Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 4893 word(s) 4893 2021-03-10 07:22:53 |
2 format correct Meta information modification 4893 2021-03-17 02:10:57 | |
3 format correct Meta information modification 4893 2021-03-17 02:11:38 | |
4 format correct Meta information modification 4893 2021-03-18 07:43:09 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Zhu, S.; Yeo, K.S. Zebrafish Models of Neuroblastoma. Encyclopedia. Available online: https://encyclopedia.pub/entry/8049 (accessed on 20 April 2024).
Zhu S, Yeo KS. Zebrafish Models of Neuroblastoma. Encyclopedia. Available at: https://encyclopedia.pub/entry/8049. Accessed April 20, 2024.
Zhu, Shizhen, Kok Siong Yeo. "Zebrafish Models of Neuroblastoma" Encyclopedia, https://encyclopedia.pub/entry/8049 (accessed April 20, 2024).
Zhu, S., & Yeo, K.S. (2021, March 16). Zebrafish Models of Neuroblastoma. In Encyclopedia. https://encyclopedia.pub/entry/8049
Zhu, Shizhen and Kok Siong Yeo. "Zebrafish Models of Neuroblastoma." Encyclopedia. Web. 16 March, 2021.
Zebrafish Models of Neuroblastoma
Edit

For nearly a decade, researchers in the field of pediatric oncology have been using zebrafish as a model for understanding the contributions of genetic alternations to the pathogenesis of neuroblastoma (NB), and exploring the molecular and cellular mechanisms that underlie neuroblastoma initiation and metastasis.

neuroblastoma zebrafish animal model

1. Introduction

Over the past ten years, zebrafish have become an increasingly popular tool for scientists conducting biomedical studies and other research. The species’ high fecundity rate, low cost of maintenance, and the ease of observation and genetic manipulation all contribute to its increasing use as an alternative and valuable vertebrate model system to study human disease. The expanding community of researchers using zebrafish has brought advanced technologies to the model, as well as a rapidly expanding inventory of transgenic and mutant lines that can be applied to different research niches. Cancer research using the zebrafish model can be traced back to 1965, when Dr. Mearle Stantion performed pioneered work to induce hepatic neoplasia in zebrafish with Diethylnitrosamine [1]. In 2003, the first zebrafish genetic cancer model was reported by Drs. David Langenau and Thomas Look, in which the MYC oncogene was overexpressed under control of the rag2 promoter, resulting in the development of T cell leukemia in the transgenic animal [2]. Since then many more zebrafish cancer models have been developed to understand the pathogenesis of leukemia, melanoma, rhabdomyosarcoma, hepatocellular carcinoma and many other tumor types [3][4][5][6]. In particular, the zebrafish model has also shown exceptional promise in dissecting the contributions of genetic alterations that were identified from integrative genomic analyses of neuroblastoma (NB) to the pathogenesis of this devastating pediatric cancer.

NB is the most common extracranial solid tumor in children and accounts for ~10% of all childhood cancer-related deaths [7]. It is derived from transformed neural crest progenitor cells in the developing peripheral sympathetic nervous system (PSNS) [8][9]. High-risk patients with amplified MYCN and over 18 months of age are often presented with widespread metastasis at diagnosis. Over the past few years, the five-year event-free survival rate for children with high-risk disease remains lower than 50% [10][11]. Very recently, a Phase III trial of immunotherapy, consisting of Dinutuximab, granulocyte macrophage-colony stimulating factor (GM-CSF) and interleukin-2 (IL2), showed significantly increased five-year overall survival rate of patients with high-risk NB to ~70% [12][13]. This immunotherapy has been approved by FDA for the treatment of patients with high-risk NB who achieve at least a partial response to prior first-line multiagent, multimodality therapy [12]. Although the improved outcomes are observed with the inclusion of Dinutuximab as part of treatment regimens for newly diagnosed NB, the prognosis for the relapsed disease remains poor (<10% progression-free survival) [14][15]. Therefore, better understanding of the pathogenesis of this disease and developing novel and more effective therapies are needed.

As an important member of the MYC proto-oncogene family identified from NB patients [16], MYCN amplification accounts for ~25% of NB cases and is associated with poor disease outcome [17][18][19]. MYCN is a bHLH transcription factor and is homologous to c-MYC structurally and functionally. It can promote neoplastic transformation of cultured mammalian cells and rat embryo fibroblasts [20][21]. In 1997, Dr. William Weiss developed the first animal model of NB by overexpressing MYCN under control of Tyrosine Hydroxylase (TH) in transgenic mice, which is by far still the most popular model for NB research [22]. Following Dr. Weiss’s effort, several genetically modified mouse (GEMM) lines with direct, conditional, inducible overexpression, knock-in or knockout of NB-relevant genes, including mutationally activated ALK (Anaplastic Lymphoma Receptor Tyrosine Kinase) [23][24][25], LIN28B (Lin-28 Homolog B) [26][27], SV40 large T antigen (Simian Vacuolating Virus 40 TAg) [28][29][30] and others [31][32] were subsequently developed. These models demonstrated a sufficient induction of NB in mice, which resemble the features of human NBs [24].

Although the mouse model provides valuable molecular insights on NB pathogenesis and opened the door for NB research, it has some disadvantages when compared to the zebrafish model. Neuroblastomas are different from adult tumors, in that they arise early in development; identifying the early onset of tumorigenesis in mice without euthanizing the animals is difficult and creates challenges in dissecting the molecular and cellular mechanisms underlying early onset tumor initiation. Zebrafish, by contrast, are translucent and develop from externally fertilized eggs, which allows for early detection of tumor onset in live animals. The zebrafish model is also more practical than GEMM, less expensive, and does not require sacrificing these animals to track tumor initiation and visualization of tumor growth. Therefore, the zebrafish model can serve as an alternative for the commonly used mouse model to conduct genetic research.

In 2012, the first zebrafish model of NB was generated and published by Zhu et al. [33]. Two oncogenes, MYCN and mutationally activated ALK (the most commonly mutated genes in primary neuroblastoma [34][35][36][37] and an attractive candidate for targeted therapy [38][39]), were expressed under control of the dopamine-beta-hydroxylase (dβh) promoter [33]. Following this initial effort on modeling NB in zebrafish, many new transgenic fish lines were developed, uncovering additional novel genetic alterations that cooperate with MYCN or c-MYC during NB pathogenesis. The evolution of zebrafish NB models has revealed the complexity of this disease at the molecular level and demonstrates the robustness of the model system in deepening our understanding of the molecular and cellular basis underlying NB pathogenesis. An overview of the NB zebrafish disease model workflow is illustrated in Figure 1.

Figure 1. An overview of the workflow using zebrafish model for NB study. Offspring from mating of wild-type (WT) or genetically engineered fish lines (A) can be subjected for (i) genome editing or transgene overexpression at one-cell stage (B), or (ii) transplantation of tumor cells at 2 days post fertilization for subsequent drug screening or functional analyses (C). The genetically modified embryos (B) can also be raised up for monitoring tumor development (D). Examples of crucial studies that can be performed using the zebrafish model are listed in the middle of the circle. This figure was created with BioRender.com.

