1
|
Wang T, Fu J, Huang Y, Fu C. Mechanism of APC truncation involved in colorectal cancer tumorigenesis (Review). Oncol Lett 2025; 29:2. [PMID: 39526304 PMCID: PMC11544694 DOI: 10.3892/ol.2024.14748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 09/26/2024] [Indexed: 11/16/2024] Open
Abstract
Adenomatous polyposis coli (APC) is widely recognized as a heavily mutated gene that suppresses tumor growth in colorectal cancer (CRC). Its mutation is considered to be the primary and early event that occurs in the development of CRC. In addition, APC has a crucial role in inhibiting the canonical Wnt signaling pathway. APC mutations in CRC result in the production of shortened gene products. This impairment of β-catenin destruction complexes causes an accumulation of active β-catenin in the cytoplasm and nucleus. In these compartments, β-catenin can bind with DNA-binding proteins of the transcription factor/lymphoid enhancer-binding factor family, thereby activating the Wnt signaling pathway. Consequently, the balance of numerous cellular processes is disrupted, ultimately driving the formation of tumors. There is a growing body of evidence indicating the prevalent occurrence of APC truncation in the majority of CRC cases. Furthermore, it has been observed that these truncated proteins have a crucial role in the activation of the Wnt signaling pathway and the subsequent loss of tumor inhibitory function. This review aimed to provide an overview of the recent advancements in understanding the mechanism behind APC truncation and its association with the onset and progression of CRC.
Collapse
Affiliation(s)
- Tuya Wang
- Department of Medicine, Hetao College, Bayannur, Inner Mongolia Autonomous Region 015000, P.R. China
| | - Jing Fu
- Department of Medicine, Hetao College, Bayannur, Inner Mongolia Autonomous Region 015000, P.R. China
| | - Ye Huang
- Department of Medicine, Hetao College, Bayannur, Inner Mongolia Autonomous Region 015000, P.R. China
| | - Chun Fu
- Department of Medicine, Hetao College, Bayannur, Inner Mongolia Autonomous Region 015000, P.R. China
| |
Collapse
|
2
|
Falconi J, Strobel K, Djiane A, Lassus P. [Drosophila as a model to study cancer biology]. Bull Cancer 2024; 111:880-892. [PMID: 38960821 DOI: 10.1016/j.bulcan.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/18/2024] [Accepted: 05/03/2024] [Indexed: 07/05/2024]
Abstract
The rising global incidence of cancer makes it the second leading cause of death worldwide. Over the past decades, significant progress has been made in both basic knowledge and the discovery of new therapeutic approaches. However, the complexity of mechanisms related to tumor development requires the use of sophisticated and adapted research tools. Among these, the fruitfly Drosophila melanogaster represents a powerful genetic model with numerous practical and conceptual advantages. Indeed, the conservation of genes implicated in cancer between this insect and mammals places Drosophila as a crucial genetic tool for understanding the fundamental mechanisms governing tumorigenesis and identifying new therapeutic targets. This review aims to describe this original model and demonstrate its relevance for studying cancer biology.
Collapse
Affiliation(s)
- Jennifer Falconi
- IRCM, Inserm, ICM, université de Montpellier, Montpellier, France
| | - Katrin Strobel
- IRCM, Inserm, ICM, université de Montpellier, Montpellier, France
| | - Alexandre Djiane
- IRCM, Inserm, ICM, université de Montpellier, Montpellier, France
| | - Patrice Lassus
- IRCM, Inserm U1194, ICM, CNRS, université de Montpellier, 208, rue des Apothicaires, 34298 Montpellier cedex, France.
| |
Collapse
|
3
|
Zhou J, Boutros M. Intestinal stem cells and their niches in homeostasis and disease. Cells Dev 2023; 175:203862. [PMID: 37271243 DOI: 10.1016/j.cdev.2023.203862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 05/21/2023] [Accepted: 06/01/2023] [Indexed: 06/06/2023]
Abstract
Tissues such as the intestine harbor stem cells that have remarkable functional plasticity in response to a dynamic environment. To adapt to the environment, stem cells constantly receive information from their surrounding microenvironment (also called the 'niche') that instructs them how to adapt to changes. The Drosophila midgut shows morphological and functional similarities to the mammalian small intestine and has been a useful model system to study signaling events in stem cells and tissue homeostasis. In this review, we summarize the current understanding of the Drosophila midgut regarding how stem cells communicate with microenvironmental niches including enteroblasts, enterocytes, enteroendocrine cells and visceral muscles to coordinate tissue regeneration and homeostasis. In addition, distant cells such as hemocytes or tracheal cells have been shown to interact with stem cells and influence the development of intestinal diseases. We discuss the contribution of stem cell niches in driving or counteracting disease progression, and review conceptual advances derived from the Drosophila intestine as a model for stem cell biology.
Collapse
Affiliation(s)
- Jun Zhou
- German Cancer Research Center (DKFZ), Heidelberg University, Division Signaling and Functional Genomics, BioQuant and Medical Faculty Mannheim, D-69120 Heidelberg, Germany; School of Biomedical Sciences, Hunan University, Changsha, China.
| | - Michael Boutros
- German Cancer Research Center (DKFZ), Heidelberg University, Division Signaling and Functional Genomics, BioQuant and Medical Faculty Mannheim, D-69120 Heidelberg, Germany.
| |
Collapse
|
4
|
Sharpe JL, Morgan J, Nisbet N, Campbell K, Casali A. Modelling Cancer Metastasis in Drosophila melanogaster. Cells 2023; 12:cells12050677. [PMID: 36899813 PMCID: PMC10000390 DOI: 10.3390/cells12050677] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/14/2023] [Accepted: 02/17/2023] [Indexed: 02/24/2023] Open
Abstract
Cancer metastasis, the process by which tumour cells spread throughout the body and form secondary tumours at distant sites, is the leading cause of cancer-related deaths. The metastatic cascade is a highly complex process encompassing initial dissemination from the primary tumour, travel through the blood stream or lymphatic system, and the colonisation of distant organs. However, the factors enabling cells to survive this stressful process and adapt to new microenvironments are not fully characterised. Drosophila have proven a powerful system in which to study this process, despite important caveats such as their open circulatory system and lack of adaptive immune system. Historically, larvae have been used to model cancer due to the presence of pools of proliferating cells in which tumours can be induced, and transplanting these larval tumours into adult hosts has enabled tumour growth to be monitored over longer periods. More recently, thanks largely to the discovery that there are stem cells in the adult midgut, adult models have been developed. We focus this review on the development of different Drosophila models of metastasis and how they have contributed to our understanding of important factors determining metastatic potential, including signalling pathways, the immune system and the microenvironment.
Collapse
Affiliation(s)
- Joanne L. Sharpe
- School of Biosciences, The University of Sheffield, Sheffield S10 2TN, UK
| | - Jason Morgan
- School of Biosciences, The University of Sheffield, Sheffield S10 2TN, UK
| | - Nicholas Nisbet
- School of Biosciences, The University of Sheffield, Sheffield S10 2TN, UK
| | - Kyra Campbell
- School of Biosciences, The University of Sheffield, Sheffield S10 2TN, UK
- Correspondence: (K.C.); (A.C.)
| | - Andreu Casali
- Departament de Ciències Mèdiques Bàsiques, Universitat de Lleida and IRBLleida, Av. Alcalde Rovira Roure, 80, 25198 Lleida, Spain
- Correspondence: (K.C.); (A.C.)
