Next Article in Journal
Ursolic Acid-Based Derivatives as Potential Anti-Cancer Agents: An Update
Next Article in Special Issue
Cell Fitness: More Than Push-Ups
Previous Article in Journal
PD-L1 in Systemic Immunity: Unraveling Its Contribution to PD-1/PD-L1 Blockade Immunotherapy
Previous Article in Special Issue
Rounding up the Usual Suspects: Assessing Yorkie, AP-1, and Stat Coactivation in Tumorigenesis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Basic and Translational Models of Cooperative Oncogenesis

1
Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
2
Wolfson Wohl Cancer Research Centre, Institute of Cancer Science, University of Glasgow, Glasgow G61 1QH, UK
3
Department of Life, Health and Environmental Sciences, University of L’Aquila, I-67100 L’Aquila, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2020, 21(16), 5919; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21165919
Submission received: 12 August 2020 / Accepted: 14 August 2020 / Published: 18 August 2020
(This article belongs to the Special Issue Basic and Translational Models of Cooperative Oncogenesis)
Cancer is a complex set of diseases involving genetic or epigenetic changes within cells, as well as interactions between the developing tumour cells and their microenvironment, which leads to uncontrolled tumour growth, altered differentiation, local invasion and metastasis to distant sites [1,2]. Molecular changes in oncogenes or tumour suppressor genes promote various cancer hallmarks, including the continued proliferation, inhibition of differentiation, inhibition of apoptosis, changes to metabolism, evasion of the immune system and the promotion of invasion/metastasis [3,4]. Although many oncogenes and tumour suppressor gene mutations promote more than one hallmark of cancer, several mutations are required to generate malignant cancers, a process referred to as cooperative oncogenesis/tumourigenesis [5,6]. Through decades of research, we have gained much knowledge on key molecular events and processes involved in the formation of cancer, and much of this knowledge has stemmed from investigations using model organisms, such as the mouse and the vinegar fly, Drosophila melanogaster, in addition to in vitro cell line studies. In this Special Issue, we present a collection of original research papers on various aspects of cancer research utilising human cell lines [7,8], or in vivo using Drosophila as a model system [9,10], as well as reviews highlighting the Drosophila model organism in cancer research [11,12,13,14,15]. Drosophila is a particularly useful model organism for the study of cancer mechanisms, because it has a rapid life cycle and is genetically manipulatable and since cancer genes and signalling pathways are highly conserved between humans and Drosophila, and interactions between tumour cells and surrounding normal cells can be readily examined in Drosophila tissues [6,16,17,18,19,20].
In a research paper pertaining to in vitro models of cancer, Di Giorgio et al. [8] analyse the transcriptional response to the expression of three key oncogenes (RAS, MYC, and HDAC4) in human fibroblasts, revealing common signalling pathways that are deregulated by these genes, and suggesting potential therapeutic avenues for the treatment of cancers driven by these oncogenes. In the second research paper on this topic, Mayer et al. [7] focus on human pancreatic cancer, where they observe in tissue sections the infiltration of Th17-like T cells expressing IL21 and IL26, and the expression of receptors for IL21 and IL26 in the pancreatic epithelial cells. They show in human pancreatic cell lines that IL21 and IL26 signal through ERK1/2 and STAT, which leads to increased tumour cell growth in colony forming assays.