 

Of course, the advances in understanding NB have not been achieved without obstacles and challenges, some of which appear daunting. In this paper, we aim to:

  • Compare and contrast the zebrafish model with other popular lab animals used as disease models in order to help cancer and other biomedical researchers determine appropriate models for experimental applications (Table 1); and
  • Provide an objective overview of the advancements in research that have occurred using the zebrafish NB model (Figure 2 and Table 2).
Figure 2. Cooperative contributions of diverse signaling pathways to the pathogenesis of NB—findings from zebrafish models. Blue lines connect cooperative genes in NB pathogenesis; Blue arrows indicate positive impact; Bar-headed lines indicate inhibitory effect; and Black lines indicate synergy between drugs. ALK, anaplastic lymphoma kinase; arid1a, AT-rich interacting domain–containing protein 1A; c-MYC, V-Myc avian myelocytomatosis viral oncogene homolog; DEF, digestive organ expansion factor; EGFR, epidermal growth factor receptor; Gab2, GRB2-associated-binding protein 2; LIN28B, lin-28 homolog B; LMO1, LIM domain only 1; MYCN, V-Myc avian myelocytomatosis viral oncogene neuroblastoma; nf1, neurofibromatosis type 1; PAG2G4, proliferation-associated protein 2G4; and PTPN11, protein tyrosine phosphatase non-receptor type 11. This figure was created with BioRender.com.
 

Table 1. Comparison of commonly used lab animal models in cancer research.

Table 2. Zebrafish models for the studies of the PSNS development and NB pathogenesis as well as evaluation of the efficacy of anti-NB drugs.

Since topics related to PSNS development in zebrafish and mammals and NB genetics have been previously covered in detail by ourselves and others [57][58], we will not be addressing these subjects in this review.

2. Advantages of Using Zebrafish as a Model for NB Research

2.1. Translucency of Juvenile and Adult Fish

The translucent appearance of both embryonic and adult zebrafish makes this species an obvious choice for NB study, since they can be observed while unharmed with the naked eye or under a fluorescent microscope using fluorescent probes (Table 1).

2.1.1. Early Detection of Tumor Onset

For the TH-MYCN mouse model, palpation is still the most common method used by researchers to examine tumor formation [22][53]. However, because tumors predominantly originate from the abdominal ganglion structures [22], it can be technically challenging for researchers to detect the early onset of tumorigenesis and track the rate of tumor progression by touch. Recently, PET or MRI scans have been used to evaluate the growth of TH-MYCN tumors in the presence or absence of different compounds, such as the Aurora A kinase or mTOR inhibitor [59][60]. These techniques seem promising and sensitive for monitoring tumor progression. However, they require expensive equipment, trained professionals, and lengthy procedures. In contrast, the onset of NB can be easily detected by fluorescent microscopy in the zebrafish model and large cohorts of animals (over 400 fish) can be monitored efficiently and regularly without harming the fish as early as 5 weeks post fertilization (wpf) [33][45][46]. Therefore, the zebrafish model allows demonstration of the cooperative contributions of multiple genetic alterations to NB tumorigenesis with high statistical power.

2.1.2. Real-Time Monitoring of Tumor Progression and Metastasis

In addition to being useful in the early detection of tumor onset, fluorescent-tagged tumor cells can be monitored for the growth of tumor, especially tumor metastasis in real-time. In transgenic fish with overexpression of MYCN and LMO1 oncogenes, fluorescent-positive tumor masses were observed using fluorescent microscopy in the distant regions from the primary tumor site, the interrenal gland region (IRG), as early as five weeks of age [46]. Using pathological and immunohistochemical analyses with antibodies against neuroblastoma markers, such as Tyrosine Hydroxylase (TH) [61] and HuC [62], widespread metastasis were detected in the orbit, gill, spleen, distal portion of kidney and heart [46]. Additional sites of metastases, such as the bone, the liver and the pancreas, were also observed in another newly developed zebrafish model with loss of function of gas7 gene [56].

Although there are anatomical differences between zebrafish and mammals, most of the organs in mammals have their functional equivalent counterparts in zebrafish. For example, the zebrafish IRG is equivalent to the human adrenal gland, which contains the chromaffin cells (an important cell lineage of origin for NB [63]) interposed with interrenal epithelial cells in the head kidney [64]. Similarly, although the lymph nodes are absent in zebrafish, the spleen and kidney (the sites where T cells, B cells, and dendritic cells reside) serve as secondary lymphoid organs in zebrafish [65][66]. The kidney marrows also function similarly to mammalian bone marrow [65]. The gills of zebrafish have a similar structure to the mammalian airways and fulfill the same gas-exchange function, making it analogous to the mammalian lung [67][68]. With such functional, physiological and anatomical homology of zebrafish to mammals, the metastases detected in the fish model match quite well to those metastatic sites commonly seen in human NB patients, including the bone marrow (70%), bone (55%), lymph nodes (30%), liver (30%), and brain (18%) [69]. In addition, increased numbers and networks of picrosirius red-stained collagen fibers, indicating the enhanced stiffness of extracellular matrix, were observed in the fish tumors with overexpression of both MYCN and LMO1 [46], which is consistent with the concept that increased ECM stiffness contributes to enhanced metastasis [70][71]. Hence, the zebrafish model offers a unique advantage in dissecting the molecular and cellular basis and the contribution of microenvironment to NB metastasis.

2.1.3. Efficient Evaluation of the Efficacy of Drug Treatment

It is challenging to develop effective and safe targeted therapies for cancer patients, especially for children. To do so requires using an entire organism to test the efficacy of new drugs or novel combinations thereof and evaluate the drug toxicity in order to develop new targeted therapies. Several NB-bearing zebrafish models have been used to demonstrate the effectiveness of novel inhibitors and combinations of new compounds in the treatment of NB, along with illuminating of the underlying mechanisms that may contribute to drug resistance in NB therapy (Figure 2).

For instance, using the dβh-MYCN zebrafish NB model (designated the MYCN line), Radic-Sarikas et al. demonstrated that the epidermal growth factor receptor (EGFR) kinase inhibitor, lapatinib, can prolong and enhance the cytotoxicity of YM155, an anti-cancer drug, by inhibiting the multidrug-resistance efflux transporter ABCB1 [47]. This led to the synergistic inhibition of the growth of MYCN-overexpressing NB in vivo. Similarly, using the transgenic fish with overexpression of both MYCN and proliferation associated 2G4 (PA2G4), Koach et al. demonstrated that a small molecule, WS6, can competitively bind to PA2G4 to prevent its interaction with MYCN, leading to destabilization and reduced expression of MYCN and in turn suppressed growth of NB [53]. Using the same zebrafish MYCN model together with the transgenic fish overexpressing both MYCN and GAB2, Zhang et al. demonstrated that the MEK inhibitor trametinib can enhance the sensitivity of MYCN and GAB2-overexpressing NB to the treatment of CBL0137, a histone chaperone FACT inhibitor [45]. In addition, He et al. applied the MEK inhibitor (trametinib) and retinoid (isotretinoin) on the juvenile compound fish with loss of nf1 in the context of MYCN overexpression and demonstrated synergistic killing of tumor cells by this combination treatment [44]. Taken together, these zebrafish NB models showed considerable translational potential for investigating new strategies to improve the treatment of this devastating childhood tumor.