| |
Collapse
|
5
|
Duneau D, Buchon N. Gut cancer increases the risk of Drosophila being preyed upon by hunting spiders. Anim Behav 2022. [DOI: 10.1016/j.anbehav.2022.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
|
6
|
Munnik C, Xaba MP, Malindisa ST, Russell BL, Sooklal SA. Drosophila melanogaster: A platform for anticancer drug discovery and personalized therapies. Front Genet 2022; 13:949241. [PMID: 36003330 PMCID: PMC9393232 DOI: 10.3389/fgene.2022.949241] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 07/06/2022] [Indexed: 12/12/2022] Open
Abstract
Cancer is a complex disease whereby multiple genetic aberrations, epigenetic modifications, metabolic reprogramming, and the microenvironment contribute to the development of a tumor. In the traditional anticancer drug discovery pipeline, drug candidates are usually screened in vitro using two-dimensional or three-dimensional cell culture. However, these methods fail to accurately mimic the human disease state. This has led to the poor success rate of anticancer drugs in the preclinical stages since many drugs are abandoned due to inefficacy or toxicity when transitioned to whole-organism models. The common fruit fly, Drosophila melanogaster, has emerged as a beneficial system for modeling human cancers. Decades of fundamental research have shown the evolutionary conservation of key genes and signaling pathways between flies and humans. Moreover, Drosophila has a lower genetic redundancy in comparison to mammals. These factors, in addition to the advancement of genetic toolkits for manipulating gene expression, allow for the generation of complex Drosophila genotypes and phenotypes. Numerous studies have successfully created Drosophila models for colorectal, lung, thyroid, and brain cancers. These models were utilized in the high-throughput screening of FDA-approved drugs which led to the identification of several compounds capable of reducing proliferation and rescuing phenotypes. More noteworthy, Drosophila has also unlocked the potential for personalized therapies. Drosophila ‘avatars’ presenting the same mutations as a patient are used to screen multiple therapeutic agents targeting multiple pathways to find the most appropriate combination of drugs. The outcomes of these studies have translated to significant responses in patients with adenoid cystic carcinoma and metastatic colorectal cancers. Despite not being widely utilized, the concept of in vivo screening of drugs in Drosophila is making significant contributions to the current drug discovery pipeline. In this review, we discuss the application of Drosophila as a platform in anticancer drug discovery; with special focus on the cancer models that have been generated, drug libraries that have been screened and the status of personalized therapies. In addition, we elaborate on the biological and technical limitations of this system.
Collapse
Affiliation(s)
- Chamoné Munnik
- Department of Life and Consumer Sciences, University of South Africa, Pretoria, South Africa
| | - Malungi P. Xaba
- Department of Life and Consumer Sciences, University of South Africa, Pretoria, South Africa
| | - Sibusiso T. Malindisa
- Department of Life and Consumer Sciences, University of South Africa, Pretoria, South Africa
| | - Bonnie L. Russell
- Department of Life and Consumer Sciences, University of South Africa, Pretoria, South Africa
- Buboo (Pty) Ltd, The Innovation Hub, Pretoria, South Africa
| | - Selisha A. Sooklal
- Department of Life and Consumer Sciences, University of South Africa, Pretoria, South Africa
- *Correspondence: Selisha A. Sooklal,
| |
Collapse
|
7
|
Swoboda J, Mittelsdorf P, Chen Y, Weiskirchen R, Stallhofer J, Schüle S, Gassler N. Intestinal Wnt in the transition from physiology to oncology. World J Clin Oncol 2022; 13:168-185. [PMID: 35433295 PMCID: PMC8966512 DOI: 10.5306/wjco.v13.i3.168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 09/07/2021] [Accepted: 02/20/2022] [Indexed: 02/06/2023] Open
Abstract
Adult stem cells are necessary for self-renewal tissues and regeneration after damage. Especially in the intestine, which self-renews every few days, they play a key role in tissue homeostasis. Therefore, complex regulatory mechanisms are needed to prevent hyperproliferation, which can lead in the worst case to carcinogenesis or under-activation of stem cells, which can result in dysfunctional epithelial. One main regulatory signaling pathway is the Wnt/β-catenin signaling pathway. It is a highly conserved pathway, with β-catenin, a transcription factor, as target protein. Translocation of β-catenin from cytoplasm to nucleus activates the transcription of numerous genes involved in regulating stem cell pluripo-tency, proliferation, cell differentiation and regulation of cell death. This review presents a brief overview of the Wnt/β-catenin signaling pathway, the regulatory mechanism of this pathway and its role in intestinal homeostasis. Additionally, this review highlights the molecular mechanisms and the histomorphological features of Wnt hyperactivation. Furthermore, the central role of the Wnt signaling pathway in intestinal carcinogenesis as well as its clinical relevance in colorectal carcinoma are discussed.
Collapse
Affiliation(s)
- Julia Swoboda
- Section Pathology, Institute of Forensic Medicine, Jena University Hospital, Jena 07747, Germany
| | - Patrick Mittelsdorf
- Section Pathology, Institute of Forensic Medicine, Jena University Hospital, Jena 07747, Germany
| | - Yuan Chen
- Section Pathology, Institute of Forensic Medicine, Jena University Hospital, Jena 07747, Germany
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH University Hospital Aachen, Aachen 52074, Germany
| | - Johannes Stallhofer
- Department of Internal Medicine IV (Gastroenterology, Hepatology, and Infectious Diseases), Jena University Hospital, Jena 07747, Germany
| | - Silke Schüle
- Department of General, Visceral and Vascular Surgery, Jena University Hospital, Jena 07747, Germany
| | - Nikolaus Gassler
- Section Pathology, Institute of Forensic Medicine, Jena University Hospital, Jena 07747, Germany
| |
Collapse
|
8
|
Conditional CRISPR-Cas Genome Editing in Drosophila to Generate Intestinal Tumors. Cells 2021; 10:cells10113156. [PMID: 34831379 PMCID: PMC8620722 DOI: 10.3390/cells10113156] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/07/2021] [Accepted: 11/10/2021] [Indexed: 01/19/2023] Open
Abstract
CRISPR-Cas has revolutionized genetics and extensive efforts have been made to enhance its editing efficiency by developing increasingly more elaborate tools. Here, we evaluate the CRISPR-Cas9 system in Drosophila melanogaster to assess its ability to induce stem cell-derived tumors in the intestine. We generated conditional tissue-specific CRISPR knockouts using different Cas9 expression vectors with guide RNAs targeting the BMP, Notch, and JNK pathways in intestinal progenitors such as stem cells (ISCs) and enteroblasts (EBs). Perturbing Notch and BMP signaling increased the proliferation of ISCs/EBs and resulted in the formation of intestinal tumors, albeit with different efficiencies. By assessing both the anterior and posterior regions of the midgut, we observed regional differences in ISC/EB proliferation and tumor formation upon mutagenesis. Surprisingly, high continuous expression of Cas9 in ISCs/EBs blocked age-dependent increase in ISCs/EBs proliferation and when combined with gRNAs targeting tumor suppressors, it prevented tumorigenesis. However, no such effects were seen when temporal parameters of Cas9 were adjusted to regulate its expression levels or with a genetically modified version, which expresses Cas9 at lower levels, suggesting that fine-tuning Cas9 expression is essential to avoid deleterious effects. Our findings suggest that modifications to Cas9 expression results in differences in editing efficiency and careful considerations are required when choosing reagents for CRISPR-Cas9 mutagenesis studies. In summary, Drosophila can serve as a powerful model for context-dependent CRISPR-Cas based perturbations and to test genome-editing systems in vivo.