In a research paper utilising the Drosophila model, Parniewska and Stocker [9] identify the novel splicing factor, SF2, which is essential for the survival and hyperproliferation of tissues that upregulate the target of rapamycin complex 1 (TORC1), a protein kinase involved in cellular growth that is upregulated in many cancers [21,22]. The identification of SF2 as a key conserved target of TORC1, which is required for early tumour growth in Drosophila, provides a potential new approach to develop anti-cancer therapies for tumours with upregulated TORC1 activity, such as those carrying loss of function of PTEN (phosphatase and tensin homolog deleted on chromosome ten) or constitutively active mutations in PI3K (phospho inositol 3 kinase) [23,24,25]. In a second research paper utilising the Drosophila model, Hamaratoglu and Atkins [10] undertook an analysis of published transcriptional data from various Drosophila imaginal disc epithelial cell models of cancer, and found that the JNK stress response pathway [26,27], and JAK/STAT [28,29], Hippo [30,31] and Notch [32,33] tissue growth signalling pathways are commonly deregulated. This important meta-analysis has opened-up new potential avenues of research to examine the cooperative interactions between these signalling pathways using model organisms, as well as to assess the co-dependency of these conserved pathways in human cancers.
The reviews in this Special Issue highlight the power of using Drosophila as an in vivo model system to study various aspects of cancer research, from its application in the study of the function of specific genes/pathways in cancer [11,15], to understanding particular cellular processes in cancer [13,14], and for functional analyses of cancer Omics data [12]. Sechi et al. [15] review the mechanisms of the Golgi phosphoprotein 3 (GOLPH3) oncogene in cancer, covering research on human cancer samples, in vitro cell line analyses and Drosophila in vivo analyses. Carmena [11] reviews the involvement of the Scribble cell polarity module [34] in asymmetric cell division (ACD) of Drosophila neural stem cells in tumourigenesis, highlighting the importance of correct ACD for appropriate differentiation and exit from the cell cycle. Casas-Tintó and Portela [14] review the involvement of specialised cellular extensions, termed cytonemes, in cell–cell communication and in tumourigenesis. Cytonemes have recently been shown to be highly important in the development of glioblastoma and the associated neural degeneration that occurs in Drosophila models and also in the human disease [35], and there is accumulating evidence for their role in tumourigenesis in other cancer types. Newman and Gregory [13] review the connection between chromosomal aberrations (aneuploidy) and metabolic changes, highlighting the role of oxidative stress and particularly reactive oxygen species (ROS) in this process. Finally, Bangi [12] reviews how Drosophila can be utilised to functionally analyse the vast amount of human cancer Omics data that is currently being generated, in order to validate key genes/pathways that contribute to cancer, to build new models to interrogate cancer mechanisms, and to screen for novel cancer therapeutics. Drosophila has already proven its worth in identifying novel drugs that target particular types of human cancers, such as multiple endocrine neoplasia type 2 (MEN2), colorectal and non-small cell lung cancers [36,37,38,39,40,41,42], and undoubtably, the development of more sophisticated Drosophila models that incorporate additional genetic lesions will enable better modelling of human cancers and new anti-cancer drug discovery.
This first iteration of this Special Issue on basic and translational models of cooperative oncogenesis presents only a snapshot of the vast amount of research into cancer currently being conducted worldwide, yet it highlights the important contribution of the simple multicellular model organism, Drosophila, to our current understanding of cancer. Undoubtably, further primary research papers and literature reviews for future iterations of this Special Issue will highlight new cancer genes/pathways and processes involved in cancer, additional in vivo models of cancer (such as worms, zebra fish, and mice), and novel approaches for the understanding of cancer mechanisms and for developing new cancer therapies.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Radisky, D.; Hagios, C.; Bissell, M.J. Tumors are unique organs defined by abnormal signaling and context. Semin. Cancer Biol. 2001, 11, 87–95. [Google Scholar] [CrossRef] [PubMed]
  2. Bissell, M.J.; Radisky, D. Putting tumours in context. Nat. Rev. Cancer 2001, 1, 46–54. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Hanahan, D.; Weinberg, R.A. The hallmarks of cancer. Cell 2000, 100, 57–70. [Google Scholar] [CrossRef] [Green Version]
  4. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
  5. Delaval, B.; Birnbaum, D. A cell cycle hypothesis of cooperative oncogenesis (Review). Int. J. Oncol. 2007, 30, 1051–1058. [Google Scholar] [CrossRef]
  6. Richardson, H.E.; Portela, M. Modelling Cooperative Tumorigenesis in Drosophila. Biomed. Res. Int. 2018, 2018, 4258387. [Google Scholar] [CrossRef] [Green Version]
  7. Mayer, P.; Linnebacher, A.; Glennemeier-Marke, H.; Marnet, N.; Bergmann, F.; Hackert, T.; Klauss, M.; Poth, T.; Gaida, M.M. The Microarchitecture of Pancreatic Cancer as Measured by Diffusion-Weighted Magnetic Resonance Imaging is Altered by T Cells with a Tumor Promoting Th17 Phenotype. Int. J. Mol. Sci. 2020, 21, 346. [Google Scholar] [CrossRef] [Green Version]
  8. Di Giorgio, E.; Paluvai, H.; Picco, R.; Brancolini, C. Genetic Programs Driving Oncogenic Transformation: Lessons from in Vitro Models. Int. J. Mol. Sci. 2020, 20, 6283. [Google Scholar] [CrossRef] [Green Version]
  9. Parniewska, M.M.; Stocker, H. The Splicing Factor SF2 is Critical for Hyperproliferation and Survival in a TORC1-Dependent Model of Early Tumorigenesis in Drosophila. Int. J. Mol. Sci. 2019, 21, 4465. [Google Scholar] [CrossRef]
  10. Hamaratoglu, F.; Atkins, M. Rounding up the Usual Suspects: Assessing Yorkie, AP-1, and Stat Coactivation in Tumorigenesis. Int. J. Mol. Sci. 2020, 21, 4580. [Google Scholar] [CrossRef]
  11. Carmena, A. The Case of the Scribble Polarity Module in Asymmetric Neuroblast Division in Development and Tumorigenesis. Int. J. Mol. Sci. 2020, 21, 2865. [Google Scholar] [CrossRef] [Green Version]
  12. Bangi, E. Strategies for Functional Interrogation of Big Cancer Data Using Drosophila Cancer Models. Int. J. Mol. Sci. 2020, 21, 3754. [Google Scholar] [CrossRef]
  13. Newman, D.L.; Gregory, S.L. Co-Operation between Aneuploidy and Metabolic Changes in Driving Tumorigenesis. Int. J. Mol. Sci. 2020, 20, 4611. [Google Scholar] [CrossRef] [Green Version]
  14. Casas-Tintó, S.; Portela, M. Cytonemes, Their Formation, Regulation, and Roles in Signaling and Communication in Tumorigenesis. Int. J. Mol. Sci. 2019, 20, 5641. [Google Scholar] [CrossRef] [Green Version]
  15. Sechi, S.; Frappaolo, A.; Karimpour-Ghahnavieh, A.; Piergentili, R.; Giansanti, M.G. Oncogenic Roles of GOLPH3 in the Physiopathology of Cancer. Int. J. Mol. Sci. 2019, 21, 933. [Google Scholar] [CrossRef] [Green Version]
  16. Rudrapatna, V.A.; Cagan, R.L.; Das, T.K. Drosophila cancer models. Dev. Dyn. 2012, 241, 107–118. [Google Scholar] [CrossRef] [Green Version]
  17. Sonoshita, M.; Cagan, R.L. Modeling Human Cancers in Drosophila. Curr. Top. Dev. Biol. 2017, 121, 287–309. [Google Scholar]
  18. Gonzalez, C. Drosophila melanogaster: A model and a tool to investigate malignancy and identify new therapeutics. Nat. Rev. Cancer 2013, 13, 172–183. [Google Scholar] [CrossRef]
  19. Tipping, M.; Perrimon, N. Drosophila as a model for context-dependent tumorigenesis. J. Cell. Physiol. 2014, 229, 27–33. [Google Scholar]
  20. Brumby, A.M.; Richardson, H.E. Using Drosophila melanogaster to map human cancer pathways. Nat. Rev. Cancer 2005, 5, 626–639. [Google Scholar] [CrossRef]
  21. Zou, Z.; Tao, T.; Li, H.; Zhu, X. mTOR signaling pathway and mTOR inhibitors in cancer: Progress and challenges. Cell Biosci. 2020, 10, 31. [Google Scholar] [CrossRef]
  22. Liu, G.Y.; Sabatini, D.M. mTOR at the nexus of nutrition, growth, ageing and disease. Nat. Rev. Mol. Cell Biol. 2020, 21, 183–203. [Google Scholar] [CrossRef]
  23. Parsons, R. Discovery of the PTEN Tumor Suppressor and Its Connection to the PI3K and AKT Oncogenes. Cold Spring Harb. Perspect. Med. 2020, 10, a036129. [Google Scholar] [CrossRef]
  24. Ngeow, J.; Eng, C. PTEN in Hereditary and Sporadic Cancer. Cold Spring Harb. Perspect. Med. 2020, 10, a036087. [Google Scholar] [CrossRef] [Green Version]
  25. Noorolyai, S.; Shajari, N.; Baghbani, E.; Sadreddini, S.; Baradaran, B. The relation between PI3K/AKT signalling pathway and cancer. Gene 2019, 698, 120–128. [Google Scholar] [CrossRef]
  26. La Marca, J.E.; Richardson, H.E. Two-Faced: Roles of JNK Signalling During Tumourigenesis in the Drosophila Model. Front. Cell Dev. Biol. 2020, 8, 42. [Google Scholar] [CrossRef] [Green Version]
  27. Wu, Q.; Wu, W.; Fu, B.; Shi, L.; Wang, X.; Kuca, K. JNK signaling in cancer cell survival. Med. Res. Rev. 2019, 39, 2082–2104. [Google Scholar] [CrossRef]
  28. Trivedi, S.; Starz-Gaiano, M. Drosophila Jak/STAT Signaling: Regulation and Relevance in Human Cancer and Metastasis. Int. J. Mol. Sci. 2018, 19, 4056. [Google Scholar] [CrossRef] [Green Version]
  29. Jin, W. Role of JAK/STAT3 Signaling in the Regulation of Metastasis, the Transition of Cancer Stem Cells, and Chemoresistance of Cancer by Epithelial-Mesenchymal Transition. Cells 2020, 9, 217. [Google Scholar] [CrossRef] [Green Version]
  30. Kulkarni, A.; Chang, M.T.; Vissers, J.H.A.; Dey, A.; Harvey, K.F. The Hippo Pathway as a Driver of Select Human Cancers. Trends Cancer 2020. [Google Scholar] [CrossRef]
  31. Zygulska, A.L.; Krzemieniecki, K.; Pierzchalski, P. Hippo pathway—Brief overview of its relevance in cancer. J. Physiol. Pharmacol. 2017, 68, 311–335. [Google Scholar]
  32. McIntyre, B.; Asahara, T.; Alev, C. Overview of Basic Mechanisms of Notch Signaling in Development and Disease. Adv. Exp. Med. Biol. 2020, 1227, 9–27. [Google Scholar]
  33. Previs, R.A.; Coleman, R.L.; Harris, A.L.; Sood, A.K. Molecular pathways: Translational and therapeutic implications of the Notch signaling pathway in cancer. Clin. Cancer Res. 2015, 21, 955–961. [Google Scholar] [CrossRef] [Green Version]
  34. Stephens, R.; Lim, K.; Portela, M.; Kvansakul, M.; Humbert, P.O.; Richardson, H.E. The Scribble Cell Polarity Module in the Regulation of Cell Signaling in Tissue Development and Tumorigenesis. J. Mol. Biol. 2018, 430, 3585–3612. [Google Scholar] [CrossRef]
  35. Portela, M.; Venkataramani, V.; Fahey-Lozano, N.; Seco, E.; Losada-Perez, M.; Winkler, F.; Casas-Tinto, S. Glioblastoma cells vampirize WNT from neurons and trigger a JNK/MMP signaling loop that enhances glioblastoma progression and neurodegeneration. PLoS Biol. 2019, 17, e3000545. [Google Scholar] [CrossRef]
  36. Bangi, E.; Ang, C.; Smibert, P.; Uzilov, A.V.; Teague, A.G.; Antipin, Y.; Chen, R.; Hecht, C.; Gruszczynski, N.; Yon, W.J.; et al. A personalized platform identifies trametinib plus zoledronate for a patient with KRAS-mutant metastatic colorectal cancer. Sci. Adv. 2019, 5, eaav6528. [Google Scholar] [CrossRef] [Green Version]
  37. Sonoshita, M.; Scopton, A.P.; Ung, P.M.U.; Murray, M.A.; Silber, L.; Maldonado, A.Y.; Real, A.; Schlessinger, A.; Cagan, R.L.; Dar, A.C. A whole-animal platform to advance a clinical kinase inhibitor into new disease space. Nat. Chem. Biol. 2018, 14, 291–298. [Google Scholar] [CrossRef]
  38. Das, T.K.; Esernio, J.; Cagan, R.L. Restraining Network Response to Targeted Cancer Therapies Improves Efficacy and Reduces Cellular Resistance. Cancer Res. 2018, 78, 4344–4359. [Google Scholar] [CrossRef] [Green Version]
  39. Das, T.K.; Cagan, R.L. Non-mammalian models of multiple endocrine neoplasia type 2. Endocr. Relat. Cancer 2018, 25, T91–T104. [Google Scholar] [CrossRef] [Green Version]
  40. Levine, B.D.; Cagan, R.L. Drosophila Lung Cancer Models Identify Trametinib plus Statin as Candidate Therapeutic. Cell Rep. 2016, 14, 1477–1487. [Google Scholar] [CrossRef] [Green Version]
  41. Bangi, E.; Murgia, C.; Teague, A.G.; Sansom, O.J.; Cagan, R.L. Functional exploration of colorectal cancer genomes using Drosophila. Nat. Commun. 2016, 7, 13615. [Google Scholar] [CrossRef] [PubMed]
  42. Dar, A.C.; Das, T.K.; Shokat, K.M.; Cagan, R.L. Chemical genetic discovery of targets and anti-targets for cancer polypharmacology. Nature 2012, 486, 80–84. [Google Scholar] [CrossRef] [PubMed] [Green Version]

Share and Cite

MDPI and ACS Style

Richardson, H.E.; Cordero, J.B.; Grifoni, D. Basic and Translational Models of Cooperative Oncogenesis. Int. J. Mol. Sci. 2020, 21, 5919. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21165919

AMA Style

Richardson HE, Cordero JB, Grifoni D. Basic and Translational Models of Cooperative Oncogenesis. International Journal of Molecular Sciences. 2020; 21(16):5919. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21165919

Chicago/Turabian Style

Richardson, Helena E., Julia B. Cordero, and Daniela Grifoni. 2020. "Basic and Translational Models of Cooperative Oncogenesis" International Journal of Molecular Sciences 21, no. 16: 5919. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21165919

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop