the embryos as well as the relative simplicity of their manipulation and maintenance allow for large numbers of embryos to be processed efficiently in a short time and at low cost. Furthermore, the possibility of fluorescence microscopic imaging of tumor progression within zebrafish embryos and larvae holds unprecedented potential for the real-time visualization of these processes in vivo. This review presents the methodologies of xenotransplantation studies on zebrafish involving research on tumor invasion, proliferation, tumor-induced angiogenesis and screening for antitumor therapeutics. We further focus on the application of these zebrafish to the study of glioma; in particular, its most common and malignant form, glioblastoma.
Content may be subject to copyright.
Journal of Histochemistry & Cytochemistry 2015, Vol. 63(10) 749 –761
© The Author(s) 2015
Reprints and permissions:
sagepub.com/journalsPermissions.nav
DOI: 10.1369/0022155415595670
jhc.sagepub.com
Review
Introduction
It is recognized that cancer results from complex interac-
tions of cancer cells with their microenvironment and the
whole organism. Thus, established in vitro cell and tissue
models of cancer must be complemented by in vivo models,
the former aiming at deciphering molecular mechanisms of
tumor progression, and the latter, elucidating multicellular
interactions during tumor progression. Commonly used
mammalian models have several drawbacks in that they are
expensive, time consuming, and ethically questionable; yet
are not necessarily a good approximation of physiological
processes taking place in the human body. In recent years,
steps have been taken towards bridging the gap between
high-throughput in vitro studies on the one hand and animal
cancer models on the other by introducing models with
higher throughput that are less ethically problematic.
Recently, the zebrafish (Danio rerio) and its embryos have
become a popular in vivo experimental model that enables
rapid, medium-throughput studies at low cost and offers the
possibility to image tumor progression directly at single-cell
resolution in real time (Armatruda et al. 2002; Konantz et al.
2005; Mione and Trede 2010; White at al. 2013; Zon and
Peterson 2005).
This review aims to provide a brief overview of the
methodology involved in studying human tumors by xeno-
transplantation into zebrafish embryos and the application
of this model to the study of gliomas, focusing on glioblas-
toma multiforme (GBM), the most common and aggressive
form of glioma (Behin et al. 2003; Ohgaki and Kleihues
2013). Several properties of GBM make these tumors dif-
ficult to treat. Their diffuse growth and invasion into sur-
rounding areas of the brain prevent their complete surgical
removal, whereas the presence of the blood-brain permea-
bility barrier (BBB) limits drug delivery (Claes et al. 2007;
595670JHCXXX10.1369/0022155415595670Vittori et al.Glioma Xenotransplantation in Zebrash
research-article2015
Received for publication April 28, 2015; accepted June 19, 2015.
Corresponding Author:
Miloš Vittori, Department of Genetic Toxicology and Cancer Biology,
National Institute of Biology, Večna pot 111, 1000 Ljubljana, Slovenia.
E-mail: milos.vittori@nib.si
The Study of Glioma by Xenotransplantation in Zebrafish
Early Life Stages
Miloš Vittori, Helena Motaln, and Tamara Lah Turnšek
Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia (MV, HM, TLT).
Summary
Zebrafish (Danio rerio) and their transparent embryos are becoming an increasingly popular tool for studying processes
involved in tumor progression and in the search for novel tumor treatment approaches. The xenotransplantation of
fluorescently labeled mammalian cancer cells into zebrafish embryos is an approach enabling relatively high-throughput in
vivo analyses. The small size of the embryos as well as the relative simplicity of their manipulation and maintenance allow
for large numbers of embryos to be processed efficiently in a short time and at low cost. Furthermore, the possibility of
fluorescence microscopic imaging of tumor progression within zebrafish embryos and larvae holds unprecedented potential
for the real-time visualization of these processes in vivo. This review presents the methodologies of xenotransplantation
studies on zebrafish involving research on tumor invasion, proliferation, tumor-induced angiogenesis and screening for
antitumor therapeutics. We further focus on the application of these zebrafish to the study of glioma; in particular, its most
common and malignant form, glioblastoma. (J Histochem Cytochem 63:749–761, 2015)
Keywords
cancer models, glioma, intravital microscopy, drug screening
750 Vittori et al.
Persano et al. 2013). The utilization of a complex in vivo
model that enables real-time imaging of cellular interac-
tions during GBM progression and its interactions with the
environment at single-cell resolution can contribute greatly
to the development of methods to improve the treatment of
this devastating disease. We discuss the potential use of
zebrafish xenotransplantation for the discovery of novel
pharmaceuticals and molecular markers with potential diag-
nostic, prognostic and therapeutic value.
The Zebrafish Cancer Model
Zebrafish and Their Early Life Stages in
Experimental Research
The zebrafish (Fig. 1) is a freshwater teleost that has been
extensively studied from developmental and genetic points
of view (Grunwald and Eisen 2002). It is an established
developmental model organism due to the ease of obtaining
and studying its embryos. The fecundity of zebrafish allows
even a small breeding facility to achieve a daily production
of embryos in their hundreds (Kari et al. 2007). Zebrafish
embryos are convenient, as they develop outside of their
parents’ bodies and can thus be monitored easily throughout
their development. The development of zebrafish embryos
is rapid: at 48 hours, an embryo already possesses a well-
developed nervous system and displays a functional circu-
lation as well as motility (Kimmel et al. 1995). In addition,
zebrafish embryos are small and are suitable for mainte-
nance on multi-well plates, making them an in vivo experi-
mental system with relatively high throughput at a
reasonable cost (Geiger et al. 2008; Kari et al. 2007; Zon
and Peterson 2005). Reverse genetic approaches are well
developed in this species (Hwang et al. 2013a, 2013b;
Lawson and Wolfe 2011; Zu et al. 2013), with a perspective
to produce knockout mutants for various genes in its
genome (Kettleborough et al. 2013).
Besides these benefits, the major advantage of zebrafish
embryos and larvae is their potential for in vivo visualiza-
tion of cellular processes at high resolution. This is due to
their small size and optical transparency, making in vivo
observations of developmental processes easy to accom-
plish at single cell resolution (Hendricks and Jesuthasan
2007; Keller et al. 2008). Intravital fluorescence micros-
copy of zebrafish embryos has been further enhanced by the
application of light sheet microscopy (Jung et al. 2012;
Kobitski et al. 2015). Although in vivo fluorescence imag-
ing of engrafted tumors has been performed on mammals
(Yang et al. 2001), they do not offer the high-resolution
imaging that can be performed in zebrafish embryos.
Furthermore, transgenic zebrafish strains with tissue-spe-
cific expression of fluorescent proteins are being produced
and are readily available for use (Distel et al. 2009; Lawson
and Weinstein 2002), enabling fluorescence imaging of
microanatomical structures and gene expression patterns;
this adds greatly to the value of this model.
Zebrafish in Cancer Research and the Tumor
Xenotransplantation Model
The zebrafish has been successfully utilized in cancer
research over the past decade (Feitsma and Cuppen 2008;
Konantz et al. 2012; Mimeault and Batra 2012; Veinotte
et al. 2014). Cancer in zebrafish has been induced by genetic
manipulation (Blackburn and Langenau 2014; Ignatius
et al. 2012; Langenau et al. 2003; Sabaawy et al. 2006) or
Figure 1. The zebrafish (Danio rerio) and its embryo. (A) Adult zebrafish. (B) The anatomy of a zebrafish embryo at 2 days after
fertilization. The areas of cancer cell implantation discussed in text are marked.
Glioma Xenotransplantation in Zebrafish 751
by xenotransplantation of cultured mammalian cancer cells
(Geiger et al. 2008; Haldi et al. 2006; Lee et al. 2005;
Konantz et al. 2012). Gene expression patterns of cancer
cells were shown to be highly similar in zebrafish and
humans (Lam et al. 2006; Zheng et al. 2014). Similar
molecular pathways may lead to tumor development in the
two species (Jung et al. 2013; Mione and Trede 2010) and
tumors in zebrafish were found to be histologically similar
to their mammalian counterparts (Armatruda et al. 2002;
Eden et al. 2014; Stern and Zon 2003). Furthermore, zebraf-
ish cells can respond to human signaling molecules
(Drabsch et al. 2013).
Because of the small size of zebrafish embryos, only a
few hundreds of cancer cells per embryo are generally
implanted (Konantz et al. 2012), and single-cell imaging
enables monitoring of the xenograft’s behavior in the
embryo. It has been proposed that implantation of a low
number of cells mirrors the early stages of tumor develop-
ment (Lal et al. 2012; Nicoli and Presta 2007). This also
makes the zebrafish xenotransplantation model particularly
suitable for the study of cells that are difficult to obtain,
such as cancer stem cells (Yang et al. 2013a).
Nevertheless, there are drawbacks or limitations to the
use of zebrafish in xenotransplantation experiments.
Because of the phylogenetic distance between teleost fish
and mammals, zebrafish might provide the implanted cells
with a different microenvironment than the human body,
especially when orthotopic implantation is impossible due
to the lack of corresponding organs in zebrafish (Konantz
et al. 2012). Furthermore, the use of embryos can be prob-
lematic due to the immaturity of their developing tissues.
For example, myelinated axonal sheaths do not develop in
the zebrafish central nervous system (CNS) until 4–7 days
post-fertilization (dpf) (Brösamle and Halpern 2002), which
may affect the invasion of implanted glioma cells (Lal et al.
2012). Furthermore, the BBB does not develop in zebrafish
embryos until 3 dpf (Xie et al. 2010) and is not mature for
another 7 days (Fleming et al. 2013), an issue important for
glioma drug screening.
Growing Human Tumors Underwater:
Approaches to Zebrafish Xenotransplantation
For cancer cell xenotransplantation, cells isolated from
tumors are grown in culture, labeled in order to distinguish
them from the recipient tissues, and implanted into the
zebrafish, after which the effect of experimental manipula-
tions on tumor progression are monitored (Fig. 2). Since the
zebrafish adaptive immune system matures at 3–4 weeks
after fertilization (Lam et al. 2004; Willett et al. 1999),
xenotransplantation in early life-stages does not necessitate
immunosuppression, making embryos particularly suitable
for xenotransplantation. An additional benefit is that
embryos do not need to be fed, relying on the yolk for
nutrition.
The zebrafish model is relatively novel and therefore
most methodology involved is not yet standardized. A vari-
ety of incubation conditions such as temperature and
Figure 2. Overview of the zebrafish glioma xenotransplantation model workflow. Cells isolated from a patient’s tumor are grown in
culture, fluorescently labeled, and implanted into the zebrafish embryos. After implantation, the embryos are incubated for several days,
during which time the behavior of the implanted cells can be monitored using fluorescence microscopy.
752 Vittori et al.
composition of the medium vary between laboratories, with
no agreement as yet on best practice.
A major issue in the use of zebrafish as recipients of
implanted mammalian cells is that the optimal temperature
for the development of zebrafish embryos is 28°C (Kimmel
et al. 1995), whereas human cells grow optimally at 37°C.
In some studies, some cancer cell lines are able to tolerate
28°C (Nicoli and Presta 2007; Zhao et al. 2009, 2011a). For
the GBM cell line, U251, it has been demonstrated that
varying the incubation temperature between 28°C and 35°C
has no effect on the survival of cells after engraftment
(Geiger et al., 2008). In other cases, the temperature has
been raised to 30°C (Geiger et al. 2008; Haldi et al. 2006;
Lally et al. 2007) or even as high as 35°C (Marques et al.
2009; Yang et al. 2013a, 2013b). Embryos can tolerate up to
35°C, although their survival is reportedly best at tempera-
tures below 33°C (Kimmel et al. 1995).
Implanted cells are generally rendered fluorescent to
visualize them within the body of the recipient embryo.
This can be achieved by staining cells prior to implantation
with stable fluorescent dyes, such as DiI, which are passed
onto the cell progeny (Jung et al. 2012; Lal et al. 2012; Lee
et al. 2009; Rouhi et al. 2010; Teng et al. 2013). The stain-
ing of cells prior to implantation is relatively fast and can
also be performed in primary cultures. In this way, the inva-
sive potential of cancer cells obtained from biopsies has
been assessed in vivo using the zebrafish model (Marques
et al. 2009), but studies on established cell lines have also
been performed with the use of dyes (Jung et al. 2012; Lal
et al. 2012). The disadvantage of using fluorescent dyes is
that their fluorescence can only decrease during the experi-
ment, making the quantification of cell proliferation by
means of fluorescence impossible. Furthermore, fluores-
cent dyes may be transferred to other cells.
An alternative to the fluorescence-labeling approach is
the establishment of cancer cell lines that stably express
fluorescent proteins (Drabsch et al. 2012; Eden et al. 2014;
Geiger et al. 2008; He et al. 2012; Yang et al. 2013a). The
fluorescence of endogenous fluorescent proteins is more
stable and correlates with cell number, enabling the quanti-
fication of proliferation by measuring fluorescence inten-
sity (Geiger et al. 2008; Vittori et al. 2014; Yang et al.
2014).
The crossing of different mutant zebrafish strains led to
the development of the Casper strain, which does not
develop skin pigmentation (White et al. 2008). The absence
of pigmentation aids microscopic observations because pig-
ments in the embryo may conceal the fluorescence emitted
by fluorescently labeled cells and only non-pigmented fish
enable quantitative intravital fluorescence imaging of
implanted cells. Despite its benefits, the Casper strain has
rarely been used in xenotransplantation studies (Corkery
et al. 2011; Eden et al. 2014). Instead, low concentrations of
phenylthiourea (PTU) are often used to inhibit melanin
synthesis (Lee et al. 2009; Yang et al. 2013a, 2013b, 2014).
Numerous studies have also used wild type embryos with
normally developed pigmentation for xenotransplantation
(Kitambi et al. 2014; Lally et al. 2007; Rampazzo et al.
2013).
Although human cells have been successfully engrafted
into zebrafish embryos during the late blastula stage (Geiger
et al. 2008; Lee et al. 2005; Zhao et al. 2009), cells are most
often implanted at 2 dpf, when the embryos develop all of
their major organ systems. Whereas some observations,
especially on angiogenesis and cell invasion, are concluded
at 5 dpf (Nicoli and Presta 2007; Yang et al. 2013a, 2013b),
longer studies, extending into the period of larval develop-
ment, have also been performed (Kitambi et al. 2014;
Pruvot et al. 2011). It is worth noting that the yolk is gener-
ally degraded at 5 dpf (Kimmel et al. 1995). When the yolk
mass is the site of implantation, this should be taken into
account, as the microenvironment of the implanted cells
will change with yolk resorption.
Applications of the Zebrafish Model
Studies on Cell Invasion and Metastasis
In the studies of cell invasion, the yolk sac is the most com-
mon area of implantation (Eguiara et al. 2011; Jung et al.
2012; Marques et al. 2009; Yang et al. 2013a). In this case,
cells are injected into the center of the yolk mass, a syncy-
tium containing nutrients required for embryonic develop-
ment. The movement of engrafted cells from the yolk to
other parts of the embryo can then be observed and quanti-
fied over a period of several days. The number of migrated
cells or the number of embryos in which invasion occurs are
determined (Eguiara et al. 2011; Marques et al. 2009; Yang
et al. 2013a).
It has been argued that quantification of the invasion of
cells from the yolk sac does not reflect their invasive poten-
tial, as cells may be passively transported to other parts of
the body via blood vessels (Drabsch et al. 2013). However,
differences in the capacity of cells to leave the yolk sac have
been demonstrated among cells grown in different culture
conditions (Eguiara et al. 2011; Yang et al. 2013a) and
among different cancer cell lines (Lee et al. 2009; Marques
et al. 2009), indicating that this is a valid model. The cor-
relation between the in vitro invasive potential of cell lines
and their ability to invade the body of the embryos has also
been established for cells implanted into the perivitelline
space, the cavity between the periderm forming the body
wall and yolk (Fig. 1B; Teng et al. 2013).
Alternatively, cells have been injected into to duct of
Cuvier (the cardinal vein of zebrafish embryos; Fig. 1B)
and allowed to spread throughout the body via the blood
circulation (Drabsch et al. 2013; He et al. 2012). Later, their
invasive potential can be assessed by counting cells located
Glioma Xenotransplantation in Zebrafish 753
within the tail fin, a structure that possesses no blood ves-
sels, thus ensuring that the cells had actively invaded the
tissue (Fig. 3A). This approach has been successfully
employed to study the involvement of neutrophils, which
may help to process the collagen matrix to facilitate cancer
cell invasion (He et al. 2012).
Angiogenesis may be involved in cancer cell dissemina-
tion from the injection site. For example, vascular endothelial
growth factor receptor (VEGFR) blockage was shown to
inhibit cell invasion from the perivitelline space (Lee et al.
2009), whereas hypoxia promoted it (Lee et al. 2009; Rouhi
et al. 2010). For the study of hypoxia-induced effects, zebraf-
ish embryos may be maintained in a hypoxic chamber (Rouhi
et al. 2010), although hypoxia may lead to developmental
abnormalities (Lee at al. 2009; Padilla and Roth 2001).
Zebrafish have also been used to study metastatic pro-
cesses. The transparency of embryos and the availability of
transgenic zebrafish embryos (fli1:GFP) expressing GFP in
their vascular endothelium (Lawson and Weinstein 2002)
has enabled the high-resolution imaging of processes
involved in metastasis (Stoletov et al. 2007). The metastatic
potential of cancer cells was associated with the upregula-
tion of specific molecular markers that are involved in
metastasis (Drabsch et al. 2013; He et al. 2012; Stoletov
et al. 2010). With cells implanted in the pericardium (Fig.
1B), cell invasion along the abluminal side of blood vessels
was also observed (Zhao et al. 2011). The results of these
studies show that zebrafish embryos can be used to evaluate
the invasive potential of cancer cells as well as the mecha-
nisms of metastasis.
Quantification of Cell Survival and Cell
Proliferation
The assessment of the effects of different treatments on the
proliferation of cancer cells through fluorescence quantifi-
cation has been successfully implemented (Eden et al.
2014; Geiger et al. 2008; Lally et al. 2007; Yang et al.
2014). An alternative approach is the use of biolumines-
cence imaging of implanted cells genetically modified to
express luciferase (Zhao et al. 2009). The advantage of bio-
luminescence is the low background signal as compared to
fluorescence, but it is less suitable for single-cell visualiza-
tion (Klerk et al. 2007). Alternative approaches for the
quantification of cancer cell proliferation include measur-
ing the projected tumor area on fluorescence micrographs
(Lal et al. 2012) and estimating the volume occupied by
cancer cells (Zhao et al. 2011a); however, these approaches
may not be indicative for proliferation, as an increase in
tumor volume can also result from cell dispersion or the
accumulation of host cells in the tumor (Lal et al. 2012).
Modeling Tumor-Induced Angiogenesis
Zebrafish embryos are particularly suitable for the study of
tumor-induced angiogenesis owing to the simplicity of their
cardiovascular system and its predictable patterning (Seabra
and Bhogal 2010; Stoletov et al. 2010; Tobia et al. 2011;
2013). The possibility to directly visualize the developing
vasculature is facilitated by the transparency of the embryos
and the availability of transgenic strains with fluorescent
Figure 3. Fluorescence observation of processes in zebrafish embryos in vivo. (A) A U87 DsRed cell (arrow) invading the tail fin of a
zebrafish embryo 2 days after implantation from the spinal cord. (B) Visualization of the vasculature in a living transgenic embryo at 1 day
after fertilization (courtesy of Marchien Dallinga). The embryo expresses GFP in the vascular endothelium. Scale, 100 µm.
754 Vittori et al.
blood vessels (Fig. 3B). To study tumor-induced angiogen-
esis, cells are usually implanted into the perivitelline space,
where the effect of implanted cells on the development of
the subintestinal vasculature is then monitored (Nicoli and
Presta 2007; Nicoli et al. 2007; Vitale et al. 2014; Zhao
et al. 2011a). Assessment of this effect can be rapid, as
changes can be observed within two days after tumor xeno-
grafting (Vitale et al. 2014; Yang et al. 2013b, 2014). The
quantification of the xenograft’s angiogenic effect involves
the determination of the percentage of embryos in which
abnormal angiogenesis is observed (Nicoli and Presta 2007;
Yang et al. 2013b) or measurement of parameters such as
vessel length, diameter, or the number of branching points
(Yang et al. 2013; Zhao et al. 2011). Alternatively, ex vivo
quantification of the frequency of tumor-induced angiogen-
esis and the number of generated blood vessels has been
performed using whole-mount alkaline phosphatase activ-
ity staining of the vasculature (Nicoli and Presta 2007;
Nicoli et al. 2007; Tobia et al. 2011).
The Zebrafish Xenotransplantation
Model in the Study of Glioma
Introducing Glioma to Zebrafish
Human glioma cells have successfully been transplanted
into zebrafish, and most of the methodological approaches
outlined above have been applied to study glioma. Unlike
many other mammalian tumors, glioma may be particularly
suitable for orthotopic xenotransplantation experiments in
zebrafish.
The yolk was the initial area of GBM cell implantation,
at first in blastula stage embryos and later in 2 dpf embryos
(Geiger et al. 2008; Lally et al. 2007; Yang et al. 2013a,
2013b). Apart from survival and proliferation, the assess-
ment of the invasion potential of glioma cells may be par-
ticularly important for the development of new treatment
approaches for this highly invasive type of cancer. Due to
the possible invasion of glioma cells into other parts of the
CNS, successful treatment of glioma by surgical removal
and directed radiotherapy is impossible (Claes et al. 2007).