Collapse
|
9
|
Gong S, Zhang Y, Tian A, Deng W. Tumor models in various Drosophila tissues. WIREs Mech Dis 2021; 13:e1525. [PMID: 34730289 PMCID: PMC8566734 DOI: 10.1002/wsbm.1525] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 01/07/2023]
Abstract
The development of cancer is a complex multistage process. Over the past few decades, the model organism Drosophila melanogaster has been crucial in identifying cancer-related genes and pathways and elucidating mechanisms underlying growth regulation in development. Investigations using Drosophila has yielded new insights into the molecular mechanisms involved in tumor initiation and progression. In this review, we describe various tumor models that have been developed in recent years using different Drosophila tissues, such as the imaginal tissue, the neural tissue, the gut, the ovary, and hematopoietic cells. We discuss underlying genetic alterations, cancer-like characteristics, as well as similarities and key differences among these models. We also discuss how disruptions in stem cell division and differentiation result in tumor formation in diverse tissues, and highlight new concepts developed using the fly model to understand context-dependent tumorigenesis. We further discuss the progress made in Drosophila to explore tumor-host interactions that involve the innate immune response to tumor growth and the cachexia wasting phenotype. This article is categorized under: Cancer > Genetics/Genomics/Epigenetics Cancer > Stem Cells and Development Cancer > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Shangyu Gong
- Department of Biochemistry and Molecular BiologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - Yichi Zhang
- Department of Biochemistry and Molecular BiologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - Aiguo Tian
- Department of Biochemistry and Molecular BiologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - Wu‐Min Deng
- Department of Biochemistry and Molecular BiologyTulane University School of MedicineNew OrleansLouisianaUSA
| |
Collapse
|
10
|
Sensitive High-Throughput Assays for Tumour Burden Reveal the Response of a Drosophila melanogaster Model of Colorectal Cancer to Standard Chemotherapies. Int J Mol Sci 2021; 22:ijms22105101. [PMID: 34065887 PMCID: PMC8151205 DOI: 10.3390/ijms22105101] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/30/2021] [Accepted: 05/05/2021] [Indexed: 01/04/2023] Open
Abstract
Drosophila melanogaster (Drosophila) models of cancer are emerging as powerful tools to investigate the basic mechanisms underlying tumour progression and identify novel therapeutics. Rapid and inexpensive, it is possible to carry out genetic and drug screens at a far larger scale than in vertebrate organisms. Such whole-organism-based drug screens permits assessment of drug absorption and toxicity, reducing the possibility of false positives. Activating mutations in the Wnt and Ras signalling pathways are common in many epithelial cancers, and when driven in the adult Drosophila midgut, it induces aggressive intestinal tumour-like outgrowths that recapitulate many aspects of human colorectal cancer (CRC). Here we have taken a Drosophila CRC model in which tumourous cells are marked with both GFP and luciferase reporter genes, and developed novel high-throughput assays for quantifying tumour burden. Leveraging these assays, we find that the Drosophila CRC model responds rapidly to treatment with standard CRC-drugs, opening the door to future rapid genetic and drug screens.
Collapse
|
11
|
Ferguson M, Foley E. Microbial recognition regulates intestinal epithelial growth in homeostasis and disease. FEBS J 2021; 289:3666-3691. [PMID: 33977656 DOI: 10.1111/febs.15910] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 04/06/2021] [Accepted: 04/30/2021] [Indexed: 12/13/2022]
Abstract
The intestine is constantly exposed to a dynamic community of microbes. Intestinal epithelial cells respond to microbes through evolutionarily conserved recognition pathways, such as the immune deficiency (IMD) pathway of Drosophila, the Toll-like receptor (TLR) response of flies and vertebrates, and the vertebrate nucleotide-binding oligomerization domain (NOD) pathway. Microbial recognition pathways are tightly controlled to respond effectively to pathogens, tolerate the microbiome, and limit intestinal disease. In this review, we focus on contributions of different model organisms to our understanding of how epithelial microbe recognition impacts intestinal proliferation and differentiation in homeostasis and disease. In particular, we compare how microbes and subsequent recognition by the intestine influences barrier integrity, intestinal repair and tumorigenesis in Drosophila, zebrafish, mice, and organoids. In addition, we discuss the importance of microbial recognition in homeostatic intestinal growth and discuss how immune pathways directly impact stem cell and crypt dynamics.
Collapse
Affiliation(s)
- Meghan Ferguson
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.,Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Edan Foley
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.,Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
12
|
Morris O, Deng H, Tam C, Jasper H. Warburg-like Metabolic Reprogramming in Aging Intestinal Stem Cells Contributes to Tissue Hyperplasia. Cell Rep 2020; 33:108423. [PMID: 33238124 PMCID: PMC8011352 DOI: 10.1016/j.celrep.2020.108423] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 09/28/2020] [Accepted: 11/02/2020] [Indexed: 12/28/2022] Open
Abstract
In many tissues, stem cell (SC) proliferation is dynamically adjusted to regenerative needs. How SCs adapt their metabolism to meet the demands of proliferation and how changes in such adaptive mechanisms contribute to age-related dysfunction remain poorly understood. Here, we identify mitochondrial Ca2+ uptake as a central coordinator of SC metabolism. Live imaging of genetically encoded metabolite sensors in intestinal SCs (ISCs) of Drosophila reveals that mitochondrial Ca2+ uptake transiently adapts electron transport chain flux to match energetic demand upon proliferative activation. This tight metabolic adaptation is lost in ISCs of old flies, as declines in mitochondrial Ca2+ uptake promote a "Warburg-like" metabolic reprogramming toward aerobic glycolysis. This switch mimics metabolic reprogramming by the oncogene RasV12 and enhances ISC hyperplasia. Our data identify a critical mechanism for metabolic adaptation of tissue SCs and reveal how its decline sets aging SCs on a metabolic trajectory reminiscent of that seen upon oncogenic transformation.
Collapse
Affiliation(s)
- Otto Morris
- Immunology Discovery, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Hansong Deng
- Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 20092, China; Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945-1400, USA
| | - Christine Tam
- Department of Biomolecular Resources, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Heinrich Jasper
- Immunology Discovery, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA; Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945-1400, USA.
| |
Collapse
|
13
|
Dissemination of Ras V12-transformed cells requires the mechanosensitive channel Piezo. Nat Commun 2020; 11:3568. [PMID: 32678085 PMCID: PMC7366633 DOI: 10.1038/s41467-020-17341-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 06/24/2020] [Indexed: 12/31/2022] Open
Abstract
Dissemination of transformed cells is a key process in metastasis. Despite its importance, how transformed cells disseminate from an intact tissue and enter the circulation is poorly understood. Here, we use a fully developed tissue, Drosophila midgut, and describe the morphologically distinct steps and the cellular events occurring over the course of RasV12-transformed cell dissemination. Notably, RasV12-transformed cells formed the Actin- and Cortactin-rich invasive protrusions that were important for breaching the extracellular matrix (ECM) and visceral muscle. Furthermore, we uncovered the essential roles of the mechanosensory channel Piezo in orchestrating dissemination of RasV12-transformed cells. Collectively, our study establishes an in vivo model for studying how transformed cells migrate out from a complex tissue and provides unique insights into the roles of Piezo in invasive cell behavior. Drosophila tumours can be utilised to study the mechanisms of cell dissemination. Here, the authors use Drosophila midgut to examine the course of RasV12-transformed cell dissemination from midgut into circulation, which requires the actions of invasive protrusions and the mechanosensitive channel Piezo.
Collapse
|
14
|
Ngo S, Liang J, Su YH, O'Brien LE. Disruption of EGF Feedback by Intestinal Tumors and Neighboring Cells in Drosophila. Curr Biol 2020; 30:1537-1546.e3. [PMID: 32243854 PMCID: PMC7409949 DOI: 10.1016/j.cub.2020.01.082] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 12/11/2019] [Accepted: 01/29/2020] [Indexed: 12/20/2022]
Abstract
In healthy adult organs, robust feedback mechanisms control cell turnover to enforce homeostatic equilibrium between cell division and death [1, 2]. Nascent tumors must subvert these mechanisms to achieve cancerous overgrowth [3-7]. Elucidating the nature of this subversion can reveal how cancers become established and may suggest strategies to prevent tumor progression. In adult Drosophila intestine, a well-studied model of homeostatic cell turnover, the linchpin of cell equilibrium is feedback control of the epidermal growth factor (EGF) protease Rhomboid (Rho). Expression of Rho in apoptotic cells enables them to secrete EGFs, which stimulate nearby stem cells to undergo replacement divisions [8]. As in mammals, loss of adenomatous polyposis coli (APC) causes Drosophila intestinal stem cells to form adenomas [9]. Here, we demonstrate that Drosophila APC-/- tumors trigger widespread Rho expression in non-apoptotic cells, resulting in chronic EGF signaling. Initially, nascent APC-/- tumors induce rho in neighboring wild-type cells via acute, non-autonomous activation of Jun N-terminal kinase (JNK). During later growth and multilayering, APC-/- tumors induce rho in tumor cells by autonomous downregulation of E-cadherin (E-cad) and consequent activity of p120-catenin. This sequential dysregulation of tumor non-autonomous and -autonomous EGF signaling converts tissue-level feedback into feed-forward activation that drives cancerous overgrowth. Because Rho, EGF receptor (EGFR), and E-cad are associated with colorectal cancer in humans [10-17], our findings may shed light on how human colorectal tumors progress.