2.2. Robustness in Genome Editing and Manipulation of Gene Expression

Since fish deposit oocytes outside of the body for external fertilization, researchers can easily collect fertilized embryos in large batches to perform genetic editing at the single-cell stage effectively and efficiently. The whole process—from breeding the fish to injections of transgene constructs or genome editing reagents into a few hundred embryos—takes only two days. Zebrafish are relatively simple to use when compared to rodent models that require multiple time-consuming steps including a period of superovulation by pregnant mare serum (PMS) in the early afternoon followed by human chorionic gonadotropin treatment [72], labor-intensive oocyte harvesting, in vitro fertilization, and embryo implantation.

The relative ease-of-use of zebrafish embryos compared to mice allows researchers to rapidly and economically develop a large number of genetically engineered fish lines in a short period of time. Various genetic editing methods have proven effective in zebrafish and this article will focus on approaches used in NB research and how they may be applicable and transferable to other research settings.

2.2.1. Retroviral-Mediated Mutagenesis

Retrovirus-mediated insertional mutagenesis in zebrafish was established in 1996 [73]. It is a powerful forward-genetic approach for identifying genes that are critical for embryonic development [74].

In 2009, led by Drs. Nancy Hopkins and Jacqueline Lees, a team of researchers performed a comprehensive insertional mutagenesis screen in zebrafish with a goal to identify genes susceptible to cancer. Four mutant fish lines with viral insertions in the fbxw4 gene were identified. Interestingly, the mis-regulated fibroblast growth factor 8 (fgf8) in these mutants was found to contribute to neuroblastomagenesis [40].

Although the forward-genetic approach was successful, it was quite challenging to screen for fish with spontaneous development of NB and to effectively identify disease-driving genes. Therefore, the reverse-genetic strategy became a more popular approach to model NB using the zebrafish in the past decade.

2.2.2. I-SceI Meganuclease-Mediated Transgenesis

Using a reverse-genetic approach, the first transgenic zebrafish model of neuroblastoma was developed in Dr. Thomas Look’s laboratory in 2012. Two stable transgenic lines with overexpression of MYCN or ALKF1174L were generated using meganuclease (I-SceI)-mediated transgenic strategy [33]. It is important to note that human genes, rather than zebrafish genes, are commonly chosen for developing a transgenic zebrafish NB model. Doing so allows researchers to avoid the potential issues of targeting transgenes or validating the efficacy of drugs designed to interact with human proteins.

The I-SceI-mediated transgenesis is highly efficient in zebrafish and frogs, although the transgene integration rate seems low in other species [75][76]. The strategy to build transgene constructs for a NB zebrafish model is similar to that of the TH-MYCN mouse model. However, the TH promoter did not work well in zebrafish. Therefore, the dβh promoter was used instead to drive expression of genes of interest in the PSNS [33]. The dβh-EGFP-MYCN construct was used to generate the first zebrafish NB model (designated as MYCN line). Due to the instability of MYCN protein, EGFP-MYCN expression was visible under fluorescence microscopy but was not strong [33]. Hence, researchers applied a strategy of co-injection of dβh-EGFP or dβh-mCherry DNA with dβh promoter-driven transgene constructs containing genes of interest for the subsequent development of multiple transgenic lines.

The co-injected DNA has been demonstrated to be co-integrated into the fish genome, leading to their co-expression in the PSNS cells [33]. The EGFP expression is detectable in the embryos as early as one-day post fertilization [33]. Hence, the EGFP-positive embryos can be easily identified and sorted using fluorescent microscopy, which eliminates additional genotyping procedures after the transgenic line is confirmed. This approach is cost-efficient for conducting large-scale experiments (which require screening hundreds of transgenic fish) at a time when lab space is at a premium.

Using a similar transgenesis approach, researchers have developed a panel of transgenic lines with overexpression of control reporters or genes of interest. These include transgenic fish lines overexpressing EGFP [33] or mCherry [46] alone as a control group, or NB-relevant genes including ALKwt [33], PTPN11 [45], GAB2 [45], LIN28B WT [54], LIN28B_MU [54], LMO1 [46], and DEF [48]. Interestingly, most of the aforementioned genes have been shown to cooperate with MYCN to contribute to the pathogenesis of NB through different cellular mechanisms (Figure 2). However, none of these genes can sufficiently drive NB tumorigenesis when they were overexpressed alone, suggesting that MYCN is a potent oncogene and a key driver for the NB initiation.

Recently, a newer stable transgenic zebrafish line with overexpression of MYCN under control of the dβh promoter was developed in Dr. Thomas Look’s lab [48]. Instead of using the DNA construct carrying a dβh-EGFP-MYCN fusion gene, two transgenic constructs, dβh-MYCN (cDNA+3′UTR) and dβh-EGFP, were coinjected into the one-cell stage of wild-type embryos in developing this line (designated as TgMYCN_TT). The advantage of the coinjection approach has been illustrated above. The major difference of the TgMYCN_TT line to the MYCN line is the inclusion of the 3′ UTR of MYCN gene, containing microRNA recognition sites [77], in the transgene construct. Thus, the MYCN expression in the TgMYCN_TT line could be regulated at the post-transcriptional level, which might link to the increased penetrance of NB to ~70% by 29 wpf [48] in the TgMYCN_TT line as compared to ~30% in the MYCN line by 25 wpf [46]. Moreover, overexpression of c-MYC—a highly expressed MYC-family gene in the MYCN non-amplified high-risk NBs—in the PSNS of transgenic fish could also induce NB tumorigenesis [49], thus implicating the key oncogenic role of MYC family genes in NB pathogenesis.

2.2.3. Genome Editing with Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) and Transcription Activator-Like Effector Nucleases (TALENs)

CRISPR can effectively target genes of interest and achieve up to 75–99% modification rate in zebrafish [78][79]. This high-throughput targeting can generate close to 30% germline mutations [80] with as low as 1% of off-target effects [81]. Shi et al. generated arid1aa and arid1ab knockout zebrafish using this robust technology in the NB model with MYCN overexpression to examine the relevance of ARID1A as a NB suppressor gene in vivo and found that both arid1aa and arid1ab deficiency increases penetration of MYCN-driven NB in zebrafish [55]. Alternatively, targeted deletion in the zebrafish genome can be achieved efficiently using the TALENs system [82]. With this method, a gas7 knockout fish line was generated and bred with the MYCN transgenic line, which led to development of widespread metastasis in the compound fish [56].

