97 results on '"Richardson HE"'
Search Results
2. TSPAN6 is a suppressor of Ras-driven cancer
- Author
-
Humbert, PO, Pryjda, TZ, Pranjic, B, Farrell, A, Fujikura, K, de Matos Simoes, R, Karim, R, Kozieradzki, I, Cronin, SJF, Neely, GG, Meyer, TF, Hagelkruys, A, Richardson, HE, Penninger, JM, Humbert, PO, Pryjda, TZ, Pranjic, B, Farrell, A, Fujikura, K, de Matos Simoes, R, Karim, R, Kozieradzki, I, Cronin, SJF, Neely, GG, Meyer, TF, Hagelkruys, A, Richardson, HE, and Penninger, JM
- Abstract
Oncogenic mutations in the small GTPase RAS contribute to ~30% of human cancers. In a Drosophila genetic screen, we identified novel and evolutionary conserved cancer genes that affect Ras-driven tumorigenesis and metastasis in Drosophila including confirmation of the tetraspanin Tsp29Fb. However, it was not known whether the mammalian Tsp29Fb orthologue, TSPAN6, has any role in RAS-driven human epithelial tumors. Here we show that TSPAN6 suppressed tumor growth and metastatic dissemination of human RAS activating mutant pancreatic cancer xenografts. Whole-body knockout as well as tumor cell autonomous inactivation using floxed alleles of Tspan6 in mice enhanced KrasG12D-driven lung tumor initiation and malignant progression. Mechanistically, TSPAN6 binds to the EGFR and blocks EGFR-induced RAS activation. Moreover, we show that inactivation of TSPAN6 induces an epithelial-to-mesenchymal transition and inhibits cell migration in vitro and in vivo. Finally, low TSPAN6 expression correlates with poor prognosis of patients with lung and pancreatic cancers with mesenchymal morphology. Our results uncover TSPAN6 as a novel tumor suppressor receptor that controls epithelial cell identify and restrains RAS-driven epithelial cancer.
- Published
- 2022
3. PTP61F Mediates Cell Competition and Mitigates Tumorigenesis
- Author
-
La Marca, JE, Willoughby, LF, Allan, K, Portela, M, Goh, PK, Tiganis, T, Richardson, HE, La Marca, JE, Willoughby, LF, Allan, K, Portela, M, Goh, PK, Tiganis, T, and Richardson, HE
- Abstract
Tissue homeostasis via the elimination of aberrant cells is fundamental for organism survival. Cell competition is a key homeostatic mechanism, contributing to the recognition and elimination of aberrant cells, preventing their malignant progression and the development of tumors. Here, using Drosophila as a model organism, we have defined a role for protein tyrosine phosphatase 61F (PTP61F) (orthologue of mammalian PTP1B and TCPTP) in the initiation and progression of epithelial cancers. We demonstrate that a Ptp61F null mutation confers cells with a competitive advantage relative to neighbouring wild-type cells, while elevating PTP61F levels has the opposite effect. Furthermore, we show that knockdown of Ptp61F affects the survival of clones with impaired cell polarity, and that this occurs through regulation of the JAK-STAT signalling pathway. Importantly, PTP61F plays a robust non-cell-autonomous role in influencing the elimination of adjacent polarity-impaired mutant cells. Moreover, in a neoplastic RAS-driven polarity-impaired tumor model, we show that PTP61F levels determine the aggressiveness of tumors, with Ptp61F knockdown or overexpression, respectively, increasing or reducing tumor size. These effects correlate with the regulation of the RAS-MAPK and JAK-STAT signalling by PTP61F. Thus, PTP61F acts as a tumor suppressor that can function in an autonomous and non-cell-autonomous manner to ensure cellular fitness and attenuate tumorigenesis.
- Published
- 2021
4. Two-Faced: Roles of JNK Signalling During Tumourigenesis in the Drosophila Model.
- Author
-
La Marca, JE, Richardson, HE, La Marca, JE, and Richardson, HE
- Abstract
The highly conserved c-Jun N-terminal Kinase (JNK) signalling pathway has many functions, regulating a diversity of processes: from cell movement during embryogenesis to the stress response of cells after environmental insults. Studies modelling cancer using the vinegar fly, Drosophila melanogaster, have identified both pro- and anti-tumourigenic roles for JNK signalling, depending on context. As a tumour suppressor, JNK signalling commonly is activated by conserved Tumour Necrosis Factor (TNF) signalling, which promotes the caspase-mediated death of tumourigenic cells. JNK pathway activation can also occur via actin cytoskeleton alterations, and after cellular damage inflicted by reactive oxygen species (ROS). Additionally, JNK signalling frequently acts in concert with Salvador-Warts-Hippo (SWH) signalling - either upstream of or parallel to this potent growth-suppressing pathway. As a tumour promoter, JNK signalling is co-opted by cells expressing activated Ras-MAPK signalling (among other pathways), and used to drive cell morphological changes, induce invasive behaviours, block differentiation, and enable persistent cell proliferation. Furthermore, JNK is capable of non-autonomous influences within tumour microenvironments by effecting the transcription of various cell growth- and proliferation-promoting molecules. In this review, we discuss these aspects of JNK signalling in Drosophila tumourigenesis models, and highlight recent publications that have expanded our knowledge of this important and versatile pathway.
- Published
- 2020
5. Basic and Translational Models of Cooperative Oncogenesis
- Author
-
Richardson, HE, Cordero, JB, Grifoni, D, Richardson, HE, Cordero, JB, and Grifoni, D
- Abstract
n/a
- Published
- 2020
6. miR-9a mediates the role of Lethal giant larvae as an epithelial growth inhibitor in Drosophila
- Author
-
Daniel, SG, Russ, AD, Guthridge, KM, Raina, AI, Estes, PS, Parsons, LM, Richardson, HE, Schroeder, JA, Zarnescu, DC, Daniel, SG, Russ, AD, Guthridge, KM, Raina, AI, Estes, PS, Parsons, LM, Richardson, HE, Schroeder, JA, and Zarnescu, DC
- Abstract
Drosophila lethal giant larvae (lgl) encodes a conserved tumor suppressor with established roles in cell polarity, asymmetric division, and proliferation control. Lgl's human orthologs, HUGL1 and HUGL2, are altered in human cancers, however, its mechanistic role as a tumor suppressor remains poorly understood. Based on a previously established connection between Lgl and Fragile X protein (FMRP), a miRNA-associated translational regulator, we hypothesized that Lgl may exert its role as a tumor suppressor by interacting with the miRNA pathway. Consistent with this model, we found that lgl is a dominant modifier of Argonaute1 overexpression in the eye neuroepithelium. Using microarray profiling we identified a core set of ten miRNAs that are altered throughout tumorigenesis in Drosophila lgl mutants. Among these are several miRNAs previously linked to human cancers including miR-9a, which we found to be downregulated in lgl neuroepithelial tissues. To determine whether miR-9a can act as an effector of Lgl in vivo, we overexpressed it in the context of lgl knock-down by RNAi and found it able to reduce the overgrowth phenotype caused by Lgl loss in epithelia. Furthermore, cross-comparisons between miRNA and mRNA profiling in lgl mutant tissues and human breast cancer cells identified thrombospondin (tsp) as a common factor altered in both fly and human breast cancer tumorigenesis models. Our work provides the first evidence of a functional connection between Lgl and the miRNA pathway, demonstrates that miR-9a mediates Lgl's role in restricting epithelial proliferation, and provides novel insights into pathways controlled by Lgl during tumor progression.
- Published
- 2018
7. Src Cooperates with Oncogenic Ras in Tumourigenesis via the JNK and PI3K Pathways in Drosophila epithelial Tissue
- Author
-
Poon, CLC, Brumby, AM, Richardson, HE, Poon, CLC, Brumby, AM, and Richardson, HE
- Abstract
The Ras oncogene (Rat Sarcoma oncogene, a small GTPase) is a key driver of human cancer, however alone it is insufficient to produce malignancy, due to the induction of cell cycle arrest or senescence. In a Drosophila melanogaster genetic screen for genes that cooperate with oncogenic Ras (bearing the RasV12 mutation, or RasACT), we identified the Drosophila Src (Sarcoma virus oncogene) family non-receptor tyrosine protein kinase genes, Src42A and Src64B, as promoting increased hyperplasia in a whole epithelial tissue context in the Drosophila eye. Moreover, overexpression of Src cooperated with RasACT in epithelial cell clones to drive neoplastic tumourigenesis. We found that Src overexpression alone activated the Jun N-terminal Kinase (JNK) signalling pathway to promote actin cytoskeletal and cell polarity defects and drive apoptosis, whereas, in cooperation with RasACT, JNK led to a loss of differentiation and an invasive phenotype. Src + RasACT cooperative tumourigenesis was dependent on JNK as well as Phosphoinositide 3-Kinase (PI3K) signalling, suggesting that targeting these pathways might provide novel therapeutic opportunities in cancers dependent on Src and Ras signalling.
- Published
- 2018
8. Modelling Cooperative Tumorigenesis in Drosophila
- Author
-
Richardson, HE, Portela, M, Richardson, HE, and Portela, M
- Abstract
The development of human metastatic cancer is a multistep process, involving the acquisition of several genetic mutations, tumour heterogeneity, and interactions with the surrounding microenvironment. Due to the complexity of cancer development in mammals, simpler model organisms, such as the vinegar fly, Drosophila melanogaster, are being utilized to provide novel insights into the molecular mechanisms involved. In this review, we highlight recent advances in modelling tumorigenesis using the Drosophila model, focusing on the cooperation of oncogenes or tumour suppressors, and the interaction of mutant cells with the surrounding tissue in epithelial tumour initiation and progression.
- Published
- 2018
9. A genome-wide Drosophila epithelial tumorigenesis screen identifies Tetraspanin 29Fb as an evolutionarily conserved suppressor of Ras-driven cancer
- Author
-
Perrimon, N, Zoranovic, T, Manent, J, Willoughby, L, de Simoes, RM, La Marca, JE, Golenkina, S, Xia, C, Gruber, S, Angjeli, B, Kanitz, EE, Cronin, SJF, Neely, GG, Wernitznig, A, Humbert, PO, Simpson, KJ, Mitsiades, CS, Richardson, HE, Penninger, JM, Perrimon, N, Zoranovic, T, Manent, J, Willoughby, L, de Simoes, RM, La Marca, JE, Golenkina, S, Xia, C, Gruber, S, Angjeli, B, Kanitz, EE, Cronin, SJF, Neely, GG, Wernitznig, A, Humbert, PO, Simpson, KJ, Mitsiades, CS, Richardson, HE, and Penninger, JM
- Abstract
Oncogenic mutations in the small GTPase Ras contribute to ~30% of human cancers. However, Ras mutations alone are insufficient for tumorigenesis, therefore it is paramount to identify cooperating cancer-relevant signaling pathways. We devised an in vivo near genome-wide, functional screen in Drosophila and discovered multiple novel, evolutionarily-conserved pathways controlling Ras-driven epithelial tumorigenesis. Human gene orthologs of the fly hits were significantly downregulated in thousands of primary tumors, revealing novel prognostic markers for human epithelial tumors. Of the top 100 candidate tumor suppressor genes, 80 were validated in secondary Drosophila assays, identifying many known cancer genes and multiple novel candidate genes that cooperate with Ras-driven tumorigenesis. Low expression of the confirmed hits significantly correlated with the KRASG12 mutation status and poor prognosis in pancreatic cancer. Among the novel top 80 candidate cancer genes, we mechanistically characterized the function of the top hit, the Tetraspanin family member Tsp29Fb, revealing that Tsp29Fb regulates EGFR signaling, epithelial architecture and restrains tumor growth and invasion. Our functional Drosophila screen uncovers multiple novel and evolutionarily conserved epithelial cancer genes, and experimentally confirmed Tsp29Fb as a key regulator of EGFR/Ras induced epithelial tumor growth and invasion.
- Published
- 2018
10. Autophagy suppresses Ras-driven epithelial tumourigenesis by limiting the accumulation of reactive oxygen species
- Author
-
Manent, J, Banerjee, S, de Matos Simoes, R, Zoranovic, T, Mitsiades, C, Penninger, JM, Simpson, KJ, Humbert, PO, Richardson, HE, Manent, J, Banerjee, S, de Matos Simoes, R, Zoranovic, T, Mitsiades, C, Penninger, JM, Simpson, KJ, Humbert, PO, and Richardson, HE
- Abstract
Activation of Ras signalling occurs in ~30% of human cancers; however, activated Ras alone is not sufficient for tumourigenesis. In a screen for tumour suppressors that cooperate with oncogenic Ras (RasV12) in Drosophila, we identified genes involved in the autophagy pathway. Bioinformatic analysis of human tumours revealed that several core autophagy genes, including GABARAP, correlate with oncogenic KRAS mutations and poor prognosis in human pancreatic cancer, supporting a potential tumour-suppressive effect of the pathway in Ras-driven human cancers. In Drosophila, we demonstrate that blocking autophagy at any step of the pathway enhances RasV12-driven epithelial tissue overgrowth via the accumulation of reactive oxygen species and activation of the Jun kinase stress response pathway. Blocking autophagy in RasV12 clones also results in non-cell-autonomous effects with autophagy, cell proliferation and caspase activation induced in adjacent wild-type cells. Our study has implications for understanding the interplay between perturbations in Ras signalling and autophagy in tumourigenesis, which might inform the development of novel therapeutics targeting Ras-driven cancers.
- Published
- 2017
11. A Kinome RNAi Screen in Drosophila Identifies Novel Genes Interacting with Lgl, aPKC, and Crb Cell Polarity Genes in Epithelial Tissues
- Author
-
Parsons, LM, Grzeschik, NA, Amaratunga, K, Burke, P, Quinn, LM, Richardson, HE, Parsons, LM, Grzeschik, NA, Amaratunga, K, Burke, P, Quinn, LM, and Richardson, HE
- Abstract
In both Drosophila melanogaster and mammalian systems, epithelial structure and underlying cell polarity are essential for proper tissue morphogenesis and organ growth. Cell polarity interfaces with multiple cellular processes that are regulated by the phosphorylation status of large protein networks. To gain insight into the molecular mechanisms that coordinate cell polarity with tissue growth, we screened a boutique collection of RNAi stocks targeting the kinome for their capacity to modify Drosophila "cell polarity" eye and wing phenotypes. Initially, we identified kinase or phosphatase genes whose depletion modified adult eye phenotypes associated with the manipulation of cell polarity complexes (via overexpression of Crb or aPKC). We next conducted a secondary screen to test whether these cell polarity modifiers altered tissue overgrowth associated with depletion of Lgl in the wing. These screens identified Hippo, Jun kinase (JNK), and Notch signaling pathways, previously linked to cell polarity regulation of tissue growth. Furthermore, novel pathways not previously connected to cell polarity regulation of tissue growth were identified, including Wingless (Wg/Wnt), Ras, and lipid/Phospho-inositol-3-kinase (PI3K) signaling pathways. Additionally, we demonstrated that the "nutrient sensing" kinases Salt Inducible Kinase 2 and 3 (SIK2 and 3) are potent modifiers of cell polarity phenotypes and regulators of tissue growth. Overall, our screen has revealed novel cell polarity-interacting kinases and phosphatases that affect tissue growth, providing a platform for investigating molecular mechanisms coordinating cell polarity and tissue growth during development.
- Published
- 2017
12. Differential regulation of protein tyrosine kinase signalling by Dock and the PTP61F variants
- Author
-
Willoughby, LF, Manent, J, Allan, K, Lee, H, Portela, M, Wiede, F, Warr, C, Meng, T-C, Tiganis, T, Richardson, HE, Willoughby, LF, Manent, J, Allan, K, Lee, H, Portela, M, Wiede, F, Warr, C, Meng, T-C, Tiganis, T, and Richardson, HE
- Abstract
Tyrosine phosphorylation-dependent signalling is coordinated by the opposing actions of protein tyrosine kinases (PTKs) and protein tyrosine phosphatases (PTPs). There is a growing list of adaptor proteins that interact with PTPs and facilitate the dephosphorylation of substrates. The extent to which any given adaptor confers selectivity for any given substrate in vivo remains unclear. Here we have taken advantage of Drosophila melanogaster as a model organism to explore the influence of the SH3/SH2 adaptor protein Dock on the abilities of the membrane (PTP61Fm)- and nuclear (PTP61Fn)-targeted variants of PTP61F (the Drosophila othologue of the mammalian enzymes PTP1B and TCPTP respectively) to repress PTK signalling pathways in vivo. PTP61Fn effectively repressed the eye overgrowth associated with activation of the epidermal growth factor receptor (EGFR), PTK, or the expression of the platelet-derived growth factor/vascular endothelial growth factor receptor (PVR) or insulin receptor (InR) PTKs. PTP61Fn repressed EGFR and PVR-induced mitogen-activated protein kinase signalling and attenuated PVR-induced STAT92E signalling. By contrast, PTP61Fm effectively repressed EGFR- and PVR-, but not InR-induced tissue overgrowth. Importantly, coexpression of Dock with PTP61F allowed for the efficient repression of the InR-induced eye overgrowth, but did not enhance the PTP61Fm-mediated inhibition of EGFR and PVR-induced signalling. Instead, Dock expression increased, and PTP61Fm coexpression further exacerbated the PVR-induced eye overgrowth. These results demonstrate that Dock selectively enhances the PTP61Fm-mediated attenuation of InR signalling and underscores the specificity of PTPs and the importance of adaptor proteins in regulating PTP function in vivo.
- Published
- 2017
13. Cooperation of the BTB-Zinc finger protein, Abrupt, with cytoskeletal regulators in Drosophila epithelial tumorigenesis
- Author
-
Turkel, N, Portela, M, Poon, C, Li, J, Brumby, AM, Richardson, HE, Turkel, N, Portela, M, Poon, C, Li, J, Brumby, AM, and Richardson, HE
- Abstract
The deregulation of cell polarity or cytoskeletal regulators is a common occurrence in human epithelial cancers. Moreover, there is accumulating evidence in human epithelial cancer that BTB-ZF genes, such as Bcl6 and ZBTB7A, are oncogenic. From our previous studies in the vinegar fly, Drosophila melanogaster, we have identified a cooperative interaction between a mutation in the apico-basal cell polarity regulator Scribble (Scrib) and overexpression of the BTB-ZF protein Abrupt (Ab). Herein, we show that co-expression of ab with actin cytoskeletal regulators, RhoGEF2 or Src64B, in the developing eye-antennal epithelial tissue results in the formation of overgrown amorphous tumours, whereas ab and DRac1 co-expression leads to non-cell autonomous overgrowth. Together with ab, these genes affect the expression of differentiation genes, resulting in tumours locked in a progenitor cell fate. Finally, we show that the expression of two mammalian genes related to ab, Bcl6 and ZBTB7A, which are oncogenes in mammalian epithelial cancers, significantly correlate with the upregulation of cytoskeletal genes or downregulation of apico-basal cell polarity neoplastic tumour suppressor genes in colorectal, lung and other human epithelial cancers. Altogether, this analysis has revealed that upregulation of cytoskeletal regulators cooperate with Abrupt in Drosophila epithelial tumorigenesis, and that high expression of human BTB-ZF genes, Bcl6 and ZBTB7A, shows significant correlations with cytoskeletal and cell polarity gene expression in specific epithelial tumour types. This highlights the need for further investigation of the cooperation between these genes in mammalian systems.
- Published
- 2015
14. BTB-Zinc Finger Oncogenes Are Required for Ras and Notch-Driven Tumorigenesis in Drosophila
- Author
-
Bergmann, A, Doggett, K, Turkel, N, Willoughby, LF, Ellul, J, Murray, MJ, Richardson, HE, Brumby, AM, Bergmann, A, Doggett, K, Turkel, N, Willoughby, LF, Ellul, J, Murray, MJ, Richardson, HE, and Brumby, AM
- Abstract
During tumorigenesis, pathways that promote the epithelial-to-mesenchymal transition (EMT) can both facilitate metastasis and endow tumor cells with cancer stem cell properties. To gain a greater understanding of how these properties are interlinked in cancers we used Drosophila epithelial tumor models, which are driven by orthologues of human oncogenes (activated alleles of Ras and Notch) in cooperation with the loss of the cell polarity regulator, scribbled (scrib). Within these tumors, both invasive, mesenchymal-like cell morphology and continual tumor overgrowth, are dependent upon Jun N-terminal kinase (JNK) activity. To identify JNK-dependent changes within the tumors we used a comparative microarray analysis to define a JNK gene signature common to both Ras and Notch-driven tumors. Amongst the JNK-dependent changes was a significant enrichment for BTB-Zinc Finger (ZF) domain genes, including chronologically inappropriate morphogenesis (chinmo). chinmo was upregulated by JNK within the tumors, and overexpression of chinmo with either RasV12 or Nintra was sufficient to promote JNK-independent epithelial tumor formation in the eye/antennal disc, and, in cooperation with RasV12, promote tumor formation in the adult midgut epithelium. Chinmo primes cells for oncogene-mediated transformation through blocking differentiation in the eye disc, and promoting an escargot-expressing stem or enteroblast cell state in the adult midgut. BTB-ZF genes are also required for Ras and Notch-driven overgrowth of scrib mutant tissue, since, although loss of chinmo alone did not significantly impede tumor development, when loss of chinmo was combined with loss of a functionally related BTB-ZF gene, abrupt, tumor overgrowth was significantly reduced. abrupt is not a JNK-induced gene, however, Abrupt is present in JNK-positive tumor cells, consistent with a JNK-associated oncogenic role. As some mammalian BTB-ZF proteins are also highly oncogenic, our work suggests that EMT-promoting
- Published
- 2015
15. The Brm-HDAC3-Erm repressor complex suppresses dedifferentiation in Drosophila type II neuroblast lineages
- Author
-
Koe, CT, Li, S, Rossi, F, Wong, JJL, Wang, Y, Zhang, Z, Chen, K, Aw, SS, Richardson, HE, Robson, P, Sung, W-K, Yu, F, Gonzalez, C, Wang, H, Koe, CT, Li, S, Rossi, F, Wong, JJL, Wang, Y, Zhang, Z, Chen, K, Aw, SS, Richardson, HE, Robson, P, Sung, W-K, Yu, F, Gonzalez, C, and Wang, H
- Abstract
The control of self-renewal and differentiation of neural stem and progenitor cells is a crucial issue in stem cell and cancer biology. Drosophila type II neuroblast lineages are prone to developing impaired neuroblast homeostasis if the limited self-renewing potential of intermediate neural progenitors (INPs) is unrestrained. Here, we demonstrate that Drosophila SWI/SNF chromatin remodeling Brahma (Brm) complex functions cooperatively with another chromatin remodeling factor, Histone deacetylase 3 (HDAC3) to suppress the formation of ectopic type II neuroblasts. We show that multiple components of the Brm complex and HDAC3 physically associate with Earmuff (Erm), a type II-specific transcription factor that prevents dedifferentiation of INPs into neuroblasts. Consistently, the predicted Erm-binding motif is present in most of known binding loci of Brm. Furthermore, brm and hdac3 genetically interact with erm to prevent type II neuroblast overgrowth. Thus, the Brm-HDAC3-Erm repressor complex suppresses dedifferentiation of INPs back into type II neuroblasts. DOI: http://dx.doi.org/10.7554/eLife.01906.001.