Glioma cell lines that have so far been studied in zebrafish
do not demonstrate a great tendency to invade if implanted
into the yolk sac. Differentiated U87 GBM cells are not
invasive when implanted in this area (Lal et al. 2012; Yang
et al. 2013a, 2014), and invasion of U251 GBM cells has
not been reported (Geiger et al. 2008; Lally et al. 2007).
The inability of implanted cells to leave the yolk sac likely
reflects their low metastatic potential.
On the other hand, implantation into the yolk sac has
provided new insights into the role of glioma stem-like cells
(GSLCs) in tumor progression. One of the hallmarks of
GSLCs is the expression of the cell-surface marker CD133,
which is also used for isolating these cells. GSLCs have
been proposed to play important roles in glioma progres-
sion and radiotherapy resistance, which is attributed to their
more efficient DNA repair mechanisms (Bao et al. 2006;
Campos and Herold-Mende 2011; Hira et al. 2015; Molina
et al. 2014; Persano et al. 2013; Tabatabai and Weller 2011;
Wang et al. 2010). As a result, GSLCs are the focus for
research into targeted therapy, including studies using the
zebrafish model. To this end, CD133+ U87 cells were
obtained by single-cell cloning of U87 cells and growing
them under specific culture conditions (Yang et al. 2013a)
or by obtaining CD133+ cells by flow cytometry-based cell
sorting (Yang et al. 2013b, 2014). The CD133+ subpopula-
tion of U87 cells implanted into the yolk sac of 2 dpf zebraf-
ish embryos invaded the body more frequently than
CD133- cells (Yang et al. 2013a). These findings suggest
that GBM cells with stem cell properties are more invasive
than their differentiated counterparts.
Studies on the proliferation of GBM cells in the yolk sac
have provided somewhat conflicting results, as U87 cells
generally do not proliferate in this area (Lal et al. 2012;
Yang et al. 2014; Vittori et al. 2014) unless injected together
with Matrigel (Yang et al. 2014). In contrast, a study assess-
ing proliferation through bioluminescence showed the pro-
liferation of U87 cells within the yolk sac (Zhao et al. 2009).
Experiments on the cell line U251 also demonstrated mea-
surable proliferation in this area (Lally et al. 2007; Geiger
et al. 2008).
To study GBM-induced angiogenesis in zebrafish
embryos, implantation into the perivitelline space—as has
been suggested for other tumor types (Nicoli and Presta
2007)—may not be necessary, as remodeling of the vascu-
lature has been observed in embryos in which U87 cells
were implanted in the center of the yolk mass (Yang et al.
2013b). The implantation of U87 cells in the yolk resulted
in better survival as compared with their implantation into
the perivitelline space. Angiogenic activity of xenotrans-
planted U251 cells in the yolk mass has also been demon-
strated (Geiger et al. 2008). This approach has been applied
to study transforming growth factor-β (TGF-β) involve-
ment in angiogenesis by pretreating U87 cells with TGF-β
and implanting them in the yolk sac of 2 dpf embryos.
Pretreatment resulted in enhanced xenograft-induced
angiogenesis in the area of U87 cell implantation (Yang
et al. 2013b).
Location Matters: Orthotopic Studies on Glioma
In zebrafish, the CNS begins to form as early as 9 hr after
fertilization and, by 2 dpf, the brain, spinal cord, eyes and
the inner ear are well formed. The CNS is a relatively large
structure within the embryo’s body (Fig. 4), which speaks in
favor of orthotopic xenotransplantation of GBM cells. The
advantages and disadvantages of implanting cells in the
embryonic brain are summarized in Table 1.
Glioma Xenotransplantation in Zebrafish 755
Orthotopic implantation of glioma cells is advanta-
geous over yolk sac implantation, as the CNS represents
an environment that is similar to that of humans. In addi-
tion, orthotopic implantation can improve the survival and
proliferation of glioma cells as compared to implantation
into the yolk sac (Figs. 5, 6; Vittori et al. 2014). Human
GBM cells have been successfully implanted into the
embryonic brain at two days after fertilization (Kitambi
et al. 2014; Vittori et al. 2014). However, most orthotopic
studies have used older developmental stages, such as lar-
vae and juveniles (Eden et al. 2014; Lal et al. 2012;
Rampazzo et al. 2013). Lal et al. (2012) assessed the inva-
siveness of GBM cells implanted into the brain of either
4-day-old embryos or 10-day-old larval zebrafish.
Implanted DiI-labeled U87 cells were shown to disperse in
the larval brain, moving predominantly along the brain
vasculature, which is also a major track for invading GBM
cells in the human brain.
Rampazzo et al. (2013) implanted patient-derived
CD133+ glioma cells, obtained by cell sorting and trans-
fected with the GFP gene, into zebrafish larvae at 7 dpf. In
this study, the implanted CD133+ cells underwent cell
cycle arrest, decreased the expression of stem cell markers
and increased the expression of neuronal markers. It was
suggested that the apparent loss of stemness is linked to
Wnt-signaling. The activity of Wnt signaling was visual-
ized in vivo by using transgenic zebrafish larvae express-
ing a fluorescent protein upon activation of a Wnt-associated
transcription regulator. As demonstrated by Yang et al.
(2013a), the percentage of CD133+ U87 cells was greatly
reduced in the body of embryos at two days after implanta-
tion of a CD133+-enriched population of this cell line into
Figure 4. Confocal image of methyl green staining in a zebrafish embryo at 3 days after fertilization, showing the relative size and basic
anatomy of its central nervous system. The brain is one of the largest structures within the body of the embryo, facilitating orthotopic
implantation of glioma cells. Scale, 200 µm.
756 Vittori et al.
the yolk sac. The mechanism of this reduction is not clear.
Taken together, these results suggest that the zebrafish
embryonal environment may promote differentiation of
GSLCs, which would have great impact on the type of
research that can be applied to GSLCs.
Bigger Fish to Fry: Studying Glioma in Juvenile
Zebrafish
The xenotransplantation approach has also been applied to
older zebrafish that are approximately 30 days old (Eden
et al. 2014; Stoletov and Klemke 2008; Stoletov et al.
2007). The possible advantages and disadvantages of this
approach are summarized in Table 2. It has been argued
that the use of juveniles may provide more similarity to
the native tumor environment. However, the application of
xenotransplantation to older life stages comes at a cost.
The brain is not as easily imaged by optical microscopy in
juvenile and adult fish as in embryos and larvae, even in
the Casper strain (White at al. 2008), making older life
stages less suitable for in vivo imaging. Xenotransplantation
of cancer cells into juvenile fish requires immunosuppres-
sion of the recipient fish through the delivery of dexa-
methasone (Eden et al. 2014; Stoletov and Klemke 2008;
Tobia et al. 2013) or radiation (Taylor and Zon 2009;
Zhang et al. 2014). However, this is not necessarily a dis-
advantage, as GBM patients are often exposed to these
treatments as well.
In the study of Eden et al. (2014), xenografting of
cells derived from mouse brain tumors (including GBM)
was performed in 30-day-old zebrafish. The formation of
secondary tumor nodules elsewhere in the brain was
observed in these experiments, and tumor progression in
zebrafish was found to recapitulate that of the parent
tumor. In addition, the authors demonstrated similarities
between the transcriptomes of zebrafish tumors and their
corresponding mammalian tumors, which is an encour-
aging validation of the orthotopic xenografting approach
in zebrafish.
Application of Zebrafish Studies to Drug
Screening
Zebrafish embryos are an attractive model to study the
effects of novel pharmaceuticals, since small molecules are
taken up directly from the aqueous environment, making
chemical delivery simple and non-invasive (Drabsch et al.
2013; Taylor and Zon 2009; Yang et al. 2014). The simulta-
neous treatment of numerous embryos with small amounts
of chemicals enables a relatively high throughput of such
studies at low cost. Zebrafish embryos and larvae can thus
potentially bridge the gap between high-throughput in vitro
screening and mammalian models, as promising chemical
hits can first be validated in zebrafish and later studied in
greater detail in mammals (Jung et al. 2012; Stern and
Weinstein 2003).
The earliest application of zebrafish embryos to validate
glioma-related cytotoxic drug screening served to identify
potential small-molecule radiosensitizers. In this approach,
drug candidates were identified by in vitro screening and the
zebrafish embryo model was used to validate the in vitro
findings in an in vivo model. The novel radiosensitizer
NS-123, which increases the effects of radiation treatment
on GBM cells in vitro, was validated in vivo by implanting
U251 GBM cells in the yolk of blastula stage embryos (Lally
et al. 2007). This test enabled the simultaneous evaluation of
the potential toxic and teratogenic effects of the chemical.
The study demonstrated that the compound effectively
decreased the survival of GBM cells in the yolk sac follow-
ing radiation treatment without toxic effects to the zebrafish
embryos. Another validation of a therapeutic candidate in
zebrafish was performed with the macropinocytosis-target-
ing compound Vacquinol-1, which is reported to selectively
induce vacuolization and death of GBM cells (Kitambi et al.
2014). Zebrafish embryos at 2 dpf were orthotopically
implanted with fluorescently labeled U3013 GBM cells and
the tumor area was monitored for 10 days. The use of
Islet1:GFP transgenic zebrafish (Higashijima et al. 2000)
with a fluorescent CNS facilitated the visualization of
Table 1. Advantages and Disadvantages of the Yolk Sac or Brain for Glioma Cell Implantation.
Yolk Sac Brain
Advantages Disadvantages Advantages Disadvantages
Studying angiogenic
effects is straightforward
Yolk sac is degraded after a few
days
Is a permanent structure Studying angiogenic effects
may be difficult
Survival and proliferation of
glioma cells are limited
Glioma cells survive and
proliferate better
Limited suitability for studying
cell invasion
Suitable for studying cell invasion
Orthotopic implantation may give
more relevant results
Glioma Xenotransplantation in Zebrafish 757
implanted cancer cells in the CNS. In the study, several drug
candidates were first tested for toxicity in zebrafish embryos,
which helped to narrow down the candidates to a few
compounds. Vacquinol-1 was then demonstrated to most
effectively decrease the survival of GBM cells in the brain of
zebrafish larvae without causing toxic effects.
Figure 5. Time-lapse imaging of U87 DsRed glioblastoma cells
implanted in the brain of a zebrafish embryo at 2 days after
fertilization. Images were obtained at 4-hr intervals. Cells can be
seen invading the spinal cord in the posterior direction (arrows).
Scale, 300 µm.
Figure 6. Orthotopic xenotransplantation of fluorescent glioma
cells. (A) Fluorescent human glioblastoma cells (red) within
the brain of a zebrafish embryo at 3 days after implantation. (B)
Proliferation of implanted U-87 glioblastoma cells expressing the
fluorescent protein DsRed within the brain of a zebrafish embryo
over the course of several days. Day 1 marks the time point of
cell implantation at 2 days after fertilization. (C) Comparison of the
proliferation of implanted U-87 glioblastoma cells within the brain
and within the yolk sac of zebrafish embryos. Approximately 100
DsRed-expressing cells were implanted into the brain or the yolk
sac of zebrafish embryos at 2 days after fertilization, and the embryos
were incubated at 31°C for 3 days. The fluorescence emitted by the
cells was measured daily. Scale (A) 100 µm; (B) 250 µm.