2.2.4. Other Potentially Useful Methods

Given that zebrafish modeling for NB research has only become common in the last decade, there are still a plethora of research methods in the zebrafish that have not been applied to the NB study, but that have potential to be used in the near future. For example, the Cre-Lox recombination, a site-specific recombinase technology, has been widely used in different model systems. Briefly, a ubiquitously or tissue-specifically expressed tyrosine recombinase enzyme (Cre) can be used to recombine a pair of short target sequences, called the Lox sequences, leading to manipulation of DNA sequence at specific sites. Such Cre-LoxP conditional expression approach has been used to target the oncogenic Kras/Ras pathway under control of the nestin promoter [83]. Given that many lineage-specific promoters are available in zebrafish, this system is highly applicable when studies require specific activation of gene of interest in a given subset of cell population. For example, Langenau et al. was able to express mouse c-Myc in lymphoblasts of zebrafish using this experimental approach [84]. Heat shock inducible promoters also appear to be applicable which has been used to track individual retina-neuron axon pathways [85]. Additionally, tamoxifen-inducible Cre recombinase can conditionally activate gene expression when needed, such as CreERT2 in zebrafish [86]. The whole system can be easily integrated using CRISPR/Cas9 in zebrafish [87]; a detailed review on the zebrafish Cre-LoxP system has previously been published [88].

2.3. High Throughput Transplantation, Patient-Derived Xenograft (PDX) and In Vivo Drug Screening Using Zebrafish Larvae

To understand disease pathogenesis and screen or validate drug efficacy in vivo, scientists have successfully transplanted tumor cells with different genetic alteration(s) or manipulated gene expression, as well as patient-derived tumor cells into zebrafish at embryonic stage or adulthood [89]. Several features of zebrafish larvae make them uniquely suited for these studies, including: (i) transparent bodies that allow for easy tumor cell injection; (ii) ability to use trackable fluorescent-tagged cells following transplantation; (iii) an immature immune system during early embryonic development, which reduces the chance of the immune rejection of transplanted tumor cells; and (iv) availability of large clutches of embryos for transplantation. Multiple injection sites, such as the perivitelline space, pericardial space, yolk, retro-optical region, and brain, have been explored in a variety of studies to understand the mechanisms of tumor metastasis, angiogenesis, cellular intravasation/extravasation [90][91][92].

Drug screening on zebrafish transplants or xenografts is another exemplary usage of this model. Both embryos and adults can be used in high-throughput drug-screening assays. Embryos are relatively easy to work with due to their permeability of small molecules [93]. Researchers have already performed small-molecule drug screening using zebrafish embryos transplanted with neural crest stem cells (NCSCs) [52]. Since NB is derived from the sympathoadrenal lineage of neural crest cells, small molecules that inhibit NCSC induction might be potentially useful for the NB treatment. Among the 640 FDA-approved drugs applied in this screen, one drug, leflunomide, was identified to inhibit NCSC induction. Leflunomide, as an inhibitor of dihydroorotate dehydrogenase (DHODH) and an immunosuppressive agent for the treatment of patients with rheumatoid arthritis, has already been shown to reduce proliferation and induce apoptosis in NB cells both in vitro and in vivo [94]. Hence, this result further demonstrates the important application of zebrafish as an unbiased in vivo system for effective drug screening. Recently, zebrafish transplanted with human NB cells have been used to demonstrate the effect of a new multi-kinase drug, TP-0903, on reducing extravasation and inducing tumor cell death, suggesting the therapeutic potential of this compound for the NB treatment [51].

Although PDX mouse models are considered the gold standard for the in vivo validation of drug efficacy, the studies led by Drs. Ferreira and Fior, have demonstrated that the patient-derived zebrafish xenografts (zPDX, also called cancer “avatars”) can be used to sense cancer behavior and screen for potential novel therapies. Using a panel of zebrafish xenografts with patient-derived colorectal cancers, Ferreira and Fior rapidly screened the available therapeutic options for the colorectal cancers and predicted the treatment outcomes [92][95], which set the groundwork for using zPDX as a rapid in vivo screening platform for future personalized cancer treatments. Following these efforts, a high-throughput zebrafish xenograft assay of neuroblastoma was performed to confirm cannabinoid receptor 2 (CNR2) and Mitogen-activated protein kinase 8 (MAPK8) as promising candidates for the treatment of high-risk NB and to identify the drugs GW405833 and AS601245 as the most effective and well-tolerated CNR2 and MAPK8 targeted compounds to inhibit the growth of xenografts in zebrafish [96].

To better mimic the cytokine-enriched microenvironment found in human patients for xenotransplantation, Dr. Berman’s group generated the first humanized zebrafish by overexpressing transgenes encoding human hematopoietic-specific cytokines, such as GM-CSF, stem cell factor (SCF), or stromal cell-derived factor 1α (SDF1α). Transgenic lines with overexpression of each of the individual gene mentioned above were developed first using Tol2 transposon-mediated transgenic approach and then incrossed to generate a compound transgenic fish line with overexpression of all of the aforementioned cytokines (GM-CSF, SCF, and SDF1α) (designated GSS fish) [97]. Patient-derived leukemias transplanted into the GSS zebrafish exhibit improved survival, self-renewal ability and broader clonal representation. Therefore, the GSS fish establish a new standard for zebrafish xenotransplantation that more accurately recapitulates the human context for evaluating personalized treatment [97].