- Published
- 2014
16. The BTB-zinc Finger Transcription Factor Abrupt Acts as an Epithelial Oncogene in Drosophila melanogaster through Maintaining a Progenitor-like Cell State
- Author
-
Horwitz, MS, Turkel, N, Sahota, VK, Bolden, JE, Goulding, KR, Doggett, K, Willoughby, LF, Blanco, E, Martin-Blanco, E, Corominas, M, Ellul, J, Aigaki, T, Richardson, HE, Brumby, AM, Horwitz, MS, Turkel, N, Sahota, VK, Bolden, JE, Goulding, KR, Doggett, K, Willoughby, LF, Blanco, E, Martin-Blanco, E, Corominas, M, Ellul, J, Aigaki, T, Richardson, HE, and Brumby, AM
- Abstract
The capacity of tumour cells to maintain continual overgrowth potential has been linked to the commandeering of normal self-renewal pathways. Using an epithelial cancer model in Drosophila melanogaster, we carried out an overexpression screen for oncogenes capable of cooperating with the loss of the epithelial apico-basal cell polarity regulator, scribbled (scrib), and identified the cell fate regulator, Abrupt, a BTB-zinc finger protein. Abrupt overexpression alone is insufficient to transform cells, but in cooperation with scrib loss of function, Abrupt promotes the formation of massive tumours in the eye/antennal disc. The steroid hormone receptor coactivator, Taiman (a homologue of SRC3/AIB1), is known to associate with Abrupt, and Taiman overexpression also drives tumour formation in cooperation with the loss of Scrib. Expression arrays and ChIP-Seq indicates that Abrupt overexpression represses a large number of genes, including steroid hormone-response genes and multiple cell fate regulators, thereby maintaining cells within an epithelial progenitor-like state. The progenitor-like state is characterised by the failure to express the conserved Eyes absent/Dachshund regulatory complex in the eye disc, and in the antennal disc by the failure to express cell fate regulators that define the temporal elaboration of the appendage along the proximo-distal axis downstream of Distalless. Loss of scrib promotes cooperation with Abrupt through impaired Hippo signalling, which is required and sufficient for cooperative overgrowth with Abrupt, and JNK (Jun kinase) signalling, which is required for tumour cell migration/invasion but not overgrowth. These results thus identify a novel cooperating oncogene, identify mammalian family members of which are also known oncogenes, and demonstrate that epithelial tumours in Drosophila can be characterised by the maintenance of a progenitor-like state.
- Published
- 2013
17. In Drosophila, RhoGEF2 cooperates with activated Ras in tumorigenesis through a pathway involving Rho1-Rok-Myosin-II and JNK signalling
- Author
-
Khoo, P, Allan, K, Willoughby, L, Brumby, AM, Richardson, HE, Khoo, P, Allan, K, Willoughby, L, Brumby, AM, and Richardson, HE
- Abstract
The Ras oncogene contributes to ≈ 30% of human cancers, but alone is not sufficient for tumorigenesis. In a Drosophila screen for oncogenes that cooperate with an activated allele of Ras (Ras(ACT)) to promote tissue overgrowth and invasion, we identified the GTP exchange factor RhoGEF2, an activator of Rho-family signalling. Here, we show that RhoGEF2 also cooperates with an activated allele of a downstream effector of Ras, Raf (Raf(GOF)). We dissect the downstream pathways through which RhoGEF2 cooperates with Ras(ACT) (and Raf(GOF)), and show that RhoGEF2 requires Rho1, but not Rac, for tumorigenesis. Furthermore, of the Rho1 effectors, we show that RhoGEF2 + Ras (Raf)-mediated tumorigenesis requires the Rho kinase (Rok)-Myosin-II pathway, but not Diaphanous, Lim kinase or protein kinase N. The Rho1-Rok-Myosin-II pathway leads to the activation of Jun kinase (JNK), in cooperation with Ras(ACT). Moreover, we show that activation of Rok or Myosin II, using constitutively active transgenes, is sufficient for cooperative tumorigenesis with Ras(ACT), and together with Ras(ACT) leads to strong activation of JNK. Our results show that Rok-Myosin-II activity is necessary and sufficient for Ras-mediated tumorigenesis. Our observation that activation of Myosin II, which regulates Filamentous actin (F-actin) contractility without affecting F-actin levels, cooperates with Ras(ACT) to promote JNK activation and tumorigenesis, suggests that increased cell contractility is a key factor in tumorigenesis. Furthermore, we show that signalling via the Tumour necrosis factor (TNF; also known as Egr)-ligand-JNK pathway is most likely the predominant pathway that activates JNK upon Rok activation. Overall, our analysis highlights the need for further analysis of the Rok-Myosin-II pathway in cooperation with Ras in human cancers.
- Published
- 2013
18. An in vivo large-scale chemical screening platform using Drosophila for anti-cancer drug discovery
- Author
-
Willoughby, LF, Schlosser, T, Manning, SA, Parisot, JP, Street, IP, Richardson, HE, Humbert, PO, Brumby, AM, Willoughby, LF, Schlosser, T, Manning, SA, Parisot, JP, Street, IP, Richardson, HE, Humbert, PO, and Brumby, AM
- Abstract
Anti-cancer drug development involves enormous expenditure and risk. For rapid and economical identification of novel, bioavailable anti-tumour chemicals, the use of appropriate in vivo tumour models suitable for large-scale screening is key. Using a Drosophila Ras-driven tumour model, we demonstrate that tumour overgrowth can be curtailed by feeding larvae with chemicals that have the in vivo pharmacokinetics essential for drug development and known efficacy against human tumour cells. We then develop an in vivo 96-well plate chemical screening platform to carry out large-scale chemical screening with the tumour model. In a proof-of-principle pilot screen of 2000 compounds, we identify the glutamine analogue, acivicin, a chemical with known activity against human tumour cells, as a potent and specific inhibitor of Drosophila tumour formation. RNAi-mediated knockdown of candidate acivicin target genes implicates an enzyme involved in pyrimidine biosynthesis, CTP synthase, as a possible crucial target of acivicin-mediated inhibition. Thus, the pilot screen has revealed that Drosophila tumours are glutamine-dependent, which is an emerging feature of many human cancers, and has validated the platform as a powerful and economical tool for in vivo chemical screening. The platform can also be adapted for use with other disease models, thus offering widespread applications in drug development.
- Published
- 2013
19. Loss of the Drosophila cell polarity regulator Scribbled promotes epithelial tissue overgrowth and cooperation with oncogenic Ras-Raf through impaired Hippo pathway signaling
- Author
-
Doggett, K, Grusche, FA, Richardson, HE, Brumby, AM, Doggett, K, Grusche, FA, Richardson, HE, and Brumby, AM
- Abstract
BACKGROUND: Epithelial neoplasias are associated with alterations in cell polarity and excessive cell proliferation, yet how these neoplastic properties are related to one another is still poorly understood. The study of Drosophila genes that function as neoplastic tumor suppressors by regulating both of these properties has significant potential to clarify this relationship. RESULTS: Here we show in Drosophila that loss of Scribbled (Scrib), a cell polarity regulator and neoplastic tumor suppressor, results in impaired Hippo pathway signaling in the epithelial tissues of both the eye and wing imaginal disc. scrib mutant tissue overgrowth, but not the loss of cell polarity, is dependent upon defective Hippo signaling and can be rescued by knockdown of either the TEAD/TEF family transcription factor Scalloped or the transcriptional coactivator Yorkie in the eye disc, or reducing levels of Yorkie in the wing disc. Furthermore, loss of Scrib sensitizes tissue to transformation by oncogenic Ras-Raf signaling, and Yorkie-Scalloped activity is required to promote this cooperative tumor overgrowth. The inhibition of Hippo signaling in scrib mutant eye disc clones is not dependent upon JNK activity, but can be significantly rescued by reducing aPKC kinase activity, and ectopic aPKC activity is sufficient to impair Hippo signaling in the eye disc, even when JNK signaling is blocked. In contrast, warts mutant overgrowth does not require aPKC activity. Moreover, reducing endogenous levels of aPKC or increasing Scrib or Lethal giant larvae levels does not promote increased Hippo signaling, suggesting that aPKC activity is not normally rate limiting for Hippo pathway activity. Epistasis experiments suggest that Hippo pathway inhibition in scrib mutants occurs, at least in part, downstream or in parallel to both the Expanded and Fat arms of Hippo pathway regulation. CONCLUSIONS: Loss of Scrib promotes Yorkie/Scalloped-dependent epithelial tissue overgrowth, and this is also impor
- Published
- 2011
20. The lethal giant larvae tumour suppressor mutation requires dMyc oncoprotein to promote clonal malignancy
- Author
-
Froldi, F, Ziosi, M, Garoia, F, Pession, A, Grzeschik, NA, Bellosta, P, Strand, D, Richardson, HE, Grifoni, D, Froldi, F, Ziosi, M, Garoia, F, Pession, A, Grzeschik, NA, Bellosta, P, Strand, D, Richardson, HE, and Grifoni, D
- Abstract
BACKGROUND: Neoplastic overgrowth depends on the cooperation of several mutations ultimately leading to major rearrangements in cellular behaviour. Precancerous cells are often removed by cell death from normal tissues in the early steps of the tumourigenic process, but the molecules responsible for such a fundamental safeguard process remain in part elusive. With the aim to investigate the molecular crosstalk occurring between precancerous and normal cells in vivo, we took advantage of the clonal analysis methods that are available in Drosophila for studying the phenotypes due to lethal giant larvae (lgl) neoplastic mutation induced in different backgrounds and tissues. RESULTS: We observed that lgl mutant cells growing in wild-type imaginal wing discs show poor viability and are eliminated by Jun N-terminal Kinase (JNK)-dependent cell death. Furthermore, they express very low levels of dMyc oncoprotein compared with those found in the surrounding normal tissue. Evidence that this is a cause of lgl mutant cells elimination was obtained by increasing dMyc levels in lgl mutant clones: their overgrowth potential was indeed re-established, with mutant cells overwhelming the neighbouring tissue and forming tumourous masses displaying several cancer hallmarks. Moreover, when lgl mutant clones were induced in backgrounds of slow-dividing cells, they upregulated dMyc, lost apical-basal cell polarity and were able to overgrow. Those phenotypes were abolished by reducing dMyc levels in the mutant clones, thereby confirming its key role in lgl-induced tumourigenesis. Furthermore, we show that the eiger-dependent Intrinsic Tumour Suppressor pathway plays only a minor role in eliminating lgl mutant cells in the wing pouch; lgl-/- clonal death in this region is instead driven mainly by dMyc-induced Cell Competition. CONCLUSIONS: Our results provide the first evidence that dMyc oncoprotein is required in lgl tumour suppressor mutant tissue to promote invasive overgrowth in larval
- Published
- 2010
21. scribble mutants promote aPKC and JNK-dependent epithelial neoplasia independently of Crumbs
- Author
-
Leong, GR, Goulding, KR, Amin, N, Richardson, HE, Brumby, AM, Leong, GR, Goulding, KR, Amin, N, Richardson, HE, and Brumby, AM
- Abstract
BACKGROUND: Metastatic neoplasias are characterized by excessive cell proliferation and disruptions to apico-basal cell polarity and tissue architecture. Understanding how alterations in cell polarity can impact upon tumour development is, therefore, a central issue in cancer biology. The Drosophila gene scribble (scrib) encodes a PDZ-domain scaffolding protein that regulates cell polarity and acts as a tumour suppressor in flies. Increasing evidence also implicates the loss of human Scrib in cancer. In this report, we investigate how loss of Scrib promotes epithelial tumourigenesis in Drosophila, both alone and in cooperation with oncogenic mutations. RESULTS: We find that genetically distinct atypical protein kinase C (aPKC)-dependent and Jun N-terminal kinase (JNK)-dependent alterations in scrib mutants drive epithelial tumourigenesis. First, we show that over-expression of the apical cell polarity determinants Crumbs (Crb) or aPKC induces similar cell morphology defects and over-proliferation phenotypes as scrib loss-of-function. However, the morphological and proliferative defects in scrib mutants are independent of Crb function, and instead can be rescued by a dominant negative (kinase dead) aPKC transgene. Secondly, we demonstrate that loss of Scrib promotes oncogene-mediated transformation through both aPKC and JNK-dependent pathways. JNK normally promotes apoptosis of scrib mutant cells. However, in cooperation with oncogenic activated Ras or Notch signalling, JNK becomes an essential driver of tumour overgrowth and invasion. aPKC-dependent signalling in scrib mutants cooperates with JNK to significantly enhance oncogene-mediated tumour overgrowth. CONCLUSION: These results demonstrate distinct aPKC and JNK-dependent pathways through which loss of Scrib promotes tumourigenesis in Drosophila. This is likely to have a direct relevance to the way in which human Scrib can similarly restrain an oncogene-mediated transformation and, more generally, on how the out
- Published
- 2009
22. The lethal giant larvae tumour suppressor mutation requires dMyc oncoprotein to promote clonal malignancy
- Author
-
Bellosta Paola, Grzeschik Nicola A, Pession Andrea, Garoia Flavio, Ziosi Marcello, Froldi Francesca, Strand Dennis, Richardson Helena E, Pession Annalisa, and Grifoni Daniela
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Abstract Background Neoplastic overgrowth depends on the cooperation of several mutations ultimately leading to major rearrangements in cellular behaviour. Precancerous cells are often removed by cell death from normal tissues in the early steps of the tumourigenic process, but the molecules responsible for such a fundamental safeguard process remain in part elusive. With the aim to investigate the molecular crosstalk occurring between precancerous and normal cells in vivo, we took advantage of the clonal analysis methods that are available in Drosophila for studying the phenotypes due to lethal giant larvae (lgl) neoplastic mutation induced in different backgrounds and tissues. Results We observed that lgl mutant cells growing in wild-type imaginal wing discs show poor viability and are eliminated by Jun N-terminal Kinase (JNK)-dependent cell death. Furthermore, they express very low levels of dMyc oncoprotein compared with those found in the surrounding normal tissue. Evidence that this is a cause of lgl mutant cells elimination was obtained by increasing dMyc levels in lgl mutant clones: their overgrowth potential was indeed re-established, with mutant cells overwhelming the neighbouring tissue and forming tumourous masses displaying several cancer hallmarks. Moreover, when lgl mutant clones were induced in backgrounds of slow-dividing cells, they upregulated dMyc, lost apical-basal cell polarity and were able to overgrow. Those phenotypes were abolished by reducing dMyc levels in the mutant clones, thereby confirming its key role in lgl-induced tumourigenesis. Furthermore, we show that the eiger-dependent Intrinsic Tumour Suppressor pathway plays only a minor role in eliminating lgl mutant cells in the wing pouch; lgl-/- clonal death in this region is instead driven mainly by dMyc-induced Cell Competition. Conclusions Our results provide the first evidence that dMyc oncoprotein is required in lgl tumour suppressor mutant tissue to promote invasive overgrowth in larval and adult epithelial tissues. Moreover, we show that dMyc abundance inside versus outside the mutant clones plays a key role in driving neoplastic overgrowth.
- Published
- 2010
- Full Text
- View/download PDF
23. Genetical genomic determinants of alcohol consumption in rats and humans
- Author
-
Mangion Jonathan, Pravenec Michal, Hübner Norbert, Heinig Matthias, Bell Richard L, Kechris Katerina, Richardson Heather N, Koob George, Goldman David, Hodgkinson Colin, Flodman Pam, Printz Morton, Saba Laura, Tabakoff Boris, Legault Lucie, Dongier Maurice, Conigrave Katherine M, Whitfield John B, Saunders John, Grant Bridget, and Hoffman Paula L
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Abstract Background We have used a genetical genomic approach, in conjunction with phenotypic analysis of alcohol consumption, to identify candidate genes that predispose to varying levels of alcohol intake by HXB/BXH recombinant inbred rat strains. In addition, in two populations of humans, we assessed genetic polymorphisms associated with alcohol consumption using a custom genotyping array for 1,350 single nucleotide polymorphisms (SNPs). Our goal was to ascertain whether our approach, which relies on statistical and informatics techniques, and non-human animal models of alcohol drinking behavior, could inform interpretation of genetic association studies with human populations. Results In the HXB/BXH recombinant inbred (RI) rats, correlation analysis of brain gene expression levels with alcohol consumption in a two-bottle choice paradigm, and filtering based on behavioral and gene expression quantitative trait locus (QTL) analyses, generated a list of candidate genes. A literature-based, functional analysis of the interactions of the products of these candidate genes defined pathways linked to presynaptic GABA release, activation of dopamine neurons, and postsynaptic GABA receptor trafficking, in brain regions including the hypothalamus, ventral tegmentum and amygdala. The analysis also implicated energy metabolism and caloric intake control as potential influences on alcohol consumption by the recombinant inbred rats. In the human populations, polymorphisms in genes associated with GABA synthesis and GABA receptors, as well as genes related to dopaminergic transmission, were associated with alcohol consumption. Conclusion Our results emphasize the importance of the signaling pathways identified using the non-human animal models, rather than single gene products, in identifying factors responsible for complex traits such as alcohol consumption. The results suggest cross-species similarities in pathways that influence predisposition to consume alcohol by rats and humans. The importance of a well-defined phenotype is also illustrated. Our results also suggest that different genetic factors predispose alcohol dependence versus the phenotype of alcohol consumption.
- Published
- 2009
- Full Text
- View/download PDF
24. scribble mutants promote aPKC and JNK-dependent epithelial neoplasia independently of Crumbs
- Author
-
Richardson Helena E, Amin Nancy, Goulding Karen R, Leong Gregory R, and Brumby Anthony M
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Abstract Background Metastatic neoplasias are characterized by excessive cell proliferation and disruptions to apico-basal cell polarity and tissue architecture. Understanding how alterations in cell polarity can impact upon tumour development is, therefore, a central issue in cancer biology. The Drosophila gene scribble (scrib) encodes a PDZ-domain scaffolding protein that regulates cell polarity and acts as a tumour suppressor in flies. Increasing evidence also implicates the loss of human Scrib in cancer. In this report, we investigate how loss of Scrib promotes epithelial tumourigenesis in Drosophila, both alone and in cooperation with oncogenic mutations. Results We find that genetically distinct atypical protein kinase C (aPKC)-dependent and Jun N-terminal kinase (JNK)-dependent alterations in scrib mutants drive epithelial tumourigenesis. First, we show that over-expression of the apical cell polarity determinants Crumbs (Crb) or aPKC induces similar cell morphology defects and over-proliferation phenotypes as scrib loss-of-function. However, the morphological and proliferative defects in scrib mutants are independent of Crb function, and instead can be rescued by a dominant negative (kinase dead) aPKC transgene. Secondly, we demonstrate that loss of Scrib promotes oncogene-mediated transformation through both aPKC and JNK-dependent pathways. JNK normally promotes apoptosis of scrib mutant cells. However, in cooperation with oncogenic activated Ras or Notch signalling, JNK becomes an essential driver of tumour overgrowth and invasion. aPKC-dependent signalling in scrib mutants cooperates with JNK to significantly enhance oncogene-mediated tumour overgrowth. Conclusion These results demonstrate distinct aPKC and JNK-dependent pathways through which loss of Scrib promotes tumourigenesis in Drosophila. This is likely to have a direct relevance to the way in which human Scrib can similarly restrain an oncogene-mediated transformation and, more generally, on how the outcome of oncogenic signalling can be profoundly perturbed by defects in apico-basal epithelial cell polarity.
- Published
- 2009
- Full Text
- View/download PDF
25. The Drosophila tumour suppressor Lgl and Vap33 activate the Hippo pathway through a dual mechanism.
- Author
-
Portela M, Mukherjee S, Paul S, La Marca JE, Parsons LM, Veraksa A, and Richardson HE
- Subjects
- Animals, Adenosine Triphosphatases metabolism, Cell Polarity, Drosophila metabolism, Drosophila melanogaster, Hippo Signaling Pathway, Intracellular Signaling Peptides and Proteins genetics, Intracellular Signaling Peptides and Proteins metabolism, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism, Signal Transduction, Tumor Suppressor Proteins metabolism, Drosophila Proteins genetics, Drosophila Proteins metabolism, Neoplasms
- Abstract
The tumour suppressor, Lethal (2) giant larvae [Lgl; also known as L(2)gl], is an evolutionarily conserved protein that was discovered in the vinegar fly Drosophila, where its depletion results in tissue overgrowth and loss of cell polarity. Lgl links cell polarity and tissue growth through regulation of the Notch and the Hippo signalling pathways. Lgl regulates the Notch pathway by inhibiting V-ATPase activity via Vap33. How Lgl regulates the Hippo pathway was unclear. In this current study, we show that V-ATPase activity inhibits the Hippo pathway, whereas Vap33 acts to activate Hippo signalling. Vap33 physically and genetically interacts with the actin cytoskeletal regulators RtGEF (Pix) and Git, which also bind to the Hippo protein (Hpo) and are involved in the activation of the Hippo pathway. Additionally, we show that the ADP ribosylation factor Arf79F (Arf1), which is a Hpo interactor, is involved in the inhibition of the Hippo pathway. Altogether, our data suggest that Lgl acts via Vap33 to activate the Hippo pathway by a dual mechanism: (1) through interaction with RtGEF, Git and Arf79F, and (2) through interaction and inhibition of the V-ATPase, thereby controlling epithelial tissue growth., Competing Interests: Competing interests The authors declare no competing or financial interests., (© 2024. Published by The Company of Biologists Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
26. A Drosophila chemical screen reveals synergistic effect of MEK and DGKα inhibition in Ras-driven cancer.
- Author
-
La Marca JE, Ely RW, Diepstraten ST, Burke P, Kelly GL, Humbert PO, and Richardson HE
- Subjects
- Animals, Humans, Mitogen-Activated Protein Kinase Kinases, Drosophila, Cell Line, Tumor, Signal Transduction, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Neoplasms drug therapy
- Abstract
Elevated Ras signalling is highly prevalent in human cancer; however, targeting Ras-driven cancers with Ras pathway inhibitors often leads to undesirable side effects and to drug resistance. Thus, identifying compounds that synergise with Ras pathway inhibitors would enable lower doses of the Ras pathway inhibitors to be used and also decrease the acquisition of drug resistance. Here, in a specialised chemical screen using a Drosophila model of Ras-driven cancer, we have identified compounds that reduce tumour size by synergising with sub-therapeutic doses of the Ras pathway inhibitor trametinib, which targets MEK, the mitogen-activated protein kinase kinase, in this pathway. Analysis of one of the hits, ritanserin, and related compounds revealed that diacyl glycerol kinase α (DGKα, Dgk in Drosophila) was the critical target required for synergism with trametinib. Human epithelial cells harbouring the H-RAS oncogene and knockdown of the cell polarity gene SCRIB were also sensitive to treatment with trametinib and DGKα inhibitors. Mechanistically, DGKα inhibition synergises with trametinib by increasing the P38 stress-response signalling pathway in H-RASG12V SCRIBRNAi cells, which could lead to cell quiescence. Our results reveal that targeting Ras-driven human cancers with Ras pathway and DGKα inhibitors should be an effective combination drug therapy., Competing Interests: Competing interests The authors declare no competing or financial interests., (© 2023. Published by The Company of Biologists Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
27. Ptp61F integrates Hippo, TOR, and actomyosin pathways to control three-dimensional organ size.
- Author
-
Liu P, Guo Y, Xu W, Song S, Li X, Wang X, Lu J, Guo X, Richardson HE, and Ma X
- Subjects
- Animals, Humans, Intracellular Signaling Peptides and Proteins genetics, Protein Serine-Threonine Kinases genetics, Organ Size, Trans-Activators metabolism, Nuclear Proteins metabolism, Drosophila metabolism, Protein Tyrosine Phosphatases, Non-Receptor, Actomyosin, Drosophila Proteins metabolism
- Abstract
Precise organ size control is fundamental for all metazoans, but how organ size is controlled in a three-dimensional (3D) way remains largely unexplored at the molecular level. Here, we screen and identify Drosophila Ptp61F as a pivotal regulator of organ size that integrates the Hippo pathway, TOR pathway, and actomyosin machinery. Pathologically, Ptp61F loss synergizes with Ras
V12 to induce tumorigenesis. Physiologically, Ptp61F depletion increases body size and drives neoplastic intestinal tumor formation and stem cell proliferation. Ptp61F also regulates cell contractility and myosin activation and controls 3D cell shape by reducing cell height and horizontal cell size. Mechanistically, Ptp61F forms a complex with Expanded (Ex) and increases endosomal localization of Ex and Yki. Furthermore, we demonstrate that PTPN2, the conserved human ortholog of Ptp61F, can functionally substitute for Ptp61F in Drosophila. Our work defines Ptp61F as an essential determinant that controls 3D organ size under both physiological and pathological conditions., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
28. TSPAN6 is a suppressor of Ras-driven cancer.
- Author
-
Humbert PO, Pryjda TZ, Pranjic B, Farrell A, Fujikura K, de Matos Simoes R, Karim R, Kozieradzki I, Cronin SJF, Neely GG, Meyer TF, Hagelkruys A, Richardson HE, and Penninger JM
- Subjects
- Animals, Carcinogenesis genetics, Cell Line, Tumor, Cell Transformation, Neoplastic genetics, Genes, ras, Humans, Mammals genetics, Mammals metabolism, Mice, Mutation, Proto-Oncogene Proteins p21(ras) genetics, Proto-Oncogene Proteins p21(ras) metabolism, Oncogenes, Pancreatic Neoplasms pathology, Tetraspanins genetics, Tetraspanins metabolism
- Abstract
Oncogenic mutations in the small GTPase RAS contribute to ~30% of human cancers. In a Drosophila genetic screen, we identified novel and evolutionary conserved cancer genes that affect Ras-driven tumorigenesis and metastasis in Drosophila including confirmation of the tetraspanin Tsp29Fb. However, it was not known whether the mammalian Tsp29Fb orthologue, TSPAN6, has any role in RAS-driven human epithelial tumors. Here we show that TSPAN6 suppressed tumor growth and metastatic dissemination of human RAS activating mutant pancreatic cancer xenografts. Whole-body knockout as well as tumor cell autonomous inactivation using floxed alleles of Tspan6 in mice enhanced Kras
G12D -driven lung tumor initiation and malignant progression. Mechanistically, TSPAN6 binds to the EGFR and blocks EGFR-induced RAS activation. Moreover, we show that inactivation of TSPAN6 induces an epithelial-to-mesenchymal transition and inhibits cell migration in vitro and in vivo. Finally, low TSPAN6 expression correlates with poor prognosis of patients with lung and pancreatic cancers with mesenchymal morphology. Our results uncover TSPAN6 as a novel tumor suppressor receptor that controls epithelial cell identify and restrains RAS-driven epithelial cancer., (© 2022. Crown.)- Published
- 2022
- Full Text
- View/download PDF
29. Correction: TSPAN6 is a suppressor of Ras-driven cancer.
- Author
-
Humbert PO, Pryjda TZ, Pranjic B, Farrell A, Fujikura K, de Matos Simoes R, Karim R, Kozieradzki I, Cronin SJF, Neely GG, Meyer TF, Hagelkruys A, Richardson HE, and Penninger JM
- Published
- 2022
- Full Text
- View/download PDF
30. PTP61F Mediates Cell Competition and Mitigates Tumorigenesis.
- Author
-
La Marca JE, Willoughby LF, Allan K, Portela M, Goh PK, Tiganis T, and Richardson HE
- Subjects
- Animals, Carcinogenesis genetics, Carcinogenesis pathology, Drosophila Proteins genetics, Drosophila melanogaster genetics, Drosophila melanogaster growth & development, Janus Kinase 1 genetics, Janus Kinase 1 metabolism, Mitogen-Activated Protein Kinase Kinases genetics, Mitogen-Activated Protein Kinase Kinases metabolism, Neoplasms metabolism, Neoplasms pathology, Protein Tyrosine Phosphatases, Non-Receptor genetics, STAT Transcription Factors genetics, STAT Transcription Factors metabolism, Signal Transduction, ras Proteins genetics, ras Proteins metabolism, Carcinogenesis metabolism, Cell Competition, Drosophila Proteins metabolism, Drosophila melanogaster metabolism, Neoplasms prevention & control, Protein Tyrosine Phosphatases, Non-Receptor metabolism
- Abstract
Tissue homeostasis via the elimination of aberrant cells is fundamental for organism survival. Cell competition is a key homeostatic mechanism, contributing to the recognition and elimination of aberrant cells, preventing their malignant progression and the development of tumors. Here, using Drosophila as a model organism, we have defined a role for protein tyrosine phosphatase 61F (PTP61F) (orthologue of mammalian PTP1B and TCPTP) in the initiation and progression of epithelial cancers. We demonstrate that a Ptp61F null mutation confers cells with a competitive advantage relative to neighbouring wild-type cells, while elevating PTP61F levels has the opposite effect. Furthermore, we show that knockdown of Ptp61F affects the survival of clones with impaired cell polarity, and that this occurs through regulation of the JAK-STAT signalling pathway. Importantly, PTP61F plays a robust non-cell-autonomous role in influencing the elimination of adjacent polarity-impaired mutant cells. Moreover, in a neoplastic RAS-driven polarity-impaired tumor model, we show that PTP61F levels determine the aggressiveness of tumors, with Ptp61F knockdown or overexpression, respectively, increasing or reducing tumor size. These effects correlate with the regulation of the RAS-MAPK and JAK-STAT signalling by PTP61F. Thus, PTP61F acts as a tumor suppressor that can function in an autonomous and non-cell-autonomous manner to ensure cellular fitness and attenuate tumorigenesis.
- Published
- 2021
- Full Text
- View/download PDF
31. Tumor-derived MMPs regulate cachexia in a Drosophila cancer model.
- Author
-
Lodge W, Zavortink M, Golenkina S, Froldi F, Dark C, Cheung S, Parker BL, Blazev R, Bakopoulos D, Christie EL, Wimmer VC, Duckworth BC, Richardson HE, and Cheng LY
- Subjects
- Animals, Basement Membrane metabolism, Drosophila metabolism, Extracellular Matrix metabolism, Muscular Atrophy metabolism, Adipose Tissue metabolism, Matrix Metalloproteinases metabolism, Muscle, Skeletal metabolism, Neoplasms metabolism
- Abstract
Cachexia, the wasting syndrome commonly observed in advanced cancer patients, accounts for up to one-third of cancer-related mortalities. We have established a Drosophila larval model of organ wasting whereby epithelial overgrowth in eye-antennal discs leads to wasting of the adipose tissue and muscles. The wasting is associated with fat-body remodeling and muscle detachment and is dependent on tumor-secreted matrix metalloproteinase 1 (Mmp1). Mmp1 can both modulate TGFβ signaling in the fat body and disrupt basement membrane (BM)/extracellular matrix (ECM) protein localization in both the fat body and the muscle. Inhibition of TGFβ signaling or Mmps in the fat body/muscle using a QF2-QUAS binary expression system rescues muscle wasting in the presence of tumor. Altogether, our study proposes that tumor-derived Mmps are central mediators of organ wasting in cancer cachexia., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
32. Basic and Translational Models of Cooperative Oncogenesis.
- Author
-
Richardson HE, Cordero JB, and Grifoni D
- Subjects
- Animals, Animals, Genetically Modified, Biomedical Research methods, Carcinogenesis pathology, Cell Line, Cell Transformation, Neoplastic pathology, Drug Screening Assays, Antitumor, Epistasis, Genetic physiology, Humans, Translational Research, Biomedical methods, Carcinogenesis genetics, Cell Transformation, Neoplastic genetics, Disease Models, Animal, Drosophila melanogaster, Epigenesis, Genetic physiology, Models, Biological
- Published
- 2020
- Full Text
- View/download PDF
33. Two-Faced: Roles of JNK Signalling During Tumourigenesis in the Drosophila Model.
- Author
-
La Marca JE and Richardson HE
- Abstract
The highly conserved c-Jun N-terminal Kinase (JNK) signalling pathway has many functions, regulating a diversity of processes: from cell movement during embryogenesis to the stress response of cells after environmental insults. Studies modelling cancer using the vinegar fly, Drosophila melanogaster , have identified both pro- and anti-tumourigenic roles for JNK signalling, depending on context. As a tumour suppressor, JNK signalling commonly is activated by conserved Tumour Necrosis Factor (TNF) signalling, which promotes the caspase-mediated death of tumourigenic cells. JNK pathway activation can also occur via actin cytoskeleton alterations, and after cellular damage inflicted by reactive oxygen species (ROS). Additionally, JNK signalling frequently acts in concert with Salvador-Warts-Hippo (SWH) signalling - either upstream of or parallel to this potent growth-suppressing pathway. As a tumour promoter, JNK signalling is co-opted by cells expressing activated Ras-MAPK signalling (among other pathways), and used to drive cell morphological changes, induce invasive behaviours, block differentiation, and enable persistent cell proliferation. Furthermore, JNK is capable of non-autonomous influences within tumour microenvironments by effecting the transcription of various cell growth- and proliferation-promoting molecules. In this review, we discuss these aspects of JNK signalling in Drosophila tumourigenesis models, and highlight recent publications that have expanded our knowledge of this important and versatile pathway., (Copyright © 2020 La Marca and Richardson.)
- Published
- 2020
- Full Text
- View/download PDF
34. Strip and Cka negatively regulate JNK signalling during Drosophila spermatogenesis.
- Author
-
La Marca JE, Diepstraten ST, Hodge AL, Wang H, Hart AH, Richardson HE, and Somers WG
- Subjects
- Animals, Animals, Genetically Modified, Cell Differentiation, Down-Regulation genetics, Male, Testis cytology, Testis growth & development, Testis metabolism, Adaptor Proteins, Signal Transducing physiology, Drosophila Proteins physiology, Drosophila melanogaster genetics, Drosophila melanogaster growth & development, MAP Kinase Signaling System genetics, Phosphate-Binding Proteins physiology, Spermatogenesis genetics
- Abstract
One fundamental property of a stem cell niche is the exchange of molecular signals between its component cells. Niche models, such as the Drosophila melanogaster testis, have been instrumental in identifying and studying the conserved genetic factors that contribute to niche molecular signalling. Here, we identify jam packed ( jam ), an allele of Striatin interacting protein ( Strip ), which is a core member of the highly conserved Striatin-interacting phosphatase and kinase (STRIPAK) complex. In the developing Drosophila testis, Strip cell-autonomously regulates the differentiation and morphology of the somatic lineage, and non-cell-autonomously regulates the proliferation and differentiation of the germline lineage. Mechanistically, Strip acts in the somatic lineage with its STRIPAK partner, Connector of kinase to AP-1 ( Cka ), where they negatively regulate the Jun N-terminal kinase (JNK) signalling pathway. Our study reveals a novel role for Strip / Cka in JNK pathway regulation during spermatogenesis within the developing Drosophila testis., Competing Interests: Competing interestsThe authors declare no competing or financial interests., (© 2019. Published by The Company of Biologists Ltd.)
- Published
- 2019
- Full Text
- View/download PDF
35. Drosophila Models of Cell Polarity and Cell Competition in Tumourigenesis.
- Author
-
Fahey-Lozano N, La Marca JE, Portela M, and Richardson HE
- Subjects
- Animals, Disease Models, Animal, Cell Polarity, Cell Transformation, Neoplastic, Drosophila, Drosophila Proteins
- Abstract
Cell competition is an important surveillance mechanism that measures relative fitness between cells in a tissue during development, homeostasis, and disease. Specifically, cells that are "less fit" (losers) are actively eliminated by relatively "more fit" (winners) neighbours, despite the less fit cells otherwise being able to survive in a genetically uniform tissue. Originally described in the epithelial tissues of Drosophila larval imaginal discs, cell competition has since been shown to occur in other epithelial and non-epithelial Drosophila tissues, as well as in mammalian model systems. Many genes and signalling pathways have been identified as playing conserved roles in the mechanisms of cell competition. Among them are genes required for the establishment and maintenance of apico-basal cell polarity: the Crumbs/Stardust/Patj (Crb/Sdt/Patj), Bazooka/Par-6/atypical Protein Kinase C (Baz/Par-6/aPKC), and Scribbled/Discs large 1/Lethal (2) giant larvae (Scrib/Dlg1/L(2)gl) modules. In this chapter, we describe the concepts and mechanisms of cell competition, with emphasis on the relationship between cell polarity proteins and cell competition, particularly the Scrib/Dlg1/L(2)gl module, since this is the best described module in this emerging field.
- Published
- 2019
- Full Text
- View/download PDF
36. A genome-wide Drosophila epithelial tumorigenesis screen identifies Tetraspanin 29Fb as an evolutionarily conserved suppressor of Ras-driven cancer.
- Author
-
Zoranovic T, Manent J, Willoughby L, Matos de Simoes R, La Marca JE, Golenkina S, Cuiping X, Gruber S, Angjeli B, Kanitz EE, Cronin SJF, Neely GG, Wernitznig A, Humbert PO, Simpson KJ, Mitsiades CS, Richardson HE, and Penninger JM
- Subjects
- Animals, Animals, Genetically Modified, Carcinogenesis genetics, Drosophila Proteins metabolism, Drosophila melanogaster genetics, Epithelial Cells metabolism, Epithelial Cells pathology, Female, Genes, ras, Genetic Testing methods, Humans, IMP Dehydrogenase metabolism, Male, Mice, Neoplasms metabolism, Neoplasms pathology, Oncogenes, Signal Transduction, Tetraspanin 29 metabolism, Tumor Suppressor Proteins genetics, Drosophila Proteins genetics, IMP Dehydrogenase genetics, Neoplasms genetics, Tetraspanin 29 genetics
- Abstract
Oncogenic mutations in the small GTPase Ras contribute to ~30% of human cancers. However, Ras mutations alone are insufficient for tumorigenesis, therefore it is paramount to identify cooperating cancer-relevant signaling pathways. We devised an in vivo near genome-wide, functional screen in Drosophila and discovered multiple novel, evolutionarily-conserved pathways controlling Ras-driven epithelial tumorigenesis. Human gene orthologs of the fly hits were significantly downregulated in thousands of primary tumors, revealing novel prognostic markers for human epithelial tumors. Of the top 100 candidate tumor suppressor genes, 80 were validated in secondary Drosophila assays, identifying many known cancer genes and multiple novel candidate genes that cooperate with Ras-driven tumorigenesis. Low expression of the confirmed hits significantly correlated with the KRASG12 mutation status and poor prognosis in pancreatic cancer. Among the novel top 80 candidate cancer genes, we mechanistically characterized the function of the top hit, the Tetraspanin family member Tsp29Fb, revealing that Tsp29Fb regulates EGFR signaling, epithelial architecture and restrains tumor growth and invasion. Our functional Drosophila screen uncovers multiple novel and evolutionarily conserved epithelial cancer genes, and experimentally confirmed Tsp29Fb as a key regulator of EGFR/Ras induced epithelial tumor growth and invasion., Competing Interests: AW is affiliated with Boehringer Ingelheim RCV GmbH & Co KG. Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria has no competing interests with this study.
- Published
- 2018
- Full Text
- View/download PDF
37. The Scribble Cell Polarity Module in the Regulation of Cell Signaling in Tissue Development and Tumorigenesis.
- Author
-
Stephens R, Lim K, Portela M, Kvansakul M, Humbert PO, and Richardson HE
- Subjects
- Animals, Gene Expression Regulation, Humans, Organ Specificity, Structure-Activity Relationship, Biomarkers, Cell Polarity physiology, Cell Transformation, Neoplastic, Organogenesis, Signal Transduction
- Abstract
The Scribble cell polarity module, comprising Scribbled (Scrib), Discs-large (Dlg) and Lethal-2-giant larvae (Lgl), has a tumor suppressive role in mammalian epithelial cancers. The Scribble module proteins play key functions in the establishment and maintenance of different modes of cell polarity, as well as in the control of tissue growth, differentiation and directed cell migration, and therefore are major regulators of tissue development and homeostasis. Whilst molecular details are known regarding the roles of Scribble module proteins in cell polarity regulation, their precise mode of action in the regulation of other key cellular processes remains enigmatic. An accumulating body of evidence indicates that Scribble module proteins play scaffolding roles in the control of various signaling pathways, which are linked to the control of tissue growth, differentiation and cell migration. Multiple Scrib, Dlg and Lgl interacting proteins have been discovered, which are involved in diverse processes, however many function in the regulation of cellular signaling. Herein, we review the components of the Scrib, Dlg and Lgl protein interactomes, and focus on the mechanism by which they regulate cellular signaling pathways in metazoans, and how their disruption leads to cancer., (Copyright © 2018 Elsevier Ltd. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
38. Lgl reduces endosomal vesicle acidification and Notch signaling by promoting the interaction between Vap33 and the V-ATPase complex.
- Author
-
Portela M, Yang L, Paul S, Li X, Veraksa A, Parsons LM, and Richardson HE
- Subjects
- Acids metabolism, Animals, Carrier Proteins genetics, Cell Polarity, Drosophila Proteins genetics, Drosophila melanogaster genetics, Drosophila melanogaster growth & development, Epithelium growth & development, Epithelium metabolism, Eye growth & development, Eye metabolism, Female, Membrane Proteins genetics, Receptors, Notch genetics, Receptors, Notch metabolism, Tumor Suppressor Proteins genetics, Vacuolar Proton-Translocating ATPases genetics, Vacuoles metabolism, Carrier Proteins metabolism, Drosophila Proteins metabolism, Drosophila melanogaster metabolism, Endosomes metabolism, Membrane Proteins metabolism, Signal Transduction, Tumor Suppressor Proteins metabolism, Vacuolar Proton-Translocating ATPases metabolism
- Abstract
Epithelial cell polarity is linked to the control of tissue growth and tumorigenesis. The tumor suppressor and cell polarity protein lethal-2-giant larvae (Lgl) promotes Hippo signaling and inhibits Notch signaling to restrict tissue growth in Drosophila melanogaster Notch signaling is greater in lgl mutant tissue than in wild-type tissue because of increased acidification of endosomal vesicles, which promotes the proteolytic processing and activation of Notch by γ-secretase. We showed that the increased Notch signaling and tissue growth defects of lgl mutant tissue depended on endosomal vesicle acidification mediated by the vacuolar adenosine triphosphatase (V-ATPase). Lgl promoted the activity of the V-ATPase by interacting with Vap33 (VAMP-associated protein of 33 kDa). Vap33 physically and genetically interacted with Lgl and V-ATPase subunits and repressed V-ATPase-mediated endosomal vesicle acidification and Notch signaling. Vap33 overexpression reduced the abundance of the V-ATPase component Vha44, whereas Lgl knockdown reduced the binding of Vap33 to the V-ATPase component Vha68-3. Our data indicate that Lgl promotes the binding of Vap33 to the V-ATPase, thus inhibiting V-ATPase-mediated endosomal vesicle acidification and thereby reducing γ-secretase activity, Notch signaling, and tissue growth. Our findings implicate the deregulation of Vap33 and V-ATPase activity in polarity-impaired epithelial cancers., (Copyright © 2018 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2018
- Full Text
- View/download PDF
39. Src Cooperates with Oncogenic Ras in Tumourigenesis via the JNK and PI3K Pathways in Drosophila epithelial Tissue.
- Author
-
Poon CLC, Brumby AM, and Richardson HE
- Subjects
- Animals, Cell Differentiation, Cell Polarity, Compound Eye, Arthropod enzymology, Compound Eye, Arthropod metabolism, Compound Eye, Arthropod pathology, Drosophila Proteins physiology, Drosophila melanogaster metabolism, Epithelium enzymology, Epithelium metabolism, Epithelium physiopathology, Female, JNK Mitogen-Activated Protein Kinases metabolism, Male, Phosphatidylinositol 3-Kinases metabolism, Protein-Tyrosine Kinases physiology, Proto-Oncogene Proteins physiology, Proto-Oncogene Proteins pp60(c-src) physiology, ras Proteins physiology, Carcinogenesis, Drosophila Proteins metabolism, Drosophila melanogaster enzymology, Protein-Tyrosine Kinases metabolism, Proto-Oncogene Proteins metabolism, Proto-Oncogene Proteins pp60(c-src) metabolism, Signal Transduction, ras Proteins metabolism
- Abstract
The Ras oncogene (Rat Sarcoma oncogene, a small GTPase) is a key driver of human cancer, however alone it is insufficient to produce malignancy, due to the induction of cell cycle arrest or senescence. In a Drosophila melanogaster genetic screen for genes that cooperate with oncogenic Ras (bearing the Ras
V12 mutation, or RasACT ), we identified the Drosophila Src (Sarcoma virus oncogene) family non-receptor tyrosine protein kinase genes, Src42A and Src64B , as promoting increased hyperplasia in a whole epithelial tissue context in the Drosophila eye. Moreover, overexpression of Src cooperated with RasACT in epithelial cell clones to drive neoplastic tumourigenesis. We found that Src overexpression alone activated the Jun N-terminal Kinase (JNK) signalling pathway to promote actin cytoskeletal and cell polarity defects and drive apoptosis, whereas, in cooperation with RasACT , JNK led to a loss of differentiation and an invasive phenotype. Src + RasACT cooperative tumourigenesis was dependent on JNK as well as Phosphoinositide 3-Kinase (PI3K) signalling, suggesting that targeting these pathways might provide novel therapeutic opportunities in cancers dependent on Src and Ras signalling.- Published
- 2018
- Full Text
- View/download PDF
40. Drosophila melanogaster Guk-holder interacts with the Scribbled PDZ1 domain and regulates epithelial development with Scribbled and Discs Large.
- Author
-
Caria S, Magtoto CM, Samiei T, Portela M, Lim KYB, How JY, Stewart BZ, Humbert PO, Richardson HE, and Kvansakul M
- Subjects
- Animals, Cell Polarity, Drosophila Proteins genetics, Drosophila melanogaster genetics, Drosophila melanogaster growth & development, Epithelial Cells metabolism, Eye metabolism, Female, Gene Expression Regulation, Developmental, Male, Membrane Proteins, Nerve Tissue Proteins genetics, PDZ Domains, Protein Binding, Tumor Suppressor Proteins genetics, Wings, Animal metabolism, Drosophila Proteins metabolism, Drosophila melanogaster metabolism, Epithelial Cells cytology, Eye cytology, Nerve Tissue Proteins metabolism, Tumor Suppressor Proteins metabolism, Wings, Animal cytology
- Abstract
Epithelial cell polarity is controlled by components of the Scribble polarity module, and its regulation is critical for tissue architecture and cell proliferation and migration. In Drosophila melanogaster , the adaptor protein Guk-holder (Gukh) binds to the Scribbled (Scrib) and Discs Large (Dlg) components of the Scribble polarity module and plays an important role in the formation of neuromuscular junctions. However, Gukh's role in epithelial tissue formation and the molecular basis for the Scrib-Gukh interaction remain to be defined. We now show using isothermal titration calorimetry that the Scrib PDZ1 domain is the major site for an interaction with Gukh. Furthermore, we defined the structural basis of this interaction by determining the crystal structure of the Scrib PDZ1-Gukh complex. The C-terminal PDZ-binding motif of Gukh is located in the canonical ligand-binding groove of Scrib PDZ1 and utilizes an unusually extensive network of hydrogen bonds and ionic interactions to enable binding to PDZ1 with high affinity. We next examined the role of Gukh along with those of Scrib and Dlg in Drosophila epithelial tissues and found that Gukh is expressed in larval-wing and eye-epithelial tissues and co-localizes with Scrib and Dlg at the apical cell cortex. Importantly, we show that Gukh functions with Scrib and Dlg in the development of Drosophila epithelial tissues, with depletion of Gukh enhancing the eye- and wing-tissue defects caused by Scrib or Dlg depletion. Overall, our findings reveal that Scrib's PDZ1 domain functions in the interaction with Gukh and that the Scrib-Gukh interaction has a key role in epithelial tissue development in Drosophila ., (© 2018 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2018
- Full Text
- View/download PDF
41. Modelling Cooperative Tumorigenesis in Drosophila .
- Author
-
Richardson HE and Portela M
- Subjects
- Animals, Drosophila Proteins genetics, Drosophila melanogaster genetics, Genes, Tumor Suppressor physiology, Humans, Neoplasms genetics, Oncogenes genetics, Carcinogenesis pathology, Drosophila melanogaster pathogenicity, Neoplasms pathology
- Abstract
The development of human metastatic cancer is a multistep process, involving the acquisition of several genetic mutations, tumour heterogeneity, and interactions with the surrounding microenvironment. Due to the complexity of cancer development in mammals, simpler model organisms, such as the vinegar fly, Drosophila melanogaster , are being utilized to provide novel insights into the molecular mechanisms involved. In this review, we highlight recent advances in modelling tumorigenesis using the Drosophila model, focusing on the cooperation of oncogenes or tumour suppressors, and the interaction of mutant cells with the surrounding tissue in epithelial tumour initiation and progression.
- Published
- 2018
- Full Text
- View/download PDF
42. POSH regulates Hippo signaling through ubiquitin-mediated expanded degradation.
- Author
-
Ma X, Guo X, Richardson HE, Xu T, and Xue L
- Subjects
- Animals, Carrier Proteins genetics, Cytoskeletal Proteins genetics, Dextran Sulfate, Drosophila Proteins genetics, Drosophila melanogaster genetics, Gene Deletion, Genome, Intestines drug effects, Intracellular Signaling Peptides and Proteins genetics, Membrane Proteins genetics, Nerve Tissue Proteins genetics, Protein Binding, Protein Serine-Threonine Kinases genetics, Proteolysis, Signal Transduction, Stem Cells drug effects, Tumor Suppressor Proteins genetics, Tumor Suppressor Proteins metabolism, Carrier Proteins metabolism, Cytoskeletal Proteins metabolism, Drosophila Proteins metabolism, Drosophila melanogaster metabolism, Intracellular Signaling Peptides and Proteins metabolism, Membrane Proteins metabolism, Nerve Tissue Proteins metabolism, Protein Serine-Threonine Kinases metabolism, Ubiquitin metabolism
- Abstract
The Hippo signaling pathway is a master regulator of organ growth, tissue homeostasis, and tumorigenesis. The activity of the Hippo pathway is controlled by various upstream components, including Expanded (Ex), but the precise molecular mechanism of how Ex is regulated remains poorly understood. Here we identify Plenty of SH3s (POSH), an E3 ubiquitin ligase, as a key component of Hippo signaling in Drosophila POSH overexpression synergizes with loss of Kibra to induce overgrowth and up-regulation of Hippo pathway target genes. Furthermore, knockdown of POSH impedes dextran sulfate sodium-induced Yorkie-dependent intestinal stem cell renewal, suggesting a physiological role of POSH in modulating Hippo signaling. Mechanistically, POSH binds to the C-terminal of Ex and is essential for the Crumbs-induced ubiquitination and degradation of Ex. Our findings establish POSH as a crucial regulator that integrates the signal from the cell surface to negatively regulate Ex-mediated Hippo activation in Drosophila ., Competing Interests: The authors declare no conflict of interest.
- Published
- 2018
- Full Text
- View/download PDF
43. miR-9a mediates the role of Lethal giant larvae as an epithelial growth inhibitor in Drosophila .
- Author
-
Daniel SG, Russ AD, Guthridge KM, Raina AI, Estes PS, Parsons LM, Richardson HE, Schroeder JA, and Zarnescu DC
- Abstract
Drosophila lethal giant larvae ( lgl ) encodes a conserved tumor suppressor with established roles in cell polarity, asymmetric division, and proliferation control. Lgl's human orthologs, HUGL1 and HUGL2, are altered in human cancers, however, its mechanistic role as a tumor suppressor remains poorly understood. Based on a previously established connection between Lgl and Fragile X protein (FMRP), a miRNA-associated translational regulator, we hypothesized that Lgl may exert its role as a tumor suppressor by interacting with the miRNA pathway. Consistent with this model, we found that lgl is a dominant modifier of Argonaute1 overexpression in the eye neuroepithelium. Using microarray profiling we identified a core set of ten miRNAs that are altered throughout tumorigenesis in Drosophila lgl mutants. Among these are several miRNAs previously linked to human cancers including miR-9a , which we found to be downregulated in lgl neuroepithelial tissues. To determine whether miR-9a can act as an effector of Lgl in vivo , we overexpressed it in the context of lgl knock-down by RNAi and found it able to reduce the overgrowth phenotype caused by Lgl loss in epithelia. Furthermore, cross-comparisons between miRNA and mRNA profiling in lgl mutant tissues and human breast cancer cells identified thrombospondin ( tsp ) as a common factor altered in both fly and human breast cancer tumorigenesis models. Our work provides the first evidence of a functional connection between Lgl and the miRNA pathway, demonstrates that miR-9a mediates Lgl's role in restricting epithelial proliferation, and provides novel insights into pathways controlled by Lgl during tumor progression., Competing Interests: Competing interestsThe authors declare no competing or financial interests., (© 2018. Published by The Company of Biologists Ltd.)
- Published
- 2018
- Full Text
- View/download PDF
44. Phosphorylation of Drosophila Brahma on CDK-phosphorylation sites is important for cell cycle regulation and differentiation.
- Author
-
Roesley SNA, La Marca JE, Deans AJ, Mckenzie L, Suryadinata R, Burke P, Portela M, Wang H, Bernard O, Sarcevic B, and Richardson HE
- Subjects
- Alleles, Animals, Cell Death, Epistasis, Genetic, Epithelium metabolism, Mutation genetics, Phosphorylation, S Phase, Signal Transduction, Wings, Animal growth & development, Cell Cycle, Cell Cycle Proteins metabolism, Cell Differentiation, Cyclin-Dependent Kinases metabolism, Drosophila Proteins metabolism, Drosophila melanogaster cytology, Drosophila melanogaster metabolism, Trans-Activators metabolism
- Abstract
The SWI/SNF ATP-dependent chromatin-remodeling complex is an important evolutionarily conserved regulator of cell cycle progression. It associates with the Retinoblastoma (pRb)/HDAC/E2F/DP transcription complex to modulate cell cycle-dependent gene expression. The key catalytic component of the SWI/SNF complex in mammals is the ATPase subunit, Brahma (BRM) or BRG1. BRG1 was previously shown to be phosphorylated by the G
1 -S phase cell cycle regulatory kinase Cyclin E/CDK2 in vitro, which was associated with the bypass of G1 arrest conferred by BRG1 expression. However, it is unknown whether direct Cyclin E/CDK2-mediated phosphorylation of BRM/BRG1 is important for G1 -S phase cell cycle progression and proliferation in vivo. Herein, we demonstrate for the first time the importance of CDK-mediated phosphorylation of Brm in cell proliferation and differentiation in vivo using the Drosophila melanogaster model organism. Expression of a CDK-site phospho-mimic mutant of Brm, brm-ASP (all the potential CDK sites are mutated from Ser/Thr to Asp), which acts genetically as a brm loss-of-function allele, dominantly accelerates progression into the S phase, and bypasses a Retinoblastoma-induced developmental G1 phase arrest in the wing epithelium. Conversely, expression of a CDK-site phospho-blocking mutation of Brm, brm-ALA, acts genetically as a brm gain-of-function mutation, and in a Brm complex compromised background reduces S phase cells. Expression of the brm phospho-mutants also affected differentiation and Decapentaplegic (BMP/TGFβ) signaling in the wing epithelium. Altogether our results show that CDK-mediated phosphorylation of Brm is important in G1 -S phase regulation and differentiation in vivo., Abbreviations: A-P: Anterior-Posterior; BAF: BRG1-associated factor; BMP: Bone Morphogenetic Protein; Brg1: Brahma-Related Gene 1; Brm: Brahma; BSA: Bovine Serum Albumin; CDK: Cyclin dependent kinase dpp: decapentaplegic; EdU: 5-Ethynyl 2'-DeoxyUridine; EGFR: Epidermal Growth Factor Receptor; en: engrailed; GFP: Green Fluorescent Protein; GST: Glutathione-S-Transferase; HDAC: Histone DeACetylase; JNK: c-Jun N-terminal Kinase; Mad: Mothers Against Dpp; MAPK: Mitogen Activated Protein Kinase; MB:: Myelin Basic Protein; nub: nubbin; pH3: phosphorylated Histone H3; PBS: Phosphate Buffered Saline; PBT: PBS Triton; PFA: ParaFormAldehydep; Rb: Retinoblastoma protein; PCV: Posterior Cross-Vein; Snr1: Snf5-Related 1; SWI/SNF: SWitch/Sucrose Non-Fermentable; TGFβ: Transforming Growth Factor β; TUNEL: TdT-mediated dUTP Nick End Labelling; Wg: Wingless; ZNC: Zone of Non-Proliferating Cells.- Published
- 2018
- Full Text
- View/download PDF
45. Autophagy suppresses Ras-driven epithelial tumourigenesis by limiting the accumulation of reactive oxygen species.
- Author
-
Manent J, Banerjee S, de Matos Simoes R, Zoranovic T, Mitsiades C, Penninger JM, Simpson KJ, Humbert PO, and Richardson HE
- Subjects
- Adaptor Proteins, Signal Transducing metabolism, Animals, Apoptosis Regulatory Proteins, Cell Transformation, Neoplastic, Drosophila melanogaster, Female, Gene Knockdown Techniques, Humans, MAP Kinase Signaling System, Male, Microtubule-Associated Proteins metabolism, Oxidative Stress, Survival Analysis, Vesicle-Associated Membrane Protein 2 metabolism, Autophagy genetics, Carcinogenesis genetics, Genes, ras, Reactive Oxygen Species metabolism
- Abstract
Activation of Ras signalling occurs in ~30% of human cancers; however, activated Ras alone is not sufficient for tumourigenesis. In a screen for tumour suppressors that cooperate with oncogenic Ras (Ras
V12 ) in Drosophila, we identified genes involved in the autophagy pathway. Bioinformatic analysis of human tumours revealed that several core autophagy genes, including GABARAP, correlate with oncogenic KRAS mutations and poor prognosis in human pancreatic cancer, supporting a potential tumour-suppressive effect of the pathway in Ras-driven human cancers. In Drosophila, we demonstrate that blocking autophagy at any step of the pathway enhances RasV12 -driven epithelial tissue overgrowth via the accumulation of reactive oxygen species and activation of the Jun kinase stress response pathway. Blocking autophagy in RasV12 clones also results in non-cell-autonomous effects with autophagy, cell proliferation and caspase activation induced in adjacent wild-type cells. Our study has implications for understanding the interplay between perturbations in Ras signalling and autophagy in tumourigenesis, which might inform the development of novel therapeutics targeting Ras-driven cancers.- Published
- 2017
- Full Text
- View/download PDF
46. Tissue growth and tumorigenesis in Drosophila: cell polarity and the Hippo pathway.
- Author
-
Richardson HE and Portela M
- Subjects
- Animals, Carcinogenesis, Cell Movement, Drosophila melanogaster metabolism, Epithelial Cells metabolism, Humans, Models, Animal, Cell Polarity, Drosophila Proteins metabolism, Drosophila melanogaster cytology, Intracellular Signaling Peptides and Proteins metabolism, Protein Serine-Threonine Kinases metabolism, Signal Transduction
- Abstract
Cell polarity regulation is critical for defining membrane domains required for the establishment and maintenance of the apical-basal axis in epithelial cells (apico-basal polarity), asymmetric cell divisions, planar organization of tissues (planar cell polarity), and the formation of the front-rear axis in cell migration (front-rear polarity). In the vinegar fly, Drosophila melanogaster, cell polarity regulators also interact with the Hippo tissue growth control signaling pathway. In this review we survey the recent Drosophila literature linking cell polarity regulators with the Hippo pathway in epithelial tissue growth, neural stem cell asymmetric divisions and in cell migration in physiological and tumorigenic settings., (Copyright © 2017 Elsevier Ltd. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
47. A Kinome RNAi Screen in Drosophila Identifies Novel Genes Interacting with Lgl, aPKC, and Crb Cell Polarity Genes in Epithelial Tissues.
- Author
-
Parsons LM, Grzeschik NA, Amaratunga K, Burke P, Quinn LM, and Richardson HE
- Subjects
- Animals, Drosophila Proteins metabolism, Female, Gene Ontology, Genes, Modifier, Male, Membrane Proteins genetics, Membrane Proteins metabolism, Organ Size genetics, Signal Transduction genetics, Tumor Suppressor Proteins genetics, Tumor Suppressor Proteins metabolism, Wings, Animal anatomy & histology, Cell Polarity genetics, Drosophila Proteins genetics, Drosophila melanogaster genetics, Epistasis, Genetic, Epithelium metabolism, Genes, Insect, Genetic Testing, RNA Interference
- Abstract
In both Drosophila melanogaster and mammalian systems, epithelial structure and underlying cell polarity are essential for proper tissue morphogenesis and organ growth. Cell polarity interfaces with multiple cellular processes that are regulated by the phosphorylation status of large protein networks. To gain insight into the molecular mechanisms that coordinate cell polarity with tissue growth, we screened a boutique collection of RNAi stocks targeting the kinome for their capacity to modify Drosophila "cell polarity" eye and wing phenotypes. Initially, we identified kinase or phosphatase genes whose depletion modified adult eye phenotypes associated with the manipulation of cell polarity complexes (via overexpression of Crb or aPKC). We next conducted a secondary screen to test whether these cell polarity modifiers altered tissue overgrowth associated with depletion of Lgl in the wing. These screens identified Hippo, Jun kinase (JNK), and Notch signaling pathways, previously linked to cell polarity regulation of tissue growth. Furthermore, novel pathways not previously connected to cell polarity regulation of tissue growth were identified, including Wingless (Wg/Wnt), Ras, and lipid/Phospho-inositol-3-kinase (PI3K) signaling pathways. Additionally, we demonstrated that the "nutrient sensing" kinases Salt Inducible Kinase 2 and 3 ( SIK2 and 3 ) are potent modifiers of cell polarity phenotypes and regulators of tissue growth. Overall, our screen has revealed novel cell polarity-interacting kinases and phosphatases that affect tissue growth, providing a platform for investigating molecular mechanisms coordinating cell polarity and tissue growth during development., (Copyright © 2017 Parsons et al.)
- Published
- 2017
- Full Text
- View/download PDF
48. Differential regulation of protein tyrosine kinase signalling by Dock and the PTP61F variants.
- Author
-
Willoughby LF, Manent J, Allan K, Lee H, Portela M, Wiede F, Warr C, Meng TC, Tiganis T, and Richardson HE
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Animals, Drosophila Proteins genetics, Drosophila melanogaster enzymology, Drosophila melanogaster genetics, Drosophila melanogaster growth & development, ErbB Receptors genetics, ErbB Receptors metabolism, Gene Expression Regulation, Male, Mutation, Nerve Tissue Proteins genetics, Protein Tyrosine Phosphatases, Non-Receptor genetics, Receptor Protein-Tyrosine Kinases genetics, STAT Transcription Factors genetics, STAT Transcription Factors metabolism, Adaptor Proteins, Signal Transducing metabolism, Drosophila Proteins metabolism, Drosophila melanogaster metabolism, Nerve Tissue Proteins metabolism, Protein Tyrosine Phosphatases, Non-Receptor metabolism, Receptor Protein-Tyrosine Kinases metabolism, Signal Transduction
- Abstract
Tyrosine phosphorylation-dependent signalling is coordinated by the opposing actions of protein tyrosine kinases (PTKs) and protein tyrosine phosphatases (PTPs). There is a growing list of adaptor proteins that interact with PTPs and facilitate the dephosphorylation of substrates. The extent to which any given adaptor confers selectivity for any given substrate in vivo remains unclear. Here we have taken advantage of Drosophila melanogaster as a model organism to explore the influence of the SH3/SH2 adaptor protein Dock on the abilities of the membrane (PTP61Fm)- and nuclear (PTP61Fn)-targeted variants of PTP61F (the Drosophila othologue of the mammalian enzymes PTP1B and TCPTP respectively) to repress PTK signalling pathways in vivo. PTP61Fn effectively repressed the eye overgrowth associated with activation of the epidermal growth factor receptor (EGFR), PTK, or the expression of the platelet-derived growth factor/vascular endothelial growth factor receptor (PVR) or insulin receptor (InR) PTKs. PTP61Fn repressed EGFR and PVR-induced mitogen-activated protein kinase signalling and attenuated PVR-induced STAT92E signalling. By contrast, PTP61Fm effectively repressed EGFR- and PVR-, but not InR-induced tissue overgrowth. Importantly, coexpression of Dock with PTP61F allowed for the efficient repression of the InR-induced eye overgrowth, but did not enhance the PTP61Fm-mediated inhibition of EGFR and PVR-induced signalling. Instead, Dock expression increased, and PTP61Fm coexpression further exacerbated the PVR-induced eye overgrowth. These results demonstrate that Dock selectively enhances the PTP61Fm-mediated attenuation of InR signalling and underscores the specificity of PTPs and the importance of adaptor proteins in regulating PTP function in vivo., (© 2017 Federation of European Biochemical Societies.)
- Published
- 2017
- Full Text
- View/download PDF
49. Robo-Enabled Tumor Cell Extrusion.
- Author
-
Richardson HE and Portela M
- Subjects
- Epithelium metabolism, Humans, Signal Transduction, Nerve Tissue Proteins metabolism, Receptors, Immunologic metabolism
- Abstract
How aberrant cells are removed from a tissue to prevent tumor formation is a key question in cancer biology. Reporting in this issue of Developmental Cell, Vaughen and Igaki (2016) show that a pathway with an important role in neural guidance also directs extrusion of tumor cells from epithelial tissues., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
50. BTB-Zinc Finger Oncogenes Are Required for Ras and Notch-Driven Tumorigenesis in Drosophila.
- Author
-
Doggett K, Turkel N, Willoughby LF, Ellul J, Murray MJ, Richardson HE, and Brumby AM
- Subjects
- Animals, Animals, Genetically Modified, Drosophila Proteins genetics, Drosophila melanogaster, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Microarray Analysis, Nuclear Proteins chemistry, Nuclear Proteins genetics, Protein Interaction Domains and Motifs genetics, Carcinogenesis genetics, Drosophila Proteins physiology, Genes, ras physiology, Oncogenes physiology, Receptors, Notch physiology, Zinc Fingers genetics
- Abstract
During tumorigenesis, pathways that promote the epithelial-to-mesenchymal transition (EMT) can both facilitate metastasis and endow tumor cells with cancer stem cell properties. To gain a greater understanding of how these properties are interlinked in cancers we used Drosophila epithelial tumor models, which are driven by orthologues of human oncogenes (activated alleles of Ras and Notch) in cooperation with the loss of the cell polarity regulator, scribbled (scrib). Within these tumors, both invasive, mesenchymal-like cell morphology and continual tumor overgrowth, are dependent upon Jun N-terminal kinase (JNK) activity. To identify JNK-dependent changes within the tumors we used a comparative microarray analysis to define a JNK gene signature common to both Ras and Notch-driven tumors. Amongst the JNK-dependent changes was a significant enrichment for BTB-Zinc Finger (ZF) domain genes, including chronologically inappropriate morphogenesis (chinmo). chinmo was upregulated by JNK within the tumors, and overexpression of chinmo with either RasV12 or Nintra was sufficient to promote JNK-independent epithelial tumor formation in the eye/antennal disc, and, in cooperation with RasV12, promote tumor formation in the adult midgut epithelium. Chinmo primes cells for oncogene-mediated transformation through blocking differentiation in the eye disc, and promoting an escargot-expressing stem or enteroblast cell state in the adult midgut. BTB-ZF genes are also required for Ras and Notch-driven overgrowth of scrib mutant tissue, since, although loss of chinmo alone did not significantly impede tumor development, when loss of chinmo was combined with loss of a functionally related BTB-ZF gene, abrupt, tumor overgrowth was significantly reduced. abrupt is not a JNK-induced gene, however, Abrupt is present in JNK-positive tumor cells, consistent with a JNK-associated oncogenic role. As some mammalian BTB-ZF proteins are also highly oncogenic, our work suggests that EMT-promoting signals in human cancers could similarly utilize networks of these proteins to promote cancer stem cell states.
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.