Collapse
Affiliation(s)
- Sang Ngo
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jackson Liang
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Yu-Han Su
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lucy Erin O'Brien
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
15
|
Tsuda-Sakurai K, Kimura M, Miura M. Diphthamide modification of eEF2 is required for gut tumor-like hyperplasia induced by oncogenic Ras. Genes Cells 2019; 25:76-85. [PMID: 31828897 DOI: 10.1111/gtc.12742] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 10/27/2019] [Accepted: 11/20/2019] [Indexed: 01/16/2023]
Abstract
Eukaryotic elongation factor 2 (eEF2) undergoes a unique post-translational modification called diphthamidation. Although eEF2 diphthamidation is highly conserved, its pathophysiological function is still largely unknown. To elucidate the function of diphthamidation in tumor, we examined the involvement of diphthamidation pathway enzyme Dph5 in tumor progression in Drosophila adult gut. Expression of oncogenic RasV12 in gut intestinal stem cells (ISCs) and enteroblasts (EBs) causes hypertrophy and disruption of gut epithelia, and shortened life span. Knockdown of Dph5 ameliorated these pathogenic phenotypes. Dph5 is required for gross translation activation and high dMyc protein level in RasV12 tumor-like hyperplasia. Transcriptome analysis revealed that Dph5 is involved in the regulation of ribosome biogenesis genes. These results suggest that diphthamidation is required for translation activation partly through the regulation of ribosome biogenesis in Ras-induced tumor-like hyperplasia model in Drosophila gut.
Collapse
Affiliation(s)
- Kayoko Tsuda-Sakurai
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Masaki Kimura
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Masayuki Miura
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
16
|
Collective cell migration and metastases induced by an epithelial-to-mesenchymal transition in Drosophila intestinal tumors. Nat Commun 2019; 10:2311. [PMID: 31127094 PMCID: PMC6534551 DOI: 10.1038/s41467-019-10269-y] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 05/01/2019] [Indexed: 12/19/2022] Open
Abstract
Metastasis underlies the majority of cancer-related deaths yet remains poorly understood due, in part, to the lack of models in vivo. Here we show that expression of the EMT master inducer Snail in primary adult Drosophila intestinal tumors leads to the dissemination of tumor cells and formation of macrometastases. Snail drives an EMT in tumor cells, which, although retaining some epithelial markers, subsequently break through the basal lamina of the midgut, undergo a collective migration and seed polyclonal metastases. While metastases re-epithelialize over time, we found that early metastases are remarkably mesenchymal, discarding the requirement for a mesenchymal-to-epithelial transition for early stages of metastatic growth. Our results demonstrate the formation of metastases in adult flies, and identify a key role for partial-EMTs in driving it. This model opens the door to investigate the basic mechanisms underlying metastasis, in a powerful in vivo system suited for rapid genetic and drug screens. Modelling and visualizing tumor metastasis in Drosophila has been a challenge. Here, the authors show that constitutive expression of Sna in primary adult Drosophila intestinal tumors drives EMT and dissemination of tumor cells, induces collective cell migration and formation of polyclonal metastases.
Collapse
|
17
|
Mirzoyan Z, Sollazzo M, Allocca M, Valenza AM, Grifoni D, Bellosta P. Drosophila melanogaster: A Model Organism to Study Cancer. Front Genet 2019; 10:51. [PMID: 30881374 PMCID: PMC6405444 DOI: 10.3389/fgene.2019.00051] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 01/21/2019] [Indexed: 12/26/2022] Open
Abstract
Cancer is a multistep disease driven by the activation of specific oncogenic pathways concomitantly with the loss of function of tumor suppressor genes that act as sentinels to control physiological growth. The conservation of most of these signaling pathways in Drosophila, and the ability to easily manipulate them genetically, has made the fruit fly a useful model organism to study cancer biology. In this review we outline the basic mechanisms and signaling pathways conserved between humans and flies responsible of inducing uncontrolled growth and cancer development. Second, we describe classic and novel Drosophila models used to study different cancers, with the objective to discuss their strengths and limitations on their use to identify signals driving growth cell autonomously and within organs, drug discovery and for therapeutic approaches.
Collapse
Affiliation(s)
- Zhasmine Mirzoyan
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Manuela Sollazzo
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Mariateresa Allocca
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | | | - Daniela Grifoni
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Paola Bellosta
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy.,Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy.,Department of Biosciences, University of Milan, Milan, Italy.,Department of Medicine, NYU Langone Medical Center, New York, NY, United States
| |
Collapse
|
18
|
Cheng Y, Chen D. Fruit fly research in China. J Genet Genomics 2018; 45:583-592. [PMID: 30455037 DOI: 10.1016/j.jgg.2018.09.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 09/21/2018] [Accepted: 09/29/2018] [Indexed: 11/19/2022]
Abstract
Served as a model organism over a century, fruit fly has significantly pushed forward the development of global scientific research, including in China. The high similarity in genomic features between fruit fly and human enables this tiny insect to benefit the biomedical studies of human diseases. In the past decades, Chinese biologists have used fruit fly to make numerous achievements on understanding the fundamental questions in many diverse areas of biology. Here, we review some of the recent fruit fly studies in China, and mainly focus on those studies in the fields of stem cell biology, cancer therapy and regeneration medicine, neurological disorders and epigenetics.
Collapse
Affiliation(s)
- Ying Cheng
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Dahua Chen
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
19
|
Wingless/Wnt Signaling in Intestinal Development, Homeostasis, Regeneration and Tumorigenesis: A Drosophila Perspective. J Dev Biol 2018; 6:jdb6020008. [PMID: 29615557 PMCID: PMC6026893 DOI: 10.3390/jdb6020008] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 03/23/2018] [Accepted: 03/24/2018] [Indexed: 02/06/2023] Open
Abstract
In mammals, the Wnt/β-catenin signal transduction pathway regulates intestinal stem cell maintenance and proliferation, whereas Wnt pathway hyperactivation, resulting primarily from the inactivation of the tumor suppressor Adenomatous polyposis coli (APC), triggers the development of the vast majority of colorectal cancers. The Drosophila adult gut has recently emerged as a powerful model to elucidate the mechanisms by which Wingless/Wnt signaling regulates intestinal development, homeostasis, regeneration, and tumorigenesis. Herein, we review recent insights on the roles of Wnt signaling in Drosophila intestinal physiology and pathology.
Collapse
|
20
|
Modelling Cooperative Tumorigenesis in Drosophila. BIOMED RESEARCH INTERNATIONAL 2018; 2018:4258387. [PMID: 29693007 PMCID: PMC5859872 DOI: 10.1155/2018/4258387] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 01/21/2018] [Indexed: 12/13/2022]
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.
Collapse
|
21
|
Perochon J, Carroll LR, Cordero JB. Wnt Signalling in Intestinal Stem Cells: Lessons from Mice and Flies. Genes (Basel) 2018; 9:genes9030138. [PMID: 29498662 PMCID: PMC5867859 DOI: 10.3390/genes9030138] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 02/17/2018] [Accepted: 02/21/2018] [Indexed: 12/12/2022] Open
Abstract
Adult stem cells play critical roles in the basal maintenance of tissue integrity, also known as homeostasis, and in tissue regeneration following damage. The highly conserved Wnt signalling pathway is a key regulator of stem cell fate. In the gastrointestinal tract, Wnt signalling activation drives homeostasis and damage-induced repair. Additionally, deregulated Wnt signalling is a common hallmark of age-associated tissue dysfunction and cancer. Studies using mouse and fruit fly models have greatly improved our understanding of the functional contribution of the Wnt signalling pathway in adult intestinal biology. Here, we summarize the latest knowledge acquired from mouse and Drosophila research regarding canonical Wnt signalling and its key functions during stem cell driven intestinal homeostasis, regeneration, ageing and cancer.
Collapse
Affiliation(s)
- Jessica Perochon
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK.
| | - Lynsey R Carroll
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK.
| | - Julia B Cordero
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK.