References

  1. Stanton, M.F. Diethylnitrosamine-Induced Hepatic Degeneration and Neoplasia in the Aquarium Fish, Brachydanio Rerio. J. Natl. Cancer Inst. 1965, 34, 117–130.
  2. Langenau, D.M.; Traver, D.; Ferrando, A.A.; Kutok, J.L.; Aster, J.C.; Kanki, J.P.; Lin, S.; Prochownik, E.; Trede, N.S.; Zon, L.I.; et al. Myc-induced T cell leukemia in transgenic zebrafish. Science 2003, 299, 887–890.
  3. Etchin, J.; Kanki, J.P.; Look, A.T. Zebrafish as a model for the study of human cancer. Methods Cell Biol. 2011, 105, 309–337.
  4. Feitsma, H.; Cuppen, E. Zebrafish as a cancer model. Mol. Cancer Res. 2008, 6, 685–694.
  5. Benjamin, D.C.; Hynes, R.O. Intravital imaging of metastasis in adult Zebrafish. BMC Cancer 2017, 17, 660.
  6. Kim, I.S.; Heilmann, S.; Kansler, E.R.; Zhang, Y.; Zimmer, M.; Ratnakumar, K.; Bowman, R.L.; Simon-Vermot, T.; Fennell, M.; Garippa, R.; et al. Microenvironment-derived factors driving metastatic plasticity in melanoma. Nat. Commun. 2017, 8, 14343.
  7. Colon, N.C.; Chung, D.H. Neuroblastoma. Adv. Pediatr. 2011, 58, 297–311.
  8. Maris, J.M. Recent advances in neuroblastoma. N. Engl. J. Med. 2010, 362, 2202–2211.
  9. Brodeur, G.M. Neuroblastoma: Biological insights into a clinical enigma. Nat. Rev. Cancer 2003, 3, 203–216.
  10. Pearson, A.D.; Pinkerton, C.R.; Lewis, I.J.; Imeson, J.; Ellershaw, C.; Machin, D. High-dose rapid and standard induction chemotherapy for patients aged over 1 year with stage 4 neuroblastoma: A randomised trial. Lancet Oncol. 2008, 9, 247–256.
  11. Proust-Houdemont, S.; Pasqualini, C.; Blanchard, P.; Dufour, C.; Benhamou, E.; Goma, G.; Semeraro, M.; Raquin, M.A.; Hartmann, O.; Valteau-Couanet, D. Busulfan-melphalan in high-risk neuroblastoma: The 30-year experience of a single institution. Bone Marrow Transplant. 2016, 51, 1076–1081.
  12. Yu, A.L.; Gilman, A.L.; Ozkaynak, M.F.; London, W.B.; Kreissman, S.G.; Chen, H.X.; Smith, M.; Anderson, B.; Villablanca, J.G.; Matthay, K.K.; et al. Anti-GD2 antibody with GM-CSF, interleukin-2, and isotretinoin for neuroblastoma. N. Engl. J. Med. 2010, 363, 1324–1334.
  13. Yu, A.L.; Gilman, A.L.; Ozkaynak, M.F.; Naranjo, A.; Diccianni, M.B.; Gan, J.; Hank, J.A.; Batova, A.; London, W.B.; Tenney, S.C.; et al. Long-term follow-up of a Phase III Study of ch14.18 (Dinutuximab) + Cytokine Immunotherapy in Children with High-risk Neuroblastoma: COG Study ANBL0032. Clin. Cancer Res. 2021.
  14. Basta, N.O.; Halliday, G.C.; Makin, G.; Birch, J.; Feltbower, R.; Bown, N.; Elliott, M.; Moreno, L.; Barone, G.; Pearson, A.D.; et al. Factors associated with recurrence and survival length following relapse in patients with neuroblastoma. Br. J. Cancer 2016, 115, 1048–1057.
  15. London, W.B.; Bagatell, R.; Weigel, B.J.; Fox, E.; Guo, D.; Van Ryn, C.; Naranjo, A.; Park, J.R. Historical time to disease progression and progression-free survival in patients with recurrent/refractory neuroblastoma treated in the modern era on Children’s Oncology Group early-phase trials. Cancer-Am. Cancer Soc. 2017, 123, 4914–4923.
  16. Schwab, M.; Alitalo, K.; Klempnauer, K.H.; Varmus, H.E.; Bishop, J.M.; Gilbert, F.; Brodeur, G.; Goldstein, M.; Trent, J. Amplified DNA with limited homology to myc cellular oncogene is shared by human neuroblastoma cell lines and a neuroblastoma tumour. Nature 1983, 305, 245–248.
  17. Huang, M.; Weiss, W.A. Neuroblastoma and MYCN. Cold Spring Harb. Perspect. Med. 2013, 3, a014415.
  18. Brodeur, G.M.; Seeger, R.C.; Schwab, M.; Varmus, H.E.; Bishop, J.M. Amplification of N-myc in untreated human neuroblastomas correlates with advanced disease stage. Science 1984, 224, 1121–1124.
  19. Seeger, R.C.; Brodeur, G.M.; Sather, H.; Dalton, A.; Siegel, S.E.; Wong, K.Y.; Hammond, D. Association of multiple copies of the N-myc oncogene with rapid progression of neuroblastomas. N. Engl. J. Med. 1985, 313, 1111–1116.
  20. Schwab, M.; Varmus, H.E.; Bishop, J.M. Human N-myc gene contributes to neoplastic transformation of mammalian cells in culture. Nature 1985, 316, 160–162.
  21. Land, H.; Parada, L.F.; Weinberg, R.A. Tumorigenic conversion of primary embryo fibroblasts requires at least two cooperating oncogenes. Nature 1983, 304, 596–602.
  22. Weiss, W.A.; Aldape, K.; Mohapatra, G.; Feuerstein, B.G.; Bishop, J.M. Targeted expression of MYCN causes neuroblastoma in transgenic mice. EMBO J. 1997, 16, 2985–2995.
  23. Berry, T.; Luther, W.; Bhatnagar, N.; Jamin, Y.; Poon, E.; Sanda, T.; Pei, D.; Sharma, B.; Vetharoy, W.R.; Hallsworth, A.; et al. The ALK(F1174L) mutation potentiates the oncogenic activity of MYCN in neuroblastoma. Cancer Cell 2012, 22, 117–130.
  24. Heukamp, L.C.; Thor, T.; Schramm, A.; De Preter, K.; Kumps, C.; De Wilde, B.; Odersky, A.; Peifer, M.; Lindner, S.; Spruessel, A.; et al. Targeted expression of mutated ALK induces neuroblastoma in transgenic mice. Sci. Transl. Med. 2012, 4, 141ra191.
  25. Cazes, A.; Lopez-Delisle, L.; Tsarovina, K.; Pierre-Eugene, C.; De Preter, K.; Peuchmaur, M.; Nicolas, A.; Provost, C.; Louis-Brennetot, C.; Daveau, R.; et al. Activated Alk triggers prolonged neurogenesis and Ret upregulation providing a therapeutic target in ALK-mutated neuroblastoma. Oncotarget 2014, 5, 2688–2702.
  26. Hennchen, M.; Stubbusch, J.; Abarchan-El Makhfi, I.; Kramer, M.; Deller, T.; Pierre-Eugene, C.; Janoueix-Lerosey, I.; Delattre, O.; Ernsberger, U.; Schulte, J.B.; et al. Lin28B and Let-7 in the Control of Sympathetic Neurogenesis and Neuroblastoma Development. J. Neurosci. 2015, 35, 16531–16544.
  27. Molenaar, J.J.; Domingo-Fernandez, R.; Ebus, M.E.; Lindner, S.; Koster, J.; Drabek, K.; Mestdagh, P.; van Sluis, P.; Valentijn, L.J.; van Nes, J.; et al. LIN28B induces neuroblastoma and enhances MYCN levels via let-7 suppression. Nat. Genet. 2012, 44, 1199–1206.
  28. Servenius, B.; Vernachio, J.; Price, J.; Andersson, L.C.; Peterson, P.A. Metastasizing neuroblastomas in mice transgenic for simian virus 40 large T (SV40T) under the olfactory marker protein gene promoter. Cancer Res. 1994, 54, 5198–5205.
  29. Iwakura, H.; Ariyasu, H.; Kanamoto, N.; Hosoda, K.; Nakao, K.; Kangawa, K.; Akamizu, T. Establishment of a novel neuroblastoma mouse model. Int. J. Oncol. 2008, 33, 1195–1199.
  30. Hattori, Y.; Kanamoto, N.; Kawano, K.; Iwakura, H.; Sone, M.; Miura, M.; Yasoda, A.; Tamura, N.; Arai, H.; Akamizu, T.; et al. Molecular characterization of tumors from a transgenic mouse adrenal tumor model: Comparison with human pheochromocytoma. Int. J. Oncol. 2010, 37, 695–705.
  31. Kamili, A.; Atkinson, C.; Trahair, T.N.; Fletcher, J.I. Mouse models of high-risk neuroblastoma. Cancer Metastasis Rev. 2020, 39, 261–274.
  32. Ornell, K.J.; Coburn, J.M. Developing preclinical models of neuroblastoma: Driving therapeutic testing. BMC Biomed. Eng. 2019, 1.
  33. Zhu, S.; Lee, J.S.; Guo, F.; Shin, J.; Perez-Atayde, A.R.; Kutok, J.L.; Rodig, S.J.; Neuberg, D.S.; Helman, D.; Feng, H.; et al. Activated ALK collaborates with MYCN in neuroblastoma pathogenesis. Cancer Cell 2012, 21, 362–373.
  34. Mosse, Y.P.; Laudenslager, M.; Longo, L.; Cole, K.A.; Wood, A.; Attiyeh, E.F.; Laquaglia, M.J.; Sennett, R.; Lynch, J.E.; Perri, P.; et al. Identification of ALK as a major familial neuroblastoma predisposition gene. Nature 2008, 455, 930–935.
  35. Chen, Y.; Takita, J.; Choi, Y.L.; Kato, M.; Ohira, M.; Sanada, M.; Wang, L.; Soda, M.; Kikuchi, A.; Igarashi, T.; et al. Oncogenic mutations of ALK kinase in neuroblastoma. Nature 2008, 455, 971–974.
  36. Janoueix-Lerosey, I.; Lequin, D.; Brugieres, L.; Ribeiro, A.; de Pontual, L.; Combaret, V.; Raynal, V.; Puisieux, A.; Schleiermacher, G.; Pierron, G.; et al. Somatic and germline activating mutations of the ALK kinase receptor in neuroblastoma. Nature 2008, 455, 967–970.
  37. George, R.E.; Sanda, T.; Hanna, M.; Frohling, S.; Luther, W., 2nd; Zhang, J.; Ahn, Y.; Zhou, W.; London, W.B.; McGrady, P.; et al. Activating mutations in ALK provide a therapeutic target in neuroblastoma. Nature 2008, 455, 975–978.
  38. Carpenter, E.L.; Mosse, Y.P. Targeting ALK in neuroblastoma—Preclinical and clinical advancements. Nat. Rev. Clin. Oncol. 2012, 9, 391–399.
  39. Umapathy, G.; Mendoza-Garcia, P.; Hallberg, B.; Palmer, R.H. Targeting anaplastic lymphoma kinase in neuroblastoma. APMIS 2019, 127, 288–302.
  40. Amsterdam, A.; Lai, K.; Komisarczuk, A.Z.; Becker, T.S.; Bronson, R.T.; Hopkins, N.; Lees, J.A. Zebrafish Hagoromo mutants up-regulate fgf8 postembryonically and develop neuroblastoma. Mol. Cancer Res. 2009, 7, 841–850.
  41. Pei, D.; Luther, W.; Wang, W.; Paw, B.H.; Stewart, R.A.; George, R.E. Distinct neuroblastoma-associated alterations of PHOX2B impair sympathetic neuronal differentiation in zebrafish models. PLoS Genet. 2013, 9, e1003533.
  42. Brodeur, G.M.; Hogarty, M.; Bagatell, R.; Mosse, Y.P.; Maris, J.M. Neuroblastoma. In Principles and Practice of Pediatric Oncology, 7th ed.; Pizzo, P.A., Poplack, D., Eds.; Lippincott Williams & Wilkins: Philadelphia, PA, USA, 2016; pp. 772–792.
  43. Pinto, N.R.; Applebaum, M.A.; Volchenboum, S.L.; Matthay, K.K.; London, W.B.; Ambros, P.F.; Nakagawara, A.; Berthold, F.; Schleiermacher, G.; Park, J.R.; et al. Advances in Risk Classification and Treatment Strategies for Neuroblastoma. J. Clin. Oncol. 2015, 33, 3008–3017.
  44. He, S.; Mansour, M.R.; Zimmerman, M.W.; Ki, D.H.; Layden, H.M.; Akahane, K.; Gjini, E.; de Groh, E.D.; Perez-Atayde, A.R.; Zhu, S.; et al. Synergy between loss of NF1 and overexpression of MYCN in neuroblastoma is mediated by the GAP-related domain. Elife 2016, 5.
  45. Zhang, X.; Dong, Z.; Zhang, C.; Ung, C.Y.; He, S.; Tao, T.; Oliveira, A.M.; Meves, A.; Ji, B.; Look, A.T.; et al. Critical Role for GAB2 in Neuroblastoma Pathogenesis through the Promotion of SHP2/MYCN Cooperation. Cell Rep. 2017, 18, 2932–2942.
  46. Zhu, S.; Zhang, X.; Weichert-Leahey, N.; Dong, Z.; Zhang, C.; Lopez, G.; Tao, T.; He, S.; Wood, A.C.; Oldridge, D.; et al. LMO1 Synergizes with MYCN to Promote Neuroblastoma Initiation and Metastasis. Cancer Cell 2017, 32, 310–323.e315.
  47. Radic-Sarikas, B.; Halasz, M.; Huber, K.V.M.; Winter, G.E.; Tsafou, K.P.; Papamarkou, T.; Brunak, S.; Kolch, W.; Superti-Furga, G. Lapatinib potentiates cytotoxicity of YM155 in neuroblastoma via inhibition of the ABCB1 efflux transporter. Sci. Rep. 2017, 7, 3091.
  48. Tao, T.; Sondalle, S.B.; Shi, H.; Zhu, S.; Perez-Atayde, A.R.; Peng, J.; Baserga, S.J.; Look, A.T. The pre-rRNA processing factor DEF is rate limiting for the pathogenesis of MYCN-driven neuroblastoma. Oncogene 2017, 36, 3852–3867.
  49. Zimmerman, M.W.; Liu, Y.; He, S.; Durbin, A.D.; Abraham, B.J.; Easton, J.; Shao, Y.; Xu, B.; Zhu, S.; Zhang, X.; et al. MYC Drives a Subset of High-Risk Pediatric Neuroblastomas and Is Activated through Mechanisms Including Enhancer Hijacking and Focal Enhancer Amplification. Cancer Discov. 2018, 8, 320–335.