758 Vittori et al.
The validity of zebrafish xenotransplantation in the
search of novel therapeutics for the treatment of glioma has
been shown in various studies. The antiangiogenic effects
of Axitinib, Suntinib and Vatalanib were tested in zebrafish
with U87 GBM cells implanted in the yolk sac. These
chemicals all inhibited tumor-induced vessel formation
(Yang et al. 2014). This study also demonstrated that the
novel anti-tumor compound Nordy inhibited angiogenesis
and increased the antiangiogenic effects of Vatalanib. The
radiosensitizing effects of temozolomide on U251 GBM
cells implanted in the yolk sac of blastula stage embryos
were also confirmed. In addition, temozolomide in concen-
trations that increased the toxicity of radiation to GBM cells
did not have a negative effect on embryo development
(Geiger et al. 2008). In a study on juvenile zebrafish at 30
dpf, the effects of two chemotherapeutics on the survival
and proliferation of implanted cancer cells were assessed:
tyrosine kinase inhibitor erlotinib and the pyrimidine ana-
log 5-fluorouracil (5-FU). In juveniles, 5-FU was adminis-
tered by dissolving it in the fish maintenance water, whereas
erlotinib was administered orally because of its low solubil-
ity (Eden et al. 2014). Quantification of cancer cell fluores-
cence demonstrated that 5-FU and erlotinib inhibited GBM
cell proliferation in vivo. Taken together, these studies dem-
onstrate that the zebrafish embryo model is a good model
for drug screening and preclinical validation.
Issues, Prospects and Conclusions
The zebrafish xenotransplantation cancer model is emerging
from its infancy and holds great promise for the visualiza-
tion of tumor progression and high-throughput therapeutic
compound validation. In this regard, the model fits between
in vitro cell studies and in vivo animal studies (Eden et al.
2014). It offers intermediate throughput, being more time
and labor consuming than in vitro experiments, yet faster
and more ethical than animal experimentation.
The strength of the model lies in its potential for single-
cell visualization and the ease of genetic manipulation; the
transgenic arsenal available for zebrafish has likely not
been fully exploited yet. The use of zebrafish with a fluo-
rescent vasculature is becoming routine in xenotransplan-
tation experiments (Geiger et al. 2008; Lee et al. 2009;
Stoletov et al. 2007; Yang et al. 2014), whereas other
structures, such as the CNS (Higashijima et al. 2000;
Zhang and Gong 2013) can also express fluorescent pro-
teins, enabling high-resolution imaging of implanted cells
and their microenvironment in vivo. In the future, several
genetically coded fluorescent labels may be used simulta-
neously to highlight different structures interacting with
the engrafted cells.
Zebrafish embryos are ideal for the use of fluorescent
reporter genes. For example, the generation of transgenic
zebrafish expressing three different molecular markers that
are characteristic for different levels of muscle cell differen-
tiation linked to three different fluorescent proteins pro-
vided unprecedented insights into the progression of
rhabdomyosarcoma induced by KRAS overexpression
(Ignatius et al. 2012). This methodology can also be applied
to xenotransplants, enabling visualization of gene expres-
sion patterns during cancer progression at single-cell reso-
lution. Indeed, the combination of the visualization of
implanted cells with the expression of fluorescent reporter
genes has been successfully applied to study glioma xeno-
grafts (Rampazzo et al. 2013), showing that this approach
has great potential.
Another possibility for future developments of the
zebrafish model is to study cancer cell heterogeneity
(Blackburn and Langenau 2014). For example, two cancer
cell lines expressing different fluorescent proteins have
been implanted simultaneously in a single embryo in order
to demonstrate the difference in their invasive potential
(Marques et al. 2009). In another study, wild type cells and
cells in which the expression of certain genes was upregu-
lated were labeled with different fluorescent proteins and
simultaneously implanted into zebrafish (Stoletov et al.
2010). This approach not only enables the simultaneous
visualization of the behavior of several cell types, but also
holds potential for the analysis of cell–cell interactions in
tumor progression.
Table 2. Advantages and Disadvantages of Zebrafish Embryos or Juveniles in Xenotransplantation Studies of Brain Tumors.
Embryos Juvenile Fish
Advantages Disadvantages Advantages Disadvantages
Transparency enables high-
resolution microscopic
observations
Experiments last only a few days Longer-term experiments are
possible
Reduced transparency limits
the resolution of microscopic
observations
No need for
immunosuppression
Can accommodate only a few
hundred mammalian cells
Can accommodate more
mammalian cells
Xenotransplantation requires
immunosuppression
No need for feeding Organ systems are still
developing
Most organ systems are mature Require feeding
Blood-brain barrier is developing Blood-brain barrier is functional
Glioma Xenotransplantation in Zebrafish 759
Steps have been taken to increase the throughput of
zebrafish xenotransplantation experiments (Snaar-Jagalska
2009), leading to a proposal of automated approaches to
cell implantation and imaging that focus predominantly on
the quantification of cell invasion from the yolk sac (Ghotra
et al. 2012). The study of this process can be primarily
applied to the screening and validation of anti-angiogenic
and anti-metastatic drugs. Nevertheless, ethical consider-
ations with respect to the use of embryos in large-scale
screens should not be ignored entirely.
Acknowledgments
The authors wish to thank Matjaž Novak, Dr. David Dobnik
(National Institute of Biology, Ljubljana) and Marchien Dallinga
(Academic Medical Center, Amsterdam) for their assistance.
Declaration of Competing Interests
The authors declared no potential competing interests with respect
to the research, authorship, and/or publication of this article.
Author Contributions
MV, HM and TLT all contributed to literature overview and writ-
ing of the manuscript.
Funding
The authors disclosed receipt of the following financial support
for the research, authorship, and/or publication of this article:
Slovenian Research Agency Program P1-0245; INTERREG EC
Project 2011, Ref. No. 42: Identification of novel biomarkers to
brain tumors - glioma, for diagnosis and as new therapeutic tar-
gets, Acronym: GLIOMA.
References
Armatruda JF, Shepard JL, Stern HM, Zon LI (2002). Zebrafish as
a cancer model system. Cancer Cell 1: 229-231.
Bao S, Wu Q, McLendon R, Hao Y, Shi Q, Hjelmeland A,
Dewhirst M, Bignern D, Rich J (2006). Glioma stem cells
promote radioresistance by preferential activation of the DNA
damage response. Nature 444:756-760.
Behin A, Hoang-Xuan K, Carpentier A, Delattre J-Y (2003).
Primary brain tumours in adults. Lancet 361:323-331.
Blackburn J, Langenau D (2014). Zebrafish as a model to assess
cancer heterogeneity, progression and relapse. Dis Model
Mech 7:755-762.
Brösamle C, Halpern M (2002). Characterization of myelination
in the developing zebrafish. Glia 39:47-57.
Campos B, Herold-Mende CC (2011). Insight into the complex
regulation of CD133 in glioma. Int J Cancer 128:501-510.
Claes A, Idema A, Wesseling P (2007). Diffuse glioma growth: a
guerilla war. Acta Neuropathol 114:443-458.
Corkery D, Dellaire G, Berman J (2011). Leukaemia xenotrans-
plantation in zebrafish – chemotherapy response assay in
vivo. Br J Haematol 153:786-789.
Distel M, Wullimann M, Köster R (2009). Optimized Gal4 genet-
ics for permanent gene expression mapping in zebrafish. Proc
Natl Acad Sci USA 106:13365-13370.
Drabsch Y, He S, Zhang L, Snaar-Jagalska B, Dijke P (2013).
Transforming growth factor beta signalling controls human
breast cancer metastasis in a zebrafish xenograft model.
Breast Cancer Res 15:R106.
Eden C, Ju B, Murugesan M, Phoenix T, Nimmervoll B, Tong
Y, Ellison D, Finkelstein D, Wright K, Boulos N, Dapper
J, Thiruvenkatam R, Lessman C, Taylor M, Gilbertson R
(2014). Orthotopic models of pediatric brain tumors in zebraf-
ish. Oncogene 34:1736-1742.
Eguiara A, Holgado O, Beloqui I, Abalde L, Sanchez Y, Callol C,
Martin A (2011). Xenografts in zebrafish embryos as a rapid
functional assay for breast cancer stem-like cell identification.
Cell Cycle 10:3751-3757.
Feitsma H, Cuppen E (2008). Zebrafish as a cancer model. Mol
Cancer Res 6:685-694.
Fleming A, Diekmann H, Goldsmith PF (2013). Functional char-
acterisation of the maturation of the blood-brain barrier in lar-
val zebrafish. PLoS One 8:e77548.
Geiger G, Fu W, Kao G (2008). Temozolomide-mediated radio-
sensitization of human glioma cells in a zebrafish embryonic
system. Cancer Res 68:3396-3404.
Ghotra V, He S, Bont H, Ent W, Spaink H, Water B, Snaar-
Jagalska B, Danen E (2012). Automated whole animal bio-
imaging assay for human cancer dissemination. PloS One
7:e31281.
Grunwald DJ, Eisen JS (2002). Headwaters of the zebrafish
– emergence of a new model vertebrate. Nat Rev Genet 3:
717-724.
Haldi M, Ton C, Seng W, McGrath P (2006). Human melanoma
cells transplanted into zebrafish proliferate, migrate, produce
melanin, form masses and stimulate angiogenesis in zebraf-
ish. Angiogenesis 9:139-151.
He S, Lamers G, Beenakker J, Cui C, Ghotra V, Danen E, Meijer
A, Spaink H, Snaar-Jagalska B (2012). Neutrophil-mediated
experimental metastasis is enhanced by VEGFR inhibition in
a zebrafish xenograft model. J Pathol 227:431-445.
Hendricks M, Jesuthasan S (2007). Electroporation-based meth-
ods for in vivo, whole mount and primary culture analysis of
zebrafish brain development. Neural Dev 2:1-10.
Higashijima S, Hotta Y, Okamoto H (2000). Visualization of cra-
nial motor neurons in live transgenic zebrafish expressing
green fluorescent protein under the control of the Islet-1 pro-
moter/enhancer. J Neurosci 20:206-218.
Hira VVV, Ploegmakers KJ, Grevers F, Verbovšek U, Silvestre-
Roig C, Aronica E, Tigchelaar W, Turnsek TL, Molenaar
RJ, Van Noorden CJF (2015). CD133+ and nestin+ glioma
stem-like cells reside around CD31+ arterioles in niches that
express SDF-1α, CXCR4, osteopontin and cathepsin K. J
Histochem Cytochem 63:481-493.
Hwang W, Fu Y, Reyon D, Maeder M, Tsai S, Sander J, Peterson
R, Yeh J-R, Joung J (2013a). Efficient genome editing in
zebrafish using a CRISPR-Cas system. Nat Biotechnol
31:227-229.
Hwang W, Fu Y, Reyon D, Maeder M, Kaini P, Sander J, Joung
J, Peterson R, Yeh JR (2013b). Heritable and precise zebraf-
ish genome editing using a CRISPR-Cas system. PLoS One
8:e68708.
Ignatius M, Chen E, Elpek N, Fuller A, Tenente I, Clagg R, Liu
S, Blackburn J, Linardic C, Rosenberg A, Nielsen P, Mempel
760 Vittori et al.
T, Langenau D (2012). In vivo imaging of tumor-propagating
cells, regional tumor heterogeneity, and dynamic cell move-
ments in embryonal rhabdomyosarcoma. Cancer Cell 21:
680-693.
Jung DW, Oh ES, Park SH, Chang YT, Kim CH, Choi SY,
Williams D (2012). A novel zebrafish human tumor xenograft
model validated for anti-cancer drug screening. Mol Biosyst
8:1930-1939.
Jung I, Leem G, Jung D, Kim M, Kim E, Kim S, Park HC, Park S
(2013). Glioma is formed by active Akt1 alone and promoted
by active Rac1 in transgenic zebrafish. Neuro Oncol 15:290-
304.
Kari G, Rodeck U, Dicker A (2007). Zebrafish: An emerging
model system for human disease and drug discovery. Clin
Pharmacol Ther 82:70-80.
Keller P, Schmidt A, Wittbrodt J, Stelzer E (2008). Reconstruction
of zebrafish early embryonic development by scanned light
sheet microscopy. Science 322:1065-1069.
Kettleborough R, Busch-Nentwich E, Harvey S, Dooley C, Bruijn
E, Eeden F, Sealy I, White R, Herd C, Nijman I, Fényes F,
Mehroke S, Scahill C, Gibbons R, Wali N, Carruthers S,
Hall A, Yen J, Cuppen E, Stemple D (2013). A systematic
genome-wide analysis of zebrafish protein-coding gene func-
tion. Nature 496:494-497.
Kimmel CB, Ballard WW, Kimmel SR, Ullmann B, Schilling TF
(1995). Stages of embryonic development of the zebrafish.
Dev Dyn 203:253-310.
Kitambi S, Toledo E, Usoskin D, Wee S, Harisankar A, Svensson
R, Sigmundsson K, Kalderén C, Niklasson M, Kundu S,
Aranda S, Westermark B, Uhrbom L, Andäng M, Damberg P,
Nelander S, Arenas E, Artursson P, Walfridsson J, Nilsson K,
Hammarström L, Ernfors P (2014). Vulnerability of glioblas-
toma cells to catastrophic vacuolization and death induced by
a small molecule. Cell 157:313-328.
Klerk CPW, Overmeer RM, Niers TMH, Versteeg HH, Richel
DJ, Buckle T, Van Noorden CJF, van Tellingen O (2007).
Validity of bioluminescence measurements for noninvasive in
vivo imaging of tumor load in small animals. BioTechniques
43:S7-S13, S30.
Kobitski A, Otte J, Takamiya M, Schäfer B, Mertes J, Stegmaier
J, Rastegar S, Rindone F, Hartmann V, Stotzka R, García A,
Wezel J, Mikut R, Strähle U, Nienhaus G (2015). An ensem-
ble-averaged, cell density-based digital model of zebrafish
embryo development derived from light-sheet microscopy
data with single-cell resolution. Sci Rep 5:8601.
Konantz M, Balci T, Hartwig U, Dellaire G, André M, Berman J,
Lengerke C (2012). Zebrafish xenografts as a tool for in vivo
studies on human cancer. Ann N Y Acad Sci 1266:124-137.
Lal S, Du J, Tanguay R, Greenwood J (2012). Calpain 2 is required
for the invasion of glioblastoma cells in the zebrafish brain
microenvironment. J Neurosci Res 90:769-781.
Lally B, Geiger G, Kridel S, Arcury-Quandt A, Robbins M, Kock
N, Wheeler K, Peddi P, Georgakilas A, Kao G, Koumenis
C (2007). Identification and biological evaluation of a novel
and potent small molecule radiation sensitizer via an unbiased
screen of a chemical library. Cancer Res 67:8791-8799.
Lam S., Chua H., Gong Z, Lam T., Sin Y (2004). Development
and maturation of the immune system in zebrafish, Danio
rerio: a gene expression profiling, in situ hybridization and
immunological study. Dev Comp Immunol 28:9-28.
Lam S, Wu Y, Vega V, Miller L, Spitsbergen J, Tong Y, Zhan H,
Govindarajan K, Lee S, Mathavan S, Murthy K, Buhler D,
Liu E, Gong Z (2006). Conservation of gene expression sig-
natures between zebrafish and human liver tumors and tumor
progression. Nat Biotechnol 24:73-75.
Langenau D, Traver D, Ferrando A, Kutok J, Aster J, Kanki J,
Lin S, Prochownik E, Trede N, Zon L, Look A (2003). Myc-
induced T cell leukemia in transgenic zebrafish. Science
299:887-890.
Lawson ND, Weinstein BM (2002). In vivo imaging of embryonic
vascular development using transgenic zebrafish. Dev Biol
248:307-318.
Lawson N, Wolfe S (2011). Forward and Reverse Genetic
Approaches for the Analysis of Vertebrate Development in
the Zebrafish. Dev Cell 21:48-64.
Lee LMJ, Seftor EA, Bonde G, Cornell RA, Hendrix MJC (2005).
The fate of human malignant melanoma cells transplanted into
zebrafish embryos: assessment of migration and cell division
in the absence of tumor formation. Dev Dyn 233:1560-1570.
Lee S, Rouhi P, Jensen L, Zhang D, Ji H, Hauptmann G, Ingham
P, Cao Y (2009). Hypoxia-induced pathological angiogenesis
mediates tumor cell dissemination, invasion, and metasta-
sis in a zebrafish tumor model. Proc Natl Acad Sci U S A
106:19485-19490.
Marques I, Weiss F, Vlecken D, Nitsche C, Bakkers J, Lagendijk
A, Partecke L, Heidecke C-D, Lerch M, Bagowski C (2009).
Metastatic behaviour of primary human tumours in a zebraf-
ish xenotransplantation model. BMC Cancer 9:128.
Mimeault M, Batra S (2013). Emergence of zebrafish models in
oncology for validating novel anticancer drug targets and
nanomaterials. Drug Discov Today 18:128-140.
Mione M, Trede N (2010). The zebrafish as a model for cancer.
Dis Model Mech 3:517-523.
Molina ES, Pillat MM, Moura-Neto V, Lah TT, Ulrich H (2014).
Glioblastoma stem-like cells: approaches for isolation and
characterization. J Cancer Stem Cell Res 2:e1007.
Nicoli S, Presta M (2007). The zebrafish/tumor xenograft angio-
genesis assay. Nat Protoc 2:2918-2923.
Nicoli S, Ribatti D, Cotelli F, Presta M (2007). Mammalian tumor
xenografts induce neovascularization in zebrafish embryos.
Cancer Res 67:2927-2931.
Ohgaki H, Kleihues P (2013). The definition of primary and sec-
ondary glioblastoma. Clin Cancer Res 19:764-772.
Padilla PA, Roth MB (2001). Oxygen deprivation causes sus-
pended animation in the zebrafish embryo. Proc Natl Acad
Sci USA 98:7331-7335.
Persano L, Rampazzo E, Basso G, Viola G (2013). Glioblastoma
cancer stem cells: role of the microenvironment and therapeu-
tic targeting. Biochem Pharmacol 85:612-622.
Pruvot B, Jacquel A, Droin N, Auberger P, Bouscary D, Tamburini
J, Muller M, Fontenay M, Chluba J, Solary E (2011).
Leukemic cell xenograft in zebrafish embryo for investigat-
ing drug efficacy. Haematologica 96:612-616.
Rampazzo E, Persano L, Pistollato F, Moro E, Frasson C, Porazzi
P, Puppa A, Bresolin S, Battilana G, Indraccolo S, Kronnie
G, Argenton F, Tiso N, Basso G (2013). Wnt activation
Glioma Xenotransplantation in Zebrafish 761
promotes neuronal differentiation of glioblastoma. Cell Death
Dis 4:e500.
Rouhi P, Jensen L, Cao Z, Hosaka K, Länne T, Wahlberg E,
Steffensen J, Cao Y (2010). Hypoxia-induced metastasis
model in embryonic zebrafish. Nat Protoc 5:1911-1918.
Sabaawy H, Azuma M, Embree L, Tsai H-J, Starost M, Hickstein
D (2006). TEL-AML1 transgenic zebrafish model of precur-
sor B cell acute lymphoblastic leukemia. Proc Natl Acad Sci
U S A 103:15166-15171.
Seabra R, Bhogal N (2010). In vivo research using early life stage
models. In Vivo 24:457-462.
Snaar-Jagalska BE (2009). ZF-CANCER: developing high-
throughput bioassays for human cancers in zebrafish.
Zebrafish 6:441-443.
Stern HM, Zon LI (2003). Cancer genetics and drug discovery in
the zebrafish. Nat Rev Cancer 3:1-7.
Stoletov K, Montel V, Lester R, Gonias S, Klemke R (2007).
High-resolution imaging of the dynamic tumor cell–vascular
interface in transparent zebrafish. Proc Natl Acad Sci U S A
104:17406-17411.
Stoletov K, Klemke R (2008). Catch of the day: zebrafish as a
human cancer model. Oncogene 27:4509-4520.
Stoletov K, Kato H, Zardouzian E, Kelber J, Yang J, Shattil S,
Klemke R (2010). Visualizing extravasation dynamics of
metastatic tumor cells. J Cell Sci 123:2332-2341.
Tabatabai G, Weller M (2011). Glioblastoma stem cells. Cell
Tissue Res 343:459-465.
Taylor AM, Zon LI (2009). Zebrafish tumor assays: the state of
transplantation. Zebrafish 6:339-346.
Teng Y, Xie X, Wlaker S, White DT, Mumm JS, Cowel JK
(2013). Evaluating human cancer cell metastasis in zebrafish.
BMC Cancer 13:453.
Tobia C, Gariano G, Sena G, Presta M (2013). Zebrafish embryo
as a tool to study tumor/endothelial cell cross-talk. Biochim
Biophys Acta 1832:1371-1377.
Tobia C, Sena G, Presta M (2011). Zebrafish embryo, a tool to
study tumor angiogenesis. Int J Dev Biol 55:505-509.
Veinotte C, Dellaire G, Berman J (2014). Hooking the big one:
the potential of zebrafish xenotransplantation to reform can-
cer drug screening in the genomic era. Dis Model Mech 7:
745-754.
Vitale G, Gaudenzi G, Dicitore A, Cotelli F, Ferone D, Persani L
(2014). Zebrafish as an innovative model for neuroendocrine
tumors. Endocr Relat Cancer 21:R67-R83.
Vittori M, Podergajs N, Motaln H, Lah TT (2014). Orthotopic
xenotransplantation of human cancer cells into zebrafish
embryos for the in vivo study of glioblastoma. In: Heart of
Europe Zebrafish Meeting, September 17-19, 2014. Warsaw:
International Institute of Molecular and Cell Biology: p. 65.
Wang J, Wakeman T, Lathia J, Hjelmeland A, Wang X-F, White
R, Rich J, Sullenger B (2010). Notch promotes radioresis-
tance of glioma stem cells. Stem Cells 28:17-28.
White R, Sessa A, Burke C, Bowman T, LeBlanc J, Ceol C,
Bourque C, Dovey M, Goessling W, Burns C, Zon L (2008).
Transparent adult zebrafish as a tool for in vivo transplanta-
tion analysis. Cell stem cell 2:183–189.
White R, Rose K, Zon L (2013). Zebrafish cancer: the state of the
art and the path forward. Nat Rev Cancer 13:624-636.
Willett C, Cortes A, Zuasti A, Zapata A (1999). Early hematopoi-
esis and developing lymphoid organs in the zebrafish. Dev
Dyn 214:323-336.
Xie J, Farage E, Sugimoto M, Anand-Apte B (2010). A novel
transgenic zebrafish model for blood-brain and blood-retinal
barrier development. BMC Dev Biol 10:76.
Yang M, Baranov E, Li XM, Wang JW, Jiang P, Li L, Moossa AR,
Penman S, Hoffman M (2001). Whole-body and intravital
optical imaging of angiogenesis in orthotopically implanted
tumors. Proc Natl Acad Sci U S A 98:2616-2621.
Yang X, Chen G, Yu S, Xu C, Xin Y, Li T, Shi Y, Gu A,
Duan J, Qian C, Cui Y, Zhang X, Bian X (2013b). TGF-
β1 enhances tumor-induced angiogenesis via JNK path-
way and macrophage infiltration in an improved zebrafish
embryo/xenograft glioma model. Int Immunopharmacol
15:191-198.
Yang X, Cui W, Gu A, Xu C, Yu S, Li T, Cui Y, Zhang X,
Bian X (2013a). A novel zebrafish xenotransplantation
model for study of glioma stem cell invasion. PLoS One
8:e61801.
Yang X, Cui W, Yu S, Xu C, Chen G, Gu A, Li T, Cui Y, Zhang
X, Bian X (2014). A synthetic dl-nordihydroguaiaretic acid
(Nordy), inhibits angiogenesis, invasion and proliferation
of glioma stem cells within a zebrafish xenotransplantation
model. PLoS One 9:e85759.
Zhang B, Shimada Y, Kuroyanagi J, Nishimura Y, Umemoto
N, Nomoto T, Shintou T, Miyazaki T, Tanaka T (2014).
Zebrafish xenotransplantation model for cancer stem-like cell
study and high-throughput screening of inhibitors. Tumor
Biol 35:11861-11869.
Zhang X, Gong Z (2013). Fluorescent transgenic zebrafish
Tg(nkx2.2a:mEGFP) provides a highly sensitive monitoring
tool for neurotoxins. PLoS One 8:e55474.
Zhao C, Wang X, Zhao Y, Li Z, Lin S, Wei Y, Yang H (2011a). A
novel xenograft model in zebrafish for high-resolution inves-
tigating dynamics of neovascularization in tumors. PloS One
6:e21768.
Zhao C, Yang H, Shi H, Wang X, Chen X, Yuan Y, Lin S, Wei Y
(2011b). Distinct contributions of angiogenesis and vascular
co-option during the initiation of primary microtumors and
micrometastases. Carcinogenesis 32:1143-1150.
Zhao H, Tang C, Cui K, Ang B-T, Wong S (2009). A screening
platform for glioma growth and invasion using biolumines-
cence imaging. J Neurosurg 111:238-246.
Zheng W, Li Z, Nguyen A, Li C, Emelyanov A, Gong Z (2014).
Xmrk, Kras and Myc transgenic zebrafish liver cancer models
share molecular signatures with subsets of human hepatocel-
lular carcinoma. PLoS One 9:e91179.
Zon LI, Peterson RT (2005). In vivo drug discovery in the zebraf-
ish. Nat Rev Drug Discov 4:35-44.
Zu Y, Tong X, Wang Z, Liu D, Pan R, Li Z, Hu Y, Luo Z, Huang
P, Wu Q, Zhu Z, Zhang B, Lin S (2013). TALEN-mediated
precise genome modification by homologous recombination
in zebrafish. Nat Methods 10:329-331.
... The multifactorial, multistep nature of carcinogenesis resulting from complex interactions of cancer cells with their microenvironment and the whole organism calls for in vitro cancer models interpreting the molecular mechanisms of tumor progression, complemented by in vivo models, deciphering the multicellular interactions of tumor progression [12]. Animals are necessarily used for preclinical brain tumor research including chemically induced, genetically engineered, and xenograft animal models, with the latter displaying high clinical relevance [5,13,14]. ...
... Recently, zebrafish (Danio rerio) have emerged as a promising alternative for in vivo studies, allowing for translatable brain cancer research and high-throughput drug screening. Their costefficient husbandry, high fecundity and rapid development ex utero, the small size and transparency of their embryos, as well as the availability of well-characterized zebrafish strains with fully sequenced genome-showcasing high genetic similarity to humans (70% genetic homology), and, thus, conferring interspecies biological processes conservation-configure zebrafish as a valuable tool to recapitulate glioblastoma in vivo with minimally invasive real-time imaging techniques at single-cell resolution [5,12,15]. ...
Article
Full-text available
Glioblastoma (GBM) constitutes the most common primary brain tumor in adults. The challenges in GBM therapeutics have shed light on zebrafish used as a promising animal model for preclinical GBM xenograft studies without a standardized methodology. This systematic review aims to summarize the advances in zebrafish GBM xenografting, compare research protocols to pinpoint advantages and underlying limitations, and designate the predominant xenografting parameters. Based on the PRISMA checklist, we systematically searched PubMed, Scopus, and ZFIN using the keywords “glioblastoma,” “xenotransplantation,” and “zebrafish” for papers published from 2005 to 2022, available in English. 46 articles meeting the review criteria were examined for the zebrafish strain, cancer cell line, cell labeling technique, injected cell number, time and site of injection, and maintenance temperature. Our review designated that AB wild-type zebrafish, Casper transparent mutants, transgenic Tg(fli1:EGFP), or crossbreeding of these predominate among the zebrafish strains. Orthotopic transplantation is more commonly employed. A number of 50–100 cells injected at 48 h post-fertilization in high density and low infusion volume is considered as an effective xenografting approach. U87 cells are used for GBM angiogenesis studies, U251 for GBM proliferation studies, and patient-derived xenograft (PDX) to achieve clinical relevance. Gradual acclimatization to 32–33 °C can partly address the temperature differential between the zebrafish and the GBM cells. Zebrafish xenograft models constitute valuable tools for preclinical studies with clinical relevance regarding PDX. The GBM xenografting research requires modification based on the objective of each research team. Automation and further optimization of the protocol parameters could scale up the anticancer drug trials.
... Additionally, this model has a similar microenvironment with regards to density, to what is observed in the human brain (50). Limitations of this model include differences in the tumor microenvironment compared to that of humans, and that the optimal temperature for human cells is 37°C compared to fish cells which is 28°C (50,69). Recently, a zebrafish model has been developed that can engraft human tumors at 37°C (70). ...
... • The brain is more difficult to image with age as optical transparency is ultimately lost • The glioma microenvironment is dissimilar to what is observed in humans • Limited glioma immunotherapy studies have been performed in this model (50,(67)(68)(69)(70)90) ...
Article
Full-text available
Gliomas have an extremely poor prognosis in both adult and pediatric patient populations as these tumors are known to grow aggressively and respond poorly to standard of care treatment. Currently, treatment for gliomas involves surgical resection followed by chemoradiation therapy. However, some gliomas, such as diffuse midline glioma, have more limited treatment options such as radiotherapy alone. Even with these interventions, the prognosis for those diagnosed with a glioma remains poor. Immunotherapy is highly effective for some cancers and there is great interest in the development of effective immunotherapies for the treatment of gliomas. Clinical trials evaluating the efficacy of immunotherapies targeted to gliomas have largely failed to date, and we believe this is partially due to the poor choice in pre-clinical mouse models that are used to evaluate these immunotherapies. A key consideration in evaluating new immunotherapies is the selection of pre-clinical models that mimic the glioma-immune response in humans. Multiple pre-clinical options are currently available, each one with their own benefits and limitations. Informed selection of pre-clinical models for testing can facilitate translation of more promising immunotherapies in the clinical setting. In this review we plan to present glioma cell lines and mouse models, as well as alternatives to mouse models, that are available for pre-clinical glioma immunotherapy studies. We plan to discuss considerations of model selection that should be made for future studies as we hope this review can serve as a guide for investigators as they choose which model is best suited for their study.
... In concert with transgenic models, zebrafish transplantation gained great attention as a useful model for biomedical research as well as for clinical studies when considering patient-derived xenografts (PDXs). This highly attractive model has been widely used in GBM research by several research teams because it faithfully reproduces tumor behavior in vivo [10,147]. The possibility to transplant patient cancer cells in zebrafish to evaluate and predict their aggressiveness, invasion, and metastatic potential characterizes this animal model as a valuable tool for prognosis. ...
Article
Full-text available
Glioblastoma multiforme (GBM) is a very aggressive and lethal primary brain cancer in adults. The multifaceted nature of GBM pathogenesis, rising from complex interactions between cells and the tumor microenvironment (TME), has posed great treatment challenges. Despite significant scientific efforts, the prognosis for GBM remains very poor, even after intensive treatment with surgery, radiation, and chemotherapy. Efficient GBM management still requires the invention of innovative treatment strategies. There is a strong necessity to complete cancer in vitro studies and in vivo studies to properly evaluate the mechanisms of tumor progression within the complex TME. In recent years, the animal models used to study GBM tumors have evolved, achieving highly invasive GBM models able to provide key information on the molecular mechanisms of GBM onset. At present, the most commonly used animal models in GBM research are represented by mammalian models, such as mouse and canine ones. However, the latter present several limitations, such as high cost and time-consuming management, making them inappropriate for large-scale anticancer drug evaluation. In recent years, the zebrafish (Danio rerio) model has emerged as a valuable tool for studying GBM. It has shown great promise in preclinical studies due to numerous advantages, such as its small size, its ability to generate a large cohort of genetically identical offspring, and its rapid development, permitting more time- and cost-effective management and high-throughput drug screening when compared to mammalian models. Moreover, due to its transparent nature in early developmental stages and genetic and anatomical similarities with humans, it allows for translatable brain cancer research and related genetic screening and drug discovery. For this reason, the aim of the present review is to highlight the potential of relevant transgenic and xenograft zebrafish models and to compare them to the traditionally used animal models in GBM research.
... Although evidence from in vitro studies indicates a protective effect against triple-negative breast cancer cells, an in vivo animal model is needed to confirm this effect in a more robust and systematic evaluation of the multicellular connections associated with tumor progression. In vivo models are valuable for gaining insights into the molecular pathways and clarifying the multicellular connections associated with tumor progression [17]. The zebrafish animal model is considered a good candidate for investigating the effect of matcha on cancer. ...
Article
Full-text available
Cancer is the second leading cause of death worldwide, and triple-negative breast cancer (TNBC) patients show the poorest prognosis and survival and the highest metastasis prevalence among all breast cancer subtypes. Matcha has recently been associated with multiple health benefits, and in vitro studies showed the potential effect of matcha in inhibiting cancer development and metastasis. We aimed to determine the safe, non-toxic dose of matcha suitable for zebrafish and to investigate the anticancer effect of matcha on the metastasis and growth of human TBNC cells using a zebrafish xenograft model. Wild-type AB zebrafish were used to conduct multiple general toxicity assessments, including developmental, neuromuscular, and cardiovascular toxicities. The safe, non-toxic concentration of matcha was determined to be 50 µg/mL and 100 µg/mL. Afterward, the zebrafish xenograft model was successfully established for MDA-MB-468 and MDA-MB-231 TNBC cells. The tumor size and metastasis of the injected cancer cells were traced through CM-Dil red fluorescent dye. Upon exposure to matcha at the safe doses, MDA-MB-231 and MDA-MB-468 showed a trend toward reduction in tumor size in a dose-dependent manner, indicated by quantified fluorescence. Matcha also visibly suppressed metastasis of cancer cells in the zebrafish body. Our results point to a potential dose-dependent anticancer effect of matcha on TNBC cells; however, more extended observation periods after xenotransplantation are required to confirm the long-term anticancer effect of matcha on tumor growth and metastasis.
... Indeed, cells stained with chemical dyes, such as CM-Dil, and the transparency of the zebrafish embryos allow for the in vivo dynamics of tumor development to be followed [24]. The xenotransplantation of human labeled GBM cells into the brain, in particular the hindbrain ventricle, allows for the orthotopic implantation of these cells and permits the evaluation of the GBM tumor invasion and the response to various drug treatments [25,26]. ...
Article
Full-text available
Glioblastoma multiforme (GBM) is the most common and malignant brain tumor in adults. The invasiveness and the rapid progression that characterize GBM negatively impact patients’ survival. Temozolomide (TMZ) is currently considered the first-choice chemotherapeutic agent. Unfortunately, over 50% of patients with GBM do not respond to TMZ treatment, and the mutation-prone nature of GBM enables the development of resistance mechanisms. Therefore, efforts have been devoted to the dissection of aberrant pathways involved in GBM insurgence and resistance in order to identify new therapeutic targets. Among them, sphingolipid signaling, Hedgehog (Hh) pathway, and the histone deacetylase 6 (HDAC6) activity are frequently dysregulated and may represent key targets to counteract GBM progression. Given the positive correlation between Hh/HDAC6/sphingolipid metabolism in GBM, we decided to perform a dual pharmacological inhibition of Hh and HDAC6 through cyclopamine and tubastatin A, respectively, in a human GMB cell line and zebrafish embryos. The combined administration of these compounds elicited a more significant reduction of GMB cell viability than did single treatments in vitro and in cells orthotopically transplanted in the zebrafish hindbrain ventricle. We demonstrated, for the first time, that the inhibition of these pathways induces lysosomal stress which results in an impaired fusion of lysosomes with autophagosomes and a block of sphingolipid degradation in GBM cell lines. This condition, which we also recapitulated in zebrafish embryos, suggests an impairment of lysosome-dependent processes involving autophagy and sphingolipid homeostasis and might be instrumental in the reduction of GBM progression.
... In zebrafish embryos, GBM cell xenografts exhibit more 3D confirmation mimicking in vivo tumour architecture. Moreover, GBM cell xenografts are surrounded by zebrafish brain and endothelial cells and extracellular matrix that can affect GBM cell phenotype and consequently their response to nanobody [31,44,45]. Importantly, the invasion inhibitory effect was not related to cytotoxicity of tumour cells, as the relative tumour growth did not decrease when the nanobody was added. ...
Article
Full-text available
Purpose: Glioblastoma (GBM) is the most common primary brain tumour and one of the deadliest cancers. In addition to late diagnosis and inadequate treatment, the extremely low survival rate is also due to the lack of appropriate therapeutic biomarkers and corresponding therapeutic agents. One of the potential therapeutic biomarkers is the intermediate filament vimentin, which is associated with epithelial-mesenchymal transition (EMT). The purpose of this study was to analyse the effect of the anti-vimentin nanobody Nb79 on cell invasion in vitro and in vivo. To further our understanding of the mechanism of action, we investigated the association between Nb79 and EMT in GBM and GBM stem cells by analysing the expression levels of key EMT-related proteins. Methods: The expression of vimentin in glioma tissues and cells was determined by RT-qPCR. An invasion assay was performed on differentiated glioblastoma cell line U-87 MG and stem cell line NCH421k in vitro as well as in vivo in zebrafish embryos. The effect of Nb79 on expression of EMT biomarkers beta-catenin, vimentin, ZEB-1 and ZO1 was determined by Western blot and immunocytochemistry. Results: Our study shows that vimentin is upregulated in glioblastoma tissue compared to lower grade glioma and non-tumour brain tissue. We demonstrated that treatment with Nb79 reduced glioblastoma cell invasion by up to 64% in vitro and up to 21% in vivo. In addition, we found that the tight junction protein ZO-1 had higher expression on the cell membrane, when treated with inhibitory anti-vimentin Nb79 compared to control. Conclusion: In conclusion, our results suggest that anti-vimentin nanobody Nb79 is a promising tool to target glioblastoma cell invasion.
... 22,84 The majority of researchers set 2 dpf as the time point for xenograft recipients, providing the possibility of more injection sites at this point. 47,[85][86][87] In addition to the yolk, the juvenile duct (located in the upper part of the yolk sac), the main vein (caudal region), and the perivitelline space are the three main sites for tumor transplantation, allowing for or direct release of cancer cells into the circulation system. 73,[88][89][90][91] Fortunately, a database named "ZenoFishDb" has already been developed to allow researchers to effectively understand the details of zebrafish tumor xenograft model construction, 92 where the specific information about the injection site and time have been demonstrated in the charts (Figures 2 and 3). ...
Article
The cost of antitumor drug development is enormous, yet the clinical outcomes are less than satisfactory. Therefore, it is of great importance to develop effective drug screening methods that enable accurate, rapid, and high‐throughput discovery of lead compounds in the process of preclinical antitumor drug research. An effective solution is to use the patient‐derived xenograft (PDX) tumor animal models, which are applicable for the elucidation of tumor pathogenesis and the preclinical testing of novel antitumor compounds. As a promising screening model organism, zebrafish has been widely applied in the construction of the PDX tumor model and the discovery of antineoplastic agents. Herein, we systematically survey the recent cutting‐edge advances in zebrafish PDX models (zPDX) for studies of pathogenesis mechanisms and drug screening. In addition, the techniques used in the construction of zPDX are summarized. The advantages and limitations of the zPDX are also discussed in detail. Finally, the prospects of zPDX in drug discovery, translational medicine, and clinical precision medicine treatment are well presented.
Article
As far as malignant tumors of the central nervous system are concerned, glioblastoma (GB) and neuroblastoma (NB) are the most prevalent, aggressive, and fatal forms in adult and pediatric populations, respectively. NB is the most prominent childhood extracranial compact neoplasm in pediatrics when the embryo develops from undifferentiated neural crest cells. Regarding malignant primary brain tumors, GB is the most lethal and difficult to treat. Currently, there are few effective treatments available for either condition. Research using zebrafish is relatively new in the field of animal cancer studies, and the first results show promise. In particular, integrated genomic investigations of NB and GB have revealed the potential of the zebrafish model in elucidating the roles of specific genetic changes in the development of this fatal childhood malignancy. Hence, this study examines the possibility of zebrafish as a model organism for discovering integrative medicines for these types of cancer. This model is an excellent animal model for study due to its transparency, ease of genetic modification, ethics and financial benefits, and preservation of the primary brain areas andbloodbrain barrier (BBB). This review provides recent developments in the zebrafish model of NB and GB to illustrate the benefits of using them in cancer studies as a model of the organism. This approach provides novel insights into delivering individualized treatment and enhancing outcomes for people coping with central nervous system malignancies.
Chapter
Reliable preclinical drug testing models for cancer research are urgently needed with zebrafish embryo models emerging as a powerful vertebrate model for xenotransplantation studies. Here, we describe the evaluation of toxicity, efficacy, and on-target activity of histone deacetylase (HDAC) inhibitors in a zebrafish embryo yolk sac xenotransplantation model of medulloblastoma and neuroblastoma cells. For this, we performed toxicity assays with our zebrafish drug library consisting of 28 clinically relevant targeted as well as chemotherapeutic drugs with zebrafish embryos. We further engrafted zebrafish embryos with fluorescently labeled pediatric tumor cells (SK-N-BE(2)-C, HD-MB03, or MED8A) and monitored the progression after HDAC inhibitor treatment of xenotransplanted tumors through tumor volume measurements with high-content confocal microscopy in a multi-well format. The on-target activity of HDAC inhibitors was verified through immunohistochemistry staining on paraffin-embedded early larvae. Overall, the zebrafish embryo xenotransplantation model allows for fast and cost-efficient in vivo evaluation of targeted drug toxicity, efficacy, and on-target activity in the field of precision oncology.
Chapter
The utility of zebrafish as a laboratory model has only been realized recently. It has helped gain great insights in the field of developmental biology and is only beginning to be employed for understanding mechanisms of diseases and finding appropriate therapeutic solutions. High fecundity, ex utero development of embryos which allow scope for manipulations, and the transparent body of developing embryos allowing in vivo imaging of the developing tissues and biological processes are some of the great advantages of zebrafish as a model organism. Tumors can be spontaneously induced in zebrafish by mutagen exposure or transgene technology. Cancers induced in zebrafish greatly resemble the human cancer in terms of histology and genetic makeup. Zebrafish not only allows modeling of various cancers but enables their characterization as well. Live imaging and chemical and genetic screens are powerful tools in the study of zebrafish cancer models. Broadly speaking, the major applications of zebrafish in the study of cancer include developing human cancer models, assessing various tumor-related processes such as angiogenesis and tumor metastasis, and screening small molecules with anticancer potential. These areas will be the major focus of this chapter, which will help to appreciate versatility of zebrafish in cancer study.KeywordsZebrafishCancerScreenModelMutationGeneticGenesTumor
Article
Full-text available
We generated germ line-transmitting transgenic zebrafish that express green fluorescent protein (GFP) in the cranial motor neurons. This was accomplished by fusing GFP sequences to Islet-1 promoter/enhancer sequences that were sufficient for neural-specific expression. The expression of GFP by the motor neurons in the transgenic fish enabled visualization of the cell bodies, main axons, and the peripheral branches within the muscles. GFP-labeled motor neurons could be followed at high resolution for at least up to day four, when most larval neural circuits become functional, and larvae begin to swim and capture prey. Using this line, we analyzed axonal outgrowth by the cranial motor neurons. Furthermore, by selective application of DiI to specific GFP-positive nerve branches, we showed that the two clusters of trigeminal motor neurons in rhombomeres 2 and 3 innervate different peripheral targets. This finding suggests that the trigeminal motor neurons in the two clusters adopt distinct fates. In future experiments, this transgenic line of zebrafish will allow for a genetic analysis of cranial motor neuron development.
Article
Full-text available
A long-standing goal of biology is to map the behavior of all cells during vertebrate embryogenesis. We developed digital scanned laser light sheet fluorescence microscopy and recorded nuclei localization and movement in entire wild-type and mutant zebrafish embryos over the first 24 hours of development. Multiview in vivo imaging at 1.5 billion voxels per minute provides “digital embryos,” that is, comprehensive databases of cell positions, divisions, and migratory tracks. Our analysis of global cell division patterns reveals a maternally defined initial morphodynamic symmetry break, which identifies the embryonic body axis. We further derive a model of germ layer formation and show that the mesendoderm forms from one-third of the embryo's cells in a single event. Our digital embryos, with 55 million nucleus entries, are provided as a resource.
Article
Full-text available
Poor survival of high-grade glioma is at least partly caused by glioma stem-like cells (GSLCs) that are resistant to therapy. GSLCs reside in niches in close vicinity of endothelium. The aim of the present study was to characterize proteins that may be functional in the GSLC niche by performing immunohistochemistry on serial cryostat sections of human high-grade glioma samples. We have found nine niches in five out of five high-grade glioma samples that were all surrounding arterioles with CD31+ endothelial cells and containing cellular structures that were CD133+ and nestin+. All nine niches expressed stromal-derived factor-1α (SDF-1α), its receptor C-X-C chemokine receptor type 4 (CXCR4), osteopontin and cathepsin K. SDF-1α plays a role in homing of CXCR4+ stem cells and leukocytes, whereas osteopontin and cathepsin K promote migration of cancer cells and leukocytes. Leukocyte-related markers, such as CD68, macrophage matrix metalloprotease-9, CD177 and neutrophil elastase were often but not always detected in the niches. We suggest that SDF-1α is involved in homing of CXCR4+ GSLCs and leukocytes and that cathepsin K and osteopontin are involved in the migration of GSLCs out of the niches.
Article
Full-text available
A new era in developmental biology has been ushered in by recent advances in the quantitative imaging of all-cell morphogenesis in living organisms. Here we have developed a light-sheet fluorescence microscopy-based framework with single-cell resolution for identification and characterization of subtle phenotypical changes of millimeter-sized organisms. Such a comparative study requires analyses of entire ensembles to be able to distinguish sample-to-sample variations from definitive phenotypical changes. We present a kinetic digital model of zebrafish embryos up to 16 h of development. The model is based on the precise overlay and averaging of data taken on multiple individuals and describes the cell density and its migration direction at every point in time. Quantitative metrics for multi-sample comparative studies have been introduced to analyze developmental variations within the ensemble. The digital model may serve as a canvas on which the behavior of cellular subpopulations can be studied. As an example, we have investigated cellular rearrangements during germ layer formation at the onset of gastrulation. A comparison of the one-eyed pinhead (oep) mutant with the digital model of the wild-type embryo reveals its abnormal development at the onset of gastrulation, many hours before changes are obvious to the eye.
Article
Full-text available
CONTENTS 1. Introduction 1 1.1. Brain tumor glioblastoma multiforme (GBM) 1 1.2. Cancer stem cell (CSC) paradigm 2 1.3. GBM stem-like cells (GSCs) 4 2. Isolation methodologies of GSCs 4 2.1. SP and ABC transporters 5 2.2. Isolation based on biomarkers 5 2.2.1. CD133 5 2.2.2. ALDH 5 2.2.3. Aptamers 5 3. Enrichment of GSC cultures 6 3.1. Effects of medium and substrates 6 3.2. Effects of hypoxia 7 3.3. Effects of endothelial cells 8 3.4. Effects of pericyte cells 9 3.5. Effects of immunosuppressed animals 9 4. Validation of the GSC phenotype 10 4.1. Sphere-formation assay 10 4.2. Tumor formation in vivo 10 4.3. Expression of biomarkers 11 4.4. Chemoresistance assay 11 4.5. Differentiation assay 12 5. Conclusions 12 Acknowledgments 13 References 13 Abstract: Glioblastoma (GBM) stem-like cells (GSCs) represent the most undifferentiated state of malignant cells with distinct biological characteristics. The fraction of these cells within a glial brain tumor, ranging from 2–30%, is correlating with the increasing WHO stage and poor prognosis of patients' survival. GSCs represent the least vulnerable, thus most preferential target cell population to be exposed to various therapeutic modalities, although the underlying mechanisms of this resistance are not yet fully understood. For the development of GSC-targeting therapies, further in depth studies are needed using enriched and stable GSC cell populations. Here, we discuss the current approaches of GSC isolation and validation based on expression of stemness and oncogenic markers as well as on functional assays. The enrichment of GSC phenotypes in established cell lines and/or primary tumor cultures, achieved by different strategies, is reviewed, providing a comprehensive comparison of selected studies and contemplating the characterization of the plethora of variants of reported GBM population exhibiting the GSC phenotype. ABBREVIATIONS: Aldehyde dehydrogenase (ALDH) Brain tumor initiating cells (BTIC) Cancer stem cell (CSC) Epidermal growth factor (EGF) Epithelial to mesenchymal transition (EMT) Fibroblast growth factor (FGF) Glioblastoma multiforme (GBM) Glioblastoma stem-like cell (GSC) Iso-dehydrogenase 1 (IDH1) Neural progenitor cell (NPC) Neural stem cell (NSC) O-6-methylguanine-DNA-methyltransferase (MGMT) Temozolomide (TMZ)
Article
Full-text available
Xenotransplantation studies are important tools for studying cancer biology, especially for assaying tumor cell malignancy and providing cancer information in vivo. Cancer stem-like cells (CSCs) have been identified in many cancer types to drive tumor growth and recurrence, from "keeping" to "keep" resistant to chemotherapy and radiation therapy. In this study, we developed the xenotransplantation of CSCs derived from the leukemia and solid tumor cell lines using the zebrafish models. In adult zebrafish, we investigated that the xenografted leukemia stem cells (LSCs) from K562 cells could proliferate in vivo and keep the cancer property by re-transplantation. As for the solid tumor, these CSCs from DU145 cells (human prostate cancer) and HepG2 cells (human liver cancer) could form the tumor mass and even metastasis after xenotransplantation. In addition, the zebrafish embryos with CSC xenotransplantation could evaluate docetaxel in vivo efficiently and be available to screen the novel inhibitors by high-throughput manner. In summary, these zebrafish xenotransplantation models devote a good platform for the CSC mechanism investigation and anti-CSC inhibitor screening.
Article
Full-text available
The current preclinical pipeline for drug discovery can be cumbersome and costly, which limits the number of compounds that can effectively be transitioned to use as therapies. Chemical screens in zebrafish have uncovered new uses for existing drugs and identified promising new compounds from large libraries. Xenotransplantation of human cancer cells into zebrafish embryos builds on this work and enables direct evaluation of patient-derived tumor specimens in vivo in a rapid and cost-effective manner. The short time frame needed for xenotransplantation studies means that the zebrafish can serve as an early preclinical drug screening tool and can also help personalize cancer therapy by providing real-time data on the response of the human cells to treatment. In this Review, we summarize the use of zebrafish embryos in drug screening and highlight the potential for xenotransplantation approaches to be adopted as a preclinical tool to identify and prioritize therapies for further clinical evaluation. We also discuss some of the limitations of using zebrafish xenografts and the benefits of using them in concert with murine xenografts in drug optimization.
Article
Full-text available
Clonal evolution is the process by which genetic and epigenetic diversity is created within malignant tumor cells. This process culminates in a heterogeneous tumor, consisting of multiple subpopulations of cancer cells that often do not contain the same underlying mutations. Continuous selective pressure permits outgrowth of clones that harbor lesions that are capable of enhancing disease progression, including those that contribute to therapy resistance, metastasis and relapse. Clonal evolution and the resulting intratumoral heterogeneity pose a substantial challenge to biomarker identification, personalized cancer therapies and the discovery of underlying driver mutations in cancer. The purpose of this Review is to highlight the unique strengths of zebrafish cancer models in assessing the roles that intratumoral heterogeneity and clonal evolution play in cancer, including transgenesis, imaging technologies, high-throughput cell transplantation approaches and in vivo single-cell functional assays.
Article
Mechanisms underlying pathological angiogenesis in relation to hypoxia in tumor invasion and metastasis remain elusive. Here, we have developed a zebrafish tumor model that allows us to study the role of pathological angiogenesis under normoxia and hypoxia in arbitrating early events of the metastatic cascade at the single cell level. Under normoxia, implantation of a murine T241 fibrosarcoma into the perivitelline cavity of developing embryos of transgenic fli1:EGFP zebrafish did not result in significant dissemination, invasion, and metastasis. In marked contrast, under hypoxia substantial tumor cells disseminated from primary sites, invaded into neighboring tissues, and metastasized to distal parts of the fish body. Similarly, expression of the hypoxia-regulated angiogenic factor, vascular endothelial growth factor (VEGF) to a high level resulted in tumor cell dissemination and metastasis, which correlated with increased tumor neovascularization. Inhibition of VEGF receptor signaling pathways by sunitinib or VEGFR2 morpholinos virtually completely ablated VEGFinduced tumor cell dissemination and metastasis. To the best of our knowledge, hypoxia- and VEGF-induced pathological angiogenesis in promoting tumor dissemination, invasion, and metastasis has not been described perviously at the single cell level. Our findings also shed light on molecular mechanisms of beneficial effects of clinically available anti-VEGF drugs for cancer therapy. Citation Format: Pegah Rouhi, Samantha Lin Lee, Lasse Dahl Jensen, Danfang Zhang, Hong Ji, Giselbert Hauptmann, Philip Ingham, Yihai Cao. Hypoxia-induced pathological angiogenesis mediates tumor cell dissemination, invasion, and metastasis in a zebrafish tumor model. [abstract]. In: Proceedings of the AACR Special Conference on Tumor Invasion and Metastasis; Jan 20-23, 2013; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2013;73(3 Suppl):Abstract nr B90.