- CRUK Beatson Institute, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK.
| |
Collapse
|
22
|
Abstract
Accumulating epidemiological evidence indicates a strong clinical association between obesity and an increased risk of cancer. The global pandemic of obesity indicates a public health trend towards a substantial increase in cancer incidence and mortality. However, the mechanisms that link obesity to cancer remain incompletely understood. The fruit fly Drosophila melanogaster has been increasingly used to model an expanding spectrum of human diseases. Fly models provide a genetically simpler system that is ideal for use as a first step towards dissecting disease interactions. Recently, the combining of fly models of diet-induced obesity with models of cancer has provided a novel model system in which to study the biological mechanisms that underlie the connections between obesity and cancer. In this Review, I summarize recent advances, made using Drosophila, in our understanding of the interplay between diet, obesity, insulin resistance and cancer. I also discuss how the biological mechanisms and therapeutic targets that have been identified in fly studies could be utilized to develop preventative interventions and treatment strategies for obesity-associated cancers. Summary: This Review highlights a Drosophila model of diet-induced obesity and cancer, and how these two models are combined to study the interplay between obesity and cancer.
Collapse
Affiliation(s)
- Susumu Hirabayashi
- Metabolism and Cell Growth Group, MRC Clinical Sciences Centre (CSC), Du Cane Road, London W12 0NN, UK Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK
| |
Collapse
|
23
|
Liu Q, Jin LH. Tissue-resident stem cell activity: a view from the adult Drosophila gastrointestinal tract. Cell Commun Signal 2017; 15:33. [PMID: 28923062 PMCID: PMC5604405 DOI: 10.1186/s12964-017-0184-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 07/12/2017] [Indexed: 12/11/2022] Open
Abstract
The gastrointestinal tract serves as a fast-renewing model for unraveling the multifaceted molecular mechanisms underlying remarkably rapid cell renewal, which is exclusively fueled by a small number of long-lived stem cells and their progeny. Stem cell activity is the best-characterized aspect of mucosal homeostasis in mitotically active tissues, and the dysregulation of regenerative capacity is a hallmark of epithelial immune defects. This dysregulation is frequently associated with pathologies ranging from chronic enteritis to malignancies in humans. Application of the adult Drosophila gastrointestinal tract model in current and future studies to analyze the immuno-physiological aspects of epithelial defense strategies, including stem cell behavior and re-epithelialization, will be necessary to improve our general understanding of stem cell participation in epithelial turnover. In this review, which describes exciting observations obtained from the adult Drosophila gastrointestinal tract, we summarize a remarkable series of recent findings in the literature to decipher the molecular mechanisms through which stem cells respond to nonsterile environments.
Collapse
Affiliation(s)
- Qiang Liu
- Department of Genetics, College of Life Sciences, Northeast Forestry University, No.26 Hexing Road Xiangfang District, Harbin, 150040, China
| | - Li Hua Jin
- Department of Genetics, College of Life Sciences, Northeast Forestry University, No.26 Hexing Road Xiangfang District, Harbin, 150040, China.
| |
Collapse
|
24
|
Rahman MM, Franch-Marro X, Maestro JL, Martin D, Casali A. Local Juvenile Hormone activity regulates gut homeostasis and tumor growth in adult Drosophila. Sci Rep 2017; 7:11677. [PMID: 28916802 PMCID: PMC5600977 DOI: 10.1038/s41598-017-11199-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 08/16/2017] [Indexed: 11/09/2022] Open
Abstract
Hormones play essential roles during development and maintaining homeostasis in adult organisms, regulating a plethora of biological processes. Generally, hormones are secreted by glands and perform a systemic action. Here we show that Juvenile Hormones (JHs), insect sesquiterpenoids synthesized by the corpora allata, are also synthesized by the adult Drosophila gut. This local, gut specific JH activity, is synthesized by and acts on the intestinal stem cell and enteroblast populations, regulating their survival and cellular growth through the JH receptors Gce/Met and the coactivator Tai. Furthermore, we show that this local JH activity is important for damage response and is necessary for intestinal tumor growth driven by activating mutations in Wnt and EGFR/Ras pathways. Together, our results identify JHs as key hormonal regulators of gut homeostasis and open the possibility that analogous hormones may play a similar role in maintaining vertebrate adult intestinal stem cell population and sustaining tumor growth.
Collapse
Affiliation(s)
- M M Rahman
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, 08028, Barcelona, Spain.,Department of Molecular Cell Biology, Centre for Cancer Biomedicine, Institute for Cancer Research. Oslo University Hospital, Montebello, N-0379, Oslo, Norway
| | - X Franch-Marro
- Institut de Biologia Evolutiva (CSIC-UPF), Barcelona, Spain
| | - J L Maestro
- Institut de Biologia Evolutiva (CSIC-UPF), Barcelona, Spain
| | - D Martin
- Institut de Biologia Evolutiva (CSIC-UPF), Barcelona, Spain
| | - A Casali
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, 08028, Barcelona, Spain.
| |
Collapse
|
25
|
Tian A, Benchabane H, Wang Z, Zimmerman C, Xin N, Perochon J, Kalna G, Sansom OJ, Cheng C, Cordero JB, Ahmed Y. Intestinal stem cell overproliferation resulting from inactivation of the APC tumor suppressor requires the transcription cofactors Earthbound and Erect wing. PLoS Genet 2017; 13:e1006870. [PMID: 28708826 PMCID: PMC5510812 DOI: 10.1371/journal.pgen.1006870] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 06/15/2017] [Indexed: 12/30/2022] Open
Abstract
Wnt/β-catenin signal transduction directs intestinal stem cell (ISC) proliferation during homeostasis. Hyperactivation of Wnt signaling initiates colorectal cancer, which most frequently results from truncation of the tumor suppressor Adenomatous polyposis coli (APC). The β-catenin-TCF transcription complex activates both the physiological expression of Wnt target genes in the normal intestinal epithelium and their aberrantly increased expression in colorectal tumors. Whether mechanistic differences in the Wnt transcription machinery drive these distinct levels of target gene activation in physiological versus pathological states remains uncertain, but is relevant for the design of new therapeutic strategies. Here, using a Drosophila model, we demonstrate that two evolutionarily conserved transcription cofactors, Earthbound (Ebd) and Erect wing (Ewg), are essential for all major consequences of Apc1 inactivation in the intestine: the hyperactivation of Wnt target gene expression, excess number of ISCs, and hyperplasia of the epithelium. In contrast, only Ebd, but not Ewg, mediates the Wnt-dependent regulation of ISC proliferation during homeostasis. Therefore, in the adult intestine, Ebd acts independently of Ewg in physiological Wnt signaling, but cooperates with Ewg to induce the hyperactivation of Wnt target gene expression following Apc1 loss. These findings have relevance for human tumorigenesis, as Jerky (JRK/JH8), the human Ebd homolog, promotes Wnt pathway hyperactivation and is overexpressed in colorectal, breast, and ovarian cancers. Together, our findings reveal distinct requirements for Ebd and Ewg in physiological Wnt pathway activation versus oncogenic Wnt pathway hyperactivation following Apc1 loss. Such differentially utilized transcription cofactors may offer new opportunities for the selective targeting of Wnt-driven cancers.