  50. Shen, J.; Najafi, S.; Stable, S.; Fabian, J.; Koeneke, E.; Kolbinger, F.R.; Wrobel, J.K.; Meder, B.; Distel, M.; Heimburg, T.; et al. A kinome-wide RNAi screen identifies ALK as a target to sensitize neuroblastoma cells for HDAC8-inhibitor treatment. Cell Death Differ. 2018, 25, 2053–2070.
  51. Aveic, S.; Corallo, D.; Porcu, E.; Pantile, M.; Boso, D.; Zanon, C.; Viola, G.; Sidarovich, V.; Mariotto, E.; Quattrone, A.; et al. TP-0903 inhibits neuroblastoma cell growth and enhances the sensitivity to conventional chemotherapy. Eur. J. Pharmacol. 2018, 818, 435–448.
  52. Seda, M.; Geerlings, M.; Lim, P.; Jeyabalan-Srikaran, J.; Cichon, A.C.; Scambler, P.J.; Beales, P.L.; Hernandez-Hernandez, V.; Stoker, A.W.; Jenkins, D. An FDA-Approved Drug Screen for Compounds Influencing Craniofacial Skeletal Development and Craniosynostosis. Mol. Syndromol. 2019, 10, 98–114.
  53. Koach, J.; Holien, J.K.; Massudi, H.; Carter, D.R.; Ciampa, O.C.; Herath, M.; Lim, T.; Seneviratne, J.A.; Milazzo, G.; Murray, J.E.; et al. Drugging MYCN Oncogenic Signaling through the MYCN-PA2G4 Binding Interface. Cancer Res. 2019, 79, 5652–5667.
  54. Tao, T.; Shi, H.; Mariani, L.; Abraham, B.J.; Durbin, A.D.; Zimmerman, M.W.; Powers, J.T.; Missios, P.; Ross, K.N.; Perez-Atayde, A.R.; et al. LIN28B regulates transcription and potentiates MYCN-induced neuroblastoma through binding to ZNF143 at target gene promotors. Proc. Natl. Acad. Sci. USA 2020, 117, 16516–16526.
  55. Shi, H.; Tao, T.; Abraham, B.J.; Durbin, A.D.; Zimmerman, M.W.; Kadoch, C.; Look, A.T. ARID1A loss in neuroblastoma promotes the adrenergic-to-mesenchymal transition by regulating enhancer-mediated gene expression. Sci. Adv. 2020, 6, eaaz3440.
  56. Dong, Z.; Yeo, K.S.; Lopez, G.; Zhang, C.; Dankert Eggum, E.N.; Rokita, J.L.; Ung, C.Y.; Levee, T.M.; Her, Z.P.; Howe, C.J.; et al. GAS7 Deficiency Promotes Metastasis in MYCN-driven Neuroblastoma. Cancer Res. 2021.
  57. Corallo, D.; Candiani, S.; Ori, M.; Aveic, S.; Tonini, G.P. The zebrafish as a model for studying neuroblastoma. Cancer Cell Int. 2016, 16, 82.
  58. Casey, M.J.; Stewart, R.A. Zebrafish as a model to study neuroblastoma development. Cell Tissue Res. 2018, 372, 223–232.
  59. Quarta, C.; Cantelli, E.; Nanni, C.; Ambrosini, V.; D’Ambrosio, D.; Di Leo, K.; Angelucci, S.; Zagni, F.; Lodi, F.; Marengo, M.; et al. Molecular imaging of neuroblastoma progression in TH-MYCN transgenic mice. Mol. Imaging Biol. 2013, 15, 194–202.
  60. Almeida, G.S.; Panek, R.; Hallsworth, A.; Webber, H.; Papaevangelou, E.; Boult, J.K.; Jamin, Y.; Chesler, L.; Robinson, S.P. Pre-clinical imaging of transgenic mouse models of neuroblastoma using a dedicated 3-element solenoid coil on a clinical 3T platform. Br. J. Cancer 2017, 117, 791–800.
  61. Teitelman, G.; Baker, H.; Joh, T.H.; Reis, D.J. Appearance of catecholamine-synthesizing enzymes during development of rat sympathetic nervous system: Possible role of tissue environment. Proc. Natl. Acad. Sci. USA 1979, 76, 509–513.
  62. Marusich, M.F.; Furneaux, H.M.; Henion, P.D.; Weston, J.A. Hu neuronal proteins are expressed in proliferating neurogenic cells. J. Neurobiol. 1994, 25, 143–155.
  63. Dong, R.; Yang, R.; Zhan, Y.; Lai, H.D.; Ye, C.J.; Yao, X.Y.; Luo, W.Q.; Cheng, X.M.; Miao, J.J.; Wang, J.F.; et al. Single-Cell Characterization of Malignant Phenotypes and Developmental Trajectories of Adrenal Neuroblastoma. Cancer Cell 2020, 38, 716–733.e716.
  64. Hsu, H.J.; Lin, G.; Chung, B.C. Parallel early development of zebrafish interrenal glands and pronephros: Differential control by wt1 and ff1b. Development 2003, 130, 2107–2116.
  65. Renshaw, S.A.; Trede, N.S. A model 450 million years in the making: Zebrafish and vertebrate immunity. Dis. Model. Mech. 2012, 5, 38–47.
  66. Lieschke, G.J.; Trede, N.S. Fish immunology. Curr. Biol. 2009, 19, R678–R682.
  67. Menke, A.L.; Spitsbergen, J.M.; Wolterbeek, A.P.; Woutersen, R.A. Normal anatomy and histology of the adult zebrafish. Toxicol. Pathol. 2011, 39, 759–775.
  68. Progatzky, F.; Cook, H.T.; Lamb, J.R.; Bugeon, L.; Dallman, M.J. Mucosal inflammation at the respiratory interface: A zebrafish model. Am. J. Physiol. Lung Cell. Mol. Physiol. 2016, 310, L551–L561.
  69. DuBois, S.G.; Kalika, Y.; Lukens, J.N.; Brodeur, G.M.; Seeger, R.C.; Atkinson, J.B.; Haase, G.M.; Black, C.T.; Perez, C.; Shimada, H.; et al. Metastatic sites in stage IV and IVS neuroblastoma correlate with age, tumor biology, and survival. J. Pediatr. Hematol. Oncol. 1999, 21, 181–189.
  70. Levental, K.R.; Yu, H.; Kass, L.; Lakins, J.N.; Egeblad, M.; Erler, J.T.; Fong, S.F.; Csiszar, K.; Giaccia, A.; Weninger, W.; et al. Matrix crosslinking forces tumor progression by enhancing integrin signaling. Cell 2009, 139, 891–906.
  71. Paszek, M.J.; Zahir, N.; Johnson, K.R.; Lakins, J.N.; Rozenberg, G.I.; Gefen, A.; Reinhart-King, C.A.; Margulies, S.S.; Dembo, M.; Boettiger, D.; et al. Tensional homeostasis and the malignant phenotype. Cancer Cell 2005, 8, 241–254.
  72. Luo, C.; Zuniga, J.; Edison, E.; Palla, S.; Dong, W.; Parker-Thornburg, J. Superovulation strategies for 6 commonly used mouse strains. J. Am. Assoc. Lab. Anim. Sci. 2011, 50, 471–478.
  73. Gaiano, N.; Amsterdam, A.; Kawakami, K.; Allende, M.; Becker, T.; Hopkins, N. Insertional mutagenesis and rapid cloning of essential genes in zebrafish. Nature 1996, 383, 829–832.
  74. Allende, M.L.; Amsterdam, A.; Becker, T.; Kawakami, K.; Gaiano, N.; Hopkins, N. Insertional mutagenesis in zebrafish identifies two novel genes, pescadillo and dead eye, essential for embryonic development. Genes Dev. 1996, 10, 3141–3155.
  75. Thermes, V.; Grabher, C.; Ristoratore, F.; Bourrat, F.; Choulika, A.; Wittbrodt, J.; Joly, J.S. I-SceI meganuclease mediates highly efficient transgenesis in fish. Mech. Dev. 2002, 118, 91–98.
  76. Pan, F.C.; Chen, Y.; Loeber, J.; Henningfeld, K.; Pieler, T. I-SceI meganuclease-mediated transgenesis in Xenopus. Dev. Dyn. 2006, 235, 247–252.
  77. Powers, J.T.; Tsanov, K.M.; Pearson, D.S.; Roels, F.; Spina, C.S.; Ebright, R.; Seligson, M.; de Soysa, Y.; Cahan, P.; Theissen, J.; et al. Multiple mechanisms disrupt the let-7 microRNA family in neuroblastoma. Nature 2016, 535, 246–251.
  78. Hwang, W.Y.; Fu, Y.; Reyon, D.; Maeder, M.L.; Tsai, S.Q.; Sander, J.D.; Peterson, R.T.; Yeh, J.R.; Joung, J.K. Efficient genome editing in zebrafish using a CRISPR-Cas system. Nat. Biotechnol. 2013, 31, 227–229.
  79. Jao, L.E.; Wente, S.R.; Chen, W. Efficient multiplex biallelic zebrafish genome editing using a CRISPR nuclease system. Proc. Natl. Acad. Sci. USA 2013, 110, 13904–13909.
  80. Varshney, G.K.; Pei, W.; LaFave, M.C.; Idol, J.; Xu, L.; Gallardo, V.; Carrington, B.; Bishop, K.; Jones, M.; Li, M.; et al. High-throughput gene targeting and phenotyping in zebrafish using CRISPR/Cas9. Genome Res. 2015, 25, 1030–1042.
  81. Hruscha, A.; Krawitz, P.; Rechenberg, A.; Heinrich, V.; Hecht, J.; Haass, C.; Schmid, B. Efficient CRISPR/Cas9 genome editing with low off-target effects in zebrafish. Development 2013, 140, 4982–4987.
  82. Bedell, V.M.; Wang, Y.; Campbell, J.M.; Poshusta, T.L.; Starker, C.G.; Krug, R.G., 2nd; Tan, W.; Penheiter, S.G.; Ma, A.C.; Leung, A.Y.; et al. In vivo genome editing using a high-efficiency TALEN system. Nature 2012, 491, 114–118.
  83. Seok, S.H.; Na, Y.R.; Han, J.H.; Kim, T.H.; Jung, H.; Lee, B.H.; Emelyanov, A.; Parinov, S.; Park, J.H. Cre/loxP-regulated transgenic zebrafish model for neural progenitor-specific oncogenic Kras expression. Cancer Sci. 2010, 101, 149–154.
  84. Langenau, D.M.; Feng, H.; Berghmans, S.; Kanki, J.P.; Kutok, J.L.; Look, A.T. Cre/lox-regulated transgenic zebrafish model with conditional myc-induced T cell acute lymphoblastic leukemia. Proc. Natl. Acad. Sci. USA 2005, 102, 6068–6073.
  85. Robles, E.; Filosa, A.; Baier, H. Precise lamination of retinal axons generates multiple parallel input pathways in the tectum. J. Neurosci. 2013, 33, 5027–5039.
  86. Felker, A.; Mosimann, C. Contemporary zebrafish transgenesis with Tol2 and application for Cre/lox recombination experiments. Methods Cell Biol. 2016, 135, 219–244.
  87. Burg, L.; Palmer, N.; Kikhi, K.; Miroshnik, E.S.; Rueckert, H.; Gaddy, E.; MacPherson Cunningham, C.; Mattonet, K.; Lai, S.L.; Marin-Juez, R.; et al. Conditional mutagenesis by oligonucleotide-mediated integration of loxP sites in zebrafish. PLoS Genet. 2018, 14, e1007754.
  88. Carney, T.J.; Mosimann, C. Switch and Trace: Recombinase Genetics in Zebrafish. Trends Genet. 2018, 34, 362–378.
  89. Veinotte, C.J.; Dellaire, G.; Berman, J.N. Hooking the big one: The potential of zebrafish xenotransplantation to reform cancer drug screening in the genomic era. Dis. Model. Mech. 2014, 7, 745–754.
  90. Haney, M.G.; Moore, L.H.; Blackburn, J.S. Drug Screening of Primary Patient Derived Tumor Xenografts in Zebrafish. J. Vis. Exp. 2020, e60996.
  91. Cabezas-Sainz, P.; Pensado-Lopez, A.; Sainz, B., Jr.; Sanchez, L. Modeling Cancer Using Zebrafish Xenografts: Drawbacks for Mimicking the Human Microenvironment. Cells 2020, 9, 1978.
  92. Costa, B.; Estrada, M.F.; Mendes, R.V.; Fior, R. Zebrafish Avatars towards Personalized Medicine-A Comparative Review between Avatar Models. Cells 2020, 9, 293.
  93. Pichler, F.B.; Laurenson, S.; Williams, L.C.; Dodd, A.; Copp, B.R.; Love, D.R. Chemical discovery and global gene expression analysis in zebrafish. Nat. Biotechnol. 2003, 21, 879–883.
  94. Zhu, S.; Yan, X.; Xiang, Z.; Ding, H.F.; Cui, H. Leflunomide reduces proliferation and induces apoptosis in neuroblastoma cells in vitro and in vivo. PLoS ONE 2013, 8, e71555.
  95. Fior, R.; Povoa, V.; Mendes, R.V.; Carvalho, T.; Gomes, A.; Figueiredo, N.; Ferreira, M.G. Single-cell functional and chemosensitive profiling of combinatorial colorectal therapy in zebrafish xenografts. Proc. Natl. Acad. Sci. USA 2017, 114, E8234–E8243.
  96. Almstedt, E.; Elgendy, R.; Hekmati, N.; Rosen, E.; Warn, C.; Olsen, T.K.; Dyberg, C.; Doroszko, M.; Larsson, I.; Sundstrom, A.; et al. Integrative discovery of treatments for high-risk neuroblastoma. Nat. Commun. 2020, 11, 71.
  97. Rajan, V.; Melong, N.; Hing Wong, W.; King, B.; Tong, S.R.; Mahajan, N.; Gaston, D.; Lund, T.; Rittenberg, D.; Dellaire, G.; et al. Humanized zebrafish enhance human hematopoietic stem cell survival and promote acute myeloid leukemia clonal diversity. Haematologica 2020, 105, 2391–2399.
More
Information
Subjects: Cell Biology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : ,
View Times: 821
Revisions: 4 times (View History)
Update Date: 18 Mar 2021
1000/1000