Collapse
Affiliation(s)
- Ai Tian
- Department of Molecular and Systems Biology and the Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, Hanover, NH, United States of America
| | - Hassina Benchabane
- Department of Molecular and Systems Biology and the Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, Hanover, NH, United States of America
| | - Zhenghan Wang
- Department of Molecular and Systems Biology and the Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, Hanover, NH, United States of America
| | - Chloe Zimmerman
- Department of Molecular and Systems Biology and the Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, Hanover, NH, United States of America
| | - Nan Xin
- Department of Molecular and Systems Biology and the Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, Hanover, NH, United States of America
| | - Jessica Perochon
- Wolfson Wohl Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Gabriela Kalna
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
| | - Owen J. Sansom
- CRUK Beatson Institute, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow, United Kingdom
| | - Chao Cheng
- Department of Biomedical Data Science, Molecular and Systems Biology and the Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, Hanover, NH, United States of America
| | - Julia B. Cordero
- Wolfson Wohl Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Yashi Ahmed
- Department of Molecular and Systems Biology and the Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, Hanover, NH, United States of America
| |
Collapse
|
26
|
High sugar diet disrupts gut homeostasis though JNK and STAT pathways in Drosophila. Biochem Biophys Res Commun 2017; 487:910-916. [DOI: 10.1016/j.bbrc.2017.04.156] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 04/30/2017] [Indexed: 01/06/2023]
|
27
|
Hu B, Zhang H, Wei H, Wang Z, Zhang F, Wang X, Li L. Does adenomatous polyposis coli gene promoter 1A methylation increase non-small cell lung cancer risk? A meta-analysis. Thorac Cancer 2017; 8:410-416. [PMID: 28497891 PMCID: PMC5582469 DOI: 10.1111/1759-7714.12450] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 03/30/2017] [Accepted: 03/30/2017] [Indexed: 12/12/2022] Open
Abstract
Background The promoter region of the adenomatous polyposis coli (APC) gene is hypermethylated in several types of cancers, including non‐small cell lung cancer (NSCLC). The prevalence of methylation in the promoter region of this gene in tumor tissues and autologous controls has not been consistent in previous studies. We evaluated the frequency of APC gene promoter 1A methylation between tumor tissues and autologous controls in NSCLC patients by meta‐analysis. Methods Open published studies of APC gene promoter 1A methylation between tumor tissues and autologous samples in NSCLC patients were identified using a systematic search. Odds ratios (OR) and 95% confidence intervals (CI) of APC gene promoter 1A methylation in lung cancer tissues versus autologous controls were calculated. Fourteen studies, involving a total of 1345 patients and 2182 samples, were finally included. Results The pooled proportion of APC promoter 1A methylation was 0.62 (95% CI 0.52–072) and 0.34 (95% CI 0.21–0.50) in cancer tissues and autologous controls, respectively. The APC gene promoter 1A methylation rate in cancer tissues was much higher than in autologous controls, with a pooled OR of 3.66 (95% CI 2.12–6.33). A strong and significant correlation of APC gene promoter 1A methylation between tumor tissues and autologous controls was detected (correlation coefficient rpearson = 0.77; P = 0.0013). Conclusion The proportion of APC promoter 1A methylation in lung cancer tissues was higher than in autologous controls, indicating that promoter 1A methylation of the APC gene may play an important role in NSCLC carcinogenesis.
Collapse
Affiliation(s)
- Baoli Hu
- Department Cardiothoracic Surgery, Huaihe Hospital of Henan University, Kaifegn, China
| | - Hangfeng Zhang
- Department Cardiothoracic Surgery, Huaihe Hospital of Henan University, Kaifegn, China
| | - Haitao Wei
- Department Cardiothoracic Surgery, Huaihe Hospital of Henan University, Kaifegn, China
| | - Zuopei Wang
- Department Cardiothoracic Surgery, Huaihe Hospital of Henan University, Kaifegn, China
| | - Feng Zhang
- Department Cardiothoracic Surgery, Huaihe Hospital of Henan University, Kaifegn, China
| | - Xiaolong Wang
- Department Cardiothoracic Surgery, Huaihe Hospital of Henan University, Kaifegn, China
| | - Li Li
- Department of Clinical Teaching and Research, School of Nursing, Henan University, Kaifegn, China
| |
Collapse
|
28
|
Arnal A, Jacqueline C, Ujvari B, Leger L, Moreno C, Faugere D, Tasiemski A, Boidin‐Wichlacz C, Misse D, Renaud F, Montagne J, Casali A, Roche B, Mery F, Thomas F. Cancer brings forward oviposition in the fly Drosophila melanogaster. Ecol Evol 2017; 7:272-276. [PMID: 28070290 PMCID: PMC5214257 DOI: 10.1002/ece3.2571] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 09/19/2016] [Accepted: 09/30/2016] [Indexed: 12/17/2022] Open
Abstract
Hosts often accelerate their reproductive effort in response to a parasitic infection, especially when their chances of future reproduction decrease with time from the onset of the infection. Because malignancies usually reduce survival, and hence potentially the fitness, it is expected that hosts with early cancer could have evolved to adjust their life-history traits to maximize their immediate reproductive effort. Despite the potential importance of these plastic responses, little attention has been devoted to explore how cancers influence animal reproduction. Here, we use an experimental setup, a colony of genetically modified flies Drosophila melanogaster which develop colorectal cancer in the anterior gut, to show the role of cancer in altering life-history traits. Specifically, we tested whether females adapt their reproductive strategy in response to harboring cancer. We found that flies with cancer reached the peak period of oviposition significantly earlier (i.e., 2 days) than healthy ones, while no difference in the length and extent of the fecundity peak was observed between the two groups of flies. Such compensatory responses to overcome the fitness-limiting effect of cancer could explain the persistence of inherited cancer-causing mutant alleles in the wild.
Collapse
Affiliation(s)
- Audrey Arnal
- CREECMIVEGECUMR IRD/CNRS/UM 5290911 Avenue Agropolis, BP 6450134394Montpellier Cedex 5France
| | - Camille Jacqueline
- CREECMIVEGECUMR IRD/CNRS/UM 5290911 Avenue Agropolis, BP 6450134394Montpellier Cedex 5France
| | - Beata Ujvari
- Centre for Integrative EcologySchool of Life and Environmental SciencesDeakin UniversityWaurn PondsVic.Australia
| | - Lucas Leger
- CREECMIVEGECUMR IRD/CNRS/UM 5290911 Avenue Agropolis, BP 6450134394Montpellier Cedex 5France
| | - Céline Moreno
- Laboratoire Évolution, Génomes, et SpéciationUnité Propre de Recherche 9034Centre National de la Recherche Scientifique, 91198 Gif sur Yvette, France; Université Paris‐Sud 1191405OrsayFrance
| | - Dominique Faugere
- CREECMIVEGECUMR IRD/CNRS/UM 5290911 Avenue Agropolis, BP 6450134394Montpellier Cedex 5France
| | | | | | - Dorothée Misse
- CREECMIVEGECUMR IRD/CNRS/UM 5290911 Avenue Agropolis, BP 6450134394Montpellier Cedex 5France
| | - François Renaud
- CREECMIVEGECUMR IRD/CNRS/UM 5290911 Avenue Agropolis, BP 6450134394Montpellier Cedex 5France
| | - Jacques Montagne
- Institute for Integrative Biology of the Cell (I2BC)CNRSUniversité Paris‐SudCEA, UMR919891190Gif‐sur‐YvetteFrance
| | - Andreu Casali
- Institute for Research in Biomedicine (IRB Barcelona)BarcelonaSpain
| | - Benjamin Roche
- CREECMIVEGECUMR IRD/CNRS/UM 5290911 Avenue Agropolis, BP 6450134394Montpellier Cedex 5France
- International Center for Mathematical and Computational Modelling of Complex Systems (UMI IRD/UPMC UMMISCO)32 Avenue Henri Varagnat93143Bondy CedexFrance
| | - Frédéric Mery
- Laboratoire Évolution, Génomes, et SpéciationUnité Propre de Recherche 9034Centre National de la Recherche Scientifique, 91198 Gif sur Yvette, France; Université Paris‐Sud 1191405OrsayFrance
| | - Frédéric Thomas
- CREECMIVEGECUMR IRD/CNRS/UM 5290911 Avenue Agropolis, BP 6450134394Montpellier Cedex 5France
| |
Collapse
|
29
|
Lim HC, Jou TS. Ras-activated RSK1 phosphorylates EBP50 to regulate its nuclear localization and promote cell proliferation. Oncotarget 2016; 7:10283-96. [PMID: 26862730 PMCID: PMC4891120 DOI: 10.18632/oncotarget.7184] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 01/25/2016] [Indexed: 11/25/2022] Open
Abstract
Differential subcellular localization of EBP50 leads to its controversial role in cancer biology either as a tumor suppressor when it resides at the membrane periphery, or a tumor facilitator at the nucleus. However, the mechanism behind nuclear localization of EBP50 remains unclear. A RNA interference screening identified the downstream effector of the Ras-ERK cascade, RSK1, as the molecule unique for nuclear transport of EBP50. RSK1 binds to EBP50 and phosphorylates it at a conserved threonine residue at position 156 (T156) under the regulation of growth factor. Mutagenesis experiments confirmed the significance of T156 residue in nuclear localization of EBP50, cellular proliferation, and oncogenic transformation. Our study sheds light on a possible therapeutic strategy targeting at this aberrant nuclear expression of EBP50 without affecting the normal physiological function of EBP50 at other subcellular localization.
Collapse
Affiliation(s)
- Hooi Cheng Lim
- Graduate Institute of Molecular Medicine, National Taiwan University, Taipei, Taiwan
| | - Tzuu-Shuh Jou
- Graduate Institute of Molecular Medicine, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Clinical Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
30
|
Guo Z, Lucchetta E, Rafel N, Ohlstein B. Maintenance of the adult Drosophila intestine: all roads lead to homeostasis. Curr Opin Genet Dev 2016; 40:81-86. [PMID: 27392294 DOI: 10.1016/j.gde.2016.06.009] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 05/20/2016] [Accepted: 06/16/2016] [Indexed: 10/21/2022]
Abstract
Maintenance of tissue homeostasis is critical in tissues with high turnover such as the intestinal epithelium. The intestinal epithelium is under constant cellular assault due to its digestive functions and its function as a barrier to chemical and bacterial insults. The resulting high rate of cellular turnover necessitates highly controlled mechanisms of regeneration to maintain the integrity of the tissue over the lifetime of the organism. Transient increase in stem cell proliferation is a commonly used and elaborate mechanism to ensure fast and efficient repair of the gut. However, tissue repair is not limited to regulating ISC proliferation, as emerging evidence demonstrates that the Drosophila intestine uses multiple strategies to ensure proper tissue homeostasis that may also extend to other tissues.
Collapse
Affiliation(s)
- Zheng Guo
- Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032, USA
| | - Elena Lucchetta
- Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032, USA
| | - Neus Rafel
- Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032, USA
| | - Benjamin Ohlstein
- Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032, USA.
| |
Collapse
|
31
|
Tian A, Benchabane H, Wang Z, Ahmed Y. Regulation of Stem Cell Proliferation and Cell Fate Specification by Wingless/Wnt Signaling Gradients Enriched at Adult Intestinal Compartment Boundaries. PLoS Genet 2016; 12:e1005822. [PMID: 26845150 PMCID: PMC4742051 DOI: 10.1371/journal.pgen.1005822] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 12/31/2015] [Indexed: 01/12/2023] Open
Abstract
Intestinal stem cell (ISC) self-renewal and proliferation are directed by Wnt/β-catenin signaling in mammals, whereas aberrant Wnt pathway activation in ISCs triggers the development of human colorectal carcinoma. Herein, we have utilized the Drosophila midgut, a powerful model for ISC regulation, to elucidate the mechanisms by which Wingless (Wg)/Wnt regulates intestinal homeostasis and development. We provide evidence that the Wg signaling pathway, activation of which peaks at each of the major compartment boundaries of the adult intestine, has essential functions. Wg pathway activation in the intestinal epithelium is required not only to specify cell fate near compartment boundaries during development, but also to control ISC proliferation within compartments during homeostasis. Further, in contrast with the previous focus on Wg pathway activation within ISCs, we demonstrate that the primary mechanism by which Wg signaling regulates ISC proliferation during homeostasis is non-autonomous. Activation of the Wg pathway in absorptive enterocytes is required to suppress JAK-STAT signaling in neighboring ISCs, and thereby their proliferation. We conclude that Wg signaling gradients have essential roles during homeostasis and development of the adult intestine, non-autonomously controlling stem cell proliferation inside compartments, and autonomously specifying cell fate near compartment boundaries. The highly conserved Wingless/Wnt signal transduction pathway directs many cellular processes in metazoans and its deregulation underlies numerous human congenital diseases and cancers. Most notably, more than 80% of colon cancers arise from aberrant activation of the Wnt pathway. A better understanding of how Wnt signaling functions in the intestinal stem cells (ISCs) during homeostasis and in disease states is thus critical. The Drosophila digestive tract provides a powerful genetic model and an entry point to study these questions. Here, we find that the Wg ligand and pathway activation are enriched at Drosophila intestinal compartment boundaries and are essential for development and homeostasis of the adult gut. During homeostasis, Wg signaling in enterocytes is required to prevent the overproliferation of ISCs non-autonomously. In addition, during development, Wg signaling ensures proper cell fate specification near compartment boundaries. These findings provide insight into the mechanisms underlying the Wg-dependent regulation of adult intestinal function.
Collapse
Affiliation(s)
- Ai Tian
- Department of Genetics and the Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, Hanover, New Hampshire, United States of America
| | - Hassina Benchabane
- Department of Genetics and the Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, Hanover, New Hampshire, United States of America
| | - Zhenghan Wang
- Department of Genetics and the Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, Hanover, New Hampshire, United States of America
| | - Yashi Ahmed
- Department of Genetics and the Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, Hanover, New Hampshire, United States of America
- * E-mail:
| |
Collapse
|
32
|
Accumulation of differentiating intestinal stem cell progenies drives tumorigenesis. Nat Commun 2015; 6:10219. [PMID: 26690827 PMCID: PMC4703904 DOI: 10.1038/ncomms10219] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 11/13/2015] [Indexed: 12/31/2022] Open
Abstract
Stem cell self-renewal and differentiation are coordinated to maintain tissue homeostasis and prevent cancer. Mutations causing stem cell proliferation are traditionally the focus of cancer studies. However, the contribution of the differentiating stem cell progenies in tumorigenesis is poorly characterized. Here we report that loss of the SOX transcription factor, Sox21a, blocks the differentiation programme of enteroblast (EB), the intestinal stem cell progeny in the adult Drosophila midgut. This results in EB accumulation and formation of tumours. Sox21a tumour initiation and growth involve stem cell proliferation induced by the unpaired 2 mitogen released from accumulating EBs generating a feed-forward loop. EBs found in the tumours are heterogeneous and grow towards the intestinal lumen. Sox21a tumours modulate their environment by secreting matrix metalloproteinase and reactive oxygen species. Enterocytes surrounding the tumours are eliminated through delamination allowing tumour progression, a process requiring JNK activation. Our data highlight the tumorigenic properties of transit differentiating cells.
Collapse
|
33
|
Wang C, Guo X, Dou K, Chen H, Xi R. Ttk69 acts as a master repressor of enteroendocrine cell specification in Drosophila intestinal stem cell lineages. Development 2015; 142:3321-31. [PMID: 26293304 DOI: 10.1242/dev.123208] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 08/10/2015] [Indexed: 01/15/2023]
Abstract
In adult Drosophila midgut, intestinal stem cells (ISCs) periodically produce progenitor cells that undergo a binary fate choice determined primarily by the levels of Notch activity that they receive, before terminally differentiating into enterocytes (ECs) or enteroendocrine (EE) cells. Here we identified Ttk69, a BTB domain-containing transcriptional repressor, as a master repressor of EE cell specification in the ISC lineages. Depletion of ttk69 in progenitor cells induced ISC proliferation and caused all committed progenitor cells to adopt EE fate, leading to the production of supernumerary EE cells in the intestinal epithelium. Conversely, forced expression of Ttk69 in progenitor cells was sufficient to prevent EE cell specification. The expression of Ttk69 was not regulated by Notch signaling, and forced activation of Notch, which is sufficient to induce EC specification of normal progenitor cells, failed to prevent EE cell specification of Ttk69-depleted progenitors. Loss of Ttk69 led to derepression of the acheate-scute complex (AS-C) genes scute and asense, which then induced prospero expression to promote EE cell specification. These studies suggest that Ttk69 functions in parallel with Notch signaling and acts as a master repressor of EE cell specification in Drosophila ISC lineages primarily by suppressing AS-C genes.
Collapse
Affiliation(s)
- Chenhui Wang
- National Institute of Biological Sciences, No. 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China
| | - Xingting Guo
- National Institute of Biological Sciences, No. 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China
| | - Kun Dou
- National Institute of Biological Sciences, No. 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China
| | - Hongyan Chen
- National Institute of Biological Sciences, No. 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China
| | - Rongwen Xi
- National Institute of Biological Sciences, No. 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China
| |
Collapse
|
34
|
Oncogenic KRAS signalling promotes the Wnt/β-catenin pathway through LRP6 in colorectal cancer. Oncogene 2014; 34:4914-27. [PMID: 25500543 PMCID: PMC4687460 DOI: 10.1038/onc.2014.416] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 11/04/2014] [Accepted: 11/08/2014] [Indexed: 12/14/2022]
Abstract
Aberrant regulation of the Wnt/β-catenin signaling pathway is one of the major causes of colorectal cancer (CRC). Loss-of-function mutations in APC are commonly found in CRC, leading to inappropriate activation of canonical Wnt signaling. Conversely, gain-of-function mutations in KRAS and BRAF genes are detected in up to 60% of CRCs. Whereas KRAS/mitogen-activated protein kinase (MAPK) and canonical Wnt/β-catenin pathways are critical for intestinal tumorigenesis, mechanisms integrating these two important signaling pathways during CRC development are unknown. Results herein demonstrate that transformation of normal intestinal epithelial cells (IECs) by oncogenic forms of KRAS, BRAF or MEK1 was associated with a marked increase in β-catenin/TCF4 and c-MYC promoter transcriptional activities and mRNA levels of c-Myc, Axin2 and Lef1. Notably, expression of a dominant-negative mutant of T-Cell Factor 4 (ΔNTCF4) severely attenuated IEC transformation induced by oncogenic MEK1 and markedly reduced their tumorigenic and metastatic potential in immunocompromised mice. Interestingly, the Frizzled co-receptor LRP6 was phosphorylated in a MEK-dependent manner in transformed IECs and in human CRC cell lines. Expression of LRP6 mutant in which serine/threonine residues in each particular ProlineProlineProlineSerine/ThreonineProline motif were mutated to alanines (LRP6-5A) significantly reduced β-catenin/TCF4 transcriptional activity. Accordingly, MEK inhibition in human CRC cells significantly diminished β-catenin/TCF4 transcriptional activity and c-MYC mRNA and protein levels without affecting β-catenin expression or stability. Lastly, LRP6 phosphorylation was also increased in human colorectal tumors, including adenomas, in comparison with healthy adjacent normal tissues. Our data indicate that oncogenic activation of KRAS/BRAF/MEK signaling stimulates the canonical Wnt/β-catenin pathway, which in turn promotes intestinal tumor growth and invasion. Moreover, LRP6 phosphorylation by ERK1/2 may provide a unique point of convergence between KRAS/MAPK and Wnt/β-catenin signalings during oncogenesis.
Collapse
|
35
|
Martorell Ò, Barriga FM, Merlos-Suárez A, Stephan-Otto Attolini C, Casanova J, Batlle E, Sancho E, Casali A. Iro/IRX transcription factors negatively regulate Dpp/TGF-β pathway activity during intestinal tumorigenesis. EMBO Rep 2014; 15:1210-8. [PMID: 25296644 DOI: 10.15252/embr.201438622] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Activating mutations in Wnt and EGFR/Ras signaling pathways are common in colorectal cancer (CRC). Remarkably, clonal co-activation of these pathways in the adult Drosophila midgut induces "tumor-like" overgrowths. Here, we show that, in these clones and in CRC cell lines, Dpp/TGF-β acts as a tumor suppressor. Moreover, we discover that the Iroquois/IRX-family-protein Mirror downregulates the transcription of core components of the Dpp pathway, reducing its tumor suppressor activity. We also show that this genetic interaction is conserved in human CRC cells, where the Iro/IRX proteins IRX3 and IRX5 diminish the response to TGF-β. IRX3 and IRX5 are upregulated in human adenomas, and their levels correlate inversely with the gene expression signature of response to TGF-β. In addition, Irx5 expression confers a growth advantage in the presence of TGF-β, but is selected against in its absence. Together, our results identify a set of Iro/IRX proteins as conserved negative regulators of Dpp/TGF-β activity. We propose that during the characteristic adenoma-to-carcinoma transition of human CRC, the activity of IRX proteins could reduce the sensitivity to the cytostatic effect of TGF-β, conferring a growth advantage to tumor cells prior to the acquisition of mutations in TGF-β pathway components.
Collapse
Affiliation(s)
- Òscar Martorell
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain Institut de Biologia Molecular de Barcelona (IBMB-CSIC), Barcelona, Spain
| | | | | | | | - Jordi Casanova
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain Institut de Biologia Molecular de Barcelona (IBMB-CSIC), Barcelona, Spain
| | - Eduard Batlle
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Elena Sancho
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain
| | - Andreu Casali
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain
| |
Collapse
|
36
|
Oostindjer M, Alexander J, Amdam GV, Andersen G, Bryan NS, Chen D, Corpet DE, De Smet S, Dragsted LO, Haug A, Karlsson AH, Kleter G, de Kok TM, Kulseng B, Milkowski AL, Martin RJ, Pajari AM, Paulsen JE, Pickova J, Rudi K, Sødring M, Weed DL, Egelandsdal B. The role of red and processed meat in colorectal cancer development: a perspective. Meat Sci 2014; 97:583-96. [DOI: 10.1016/j.meatsci.2014.02.011] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 02/14/2014] [Accepted: 02/17/2014] [Indexed: 02/07/2023]
|
37
|
Patel PH, Edgar BA. Tissue design: how Drosophila tumors remodel their neighborhood. Semin Cell Dev Biol 2014; 28:86-95. [PMID: 24685612 DOI: 10.1016/j.semcdb.2014.03.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 03/03/2014] [Indexed: 12/24/2022]
Abstract
Drosophila genetics has long been appreciated as a powerful approach for discovering the normal functions of genes that act as oncogenes and tumor suppressors in human cancer. Recent studies have also highlighted its advantages for deciphering how such genes function during tumorigenesis itself. Here we detail studies relating to how tumors, generated in developing organs and adult stem cell-based tissues, remodel the tissue landscape to their benefit. Like mammalian tumors, insect tumors can dissolve extracellular matrix, recruit blood cells, migrate and invade other tissues. While much is known about how mammalian fibroblasts, immune cells and vasculature promote late tumorigenesis, less is understood about the very earliest stages of tumor development in mammals. Because Drosophila has fewer mitotic cells and a simpler tissue architecture, it affords easy detection and analysis of early clonal tumor growth. Drosophila studies have revealed both cooperative and competitive interactions between tumor and normal cells during early tumor growth. During development, these interactions typically occur with other proliferative progenitor cells, but in adult stem cell-based tissues, the stem cell niche can fuel tumor growth.
Collapse
Affiliation(s)
- Parthive H Patel
- German Cancer Research Center (DKFZ) and Center for Molecular Biology Heidelberg (ZMBH) Alliance, Im Neuenheimer Feld 282, D-69120 Heidelberg, Germany.
| | - Bruce A Edgar
- German Cancer Research Center (DKFZ) and Center for Molecular Biology Heidelberg (ZMBH) Alliance, Im Neuenheimer Feld 282, D-69120 Heidelberg, Germany.
| |
Collapse
|
38
|
Wang C, Guo X, Xi R. EGFR and Notch signaling respectively regulate proliferative activity and multiple cell lineage differentiation of Drosophila gastric stem cells. Cell Res 2014; 24:610-27. [PMID: 24603358 DOI: 10.1038/cr.2014.27] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Revised: 12/16/2013] [Accepted: 01/17/2014] [Indexed: 12/12/2022] Open
Abstract
Quiescent, multipotent gastric stem cells (GSSCs) in the copper cell region of adult Drosophila midgut can produce all epithelial cell lineages found in the region, including acid-secreting copper cells, interstitial cells and enteroendocrine cells, but mechanisms controlling their quiescence and the ternary lineage differentiation are unknown. By using cell ablation or damage-induced regeneration assays combined with cell lineage tracing and genetic analysis, here we demonstrate that Delta (Dl)-expressing cells in the copper cell region are the authentic GSSCs that can self-renew and continuously regenerate the gastric epithelium after a sustained damage. Lineage tracing analysis reveals that the committed GSSC daughter with activated Notch will invariably differentiate into either a copper cell or an interstitial cell, but not the enteroendocrine cell lineage, and loss-of-function and gain-of-function studies revealed that Notch signaling is both necessary and sufficient for copper cell/interstitial cell differentiation. We also demonstrate that elevated epidermal growth factor receptor (EGFR) signaling, which is achieved by the activation of ligand Vein from the surrounding muscle cells and ligand Spitz from progenitor cells, mediates the regenerative proliferation of GSSCs following damage. Taken together, we demonstrate that Dl is a specific marker for Drosophila GSSCs, whose cell cycle status is dependent on the levels of EGFR signaling activity, and the Notch signaling has a central role in controlling cell lineage differentiation from GSSCs by separating copper/interstitial cell lineage from enteroendocrine cell lineage.
Collapse
Affiliation(s)
- Chenhui Wang
- 1] National Institute of Biological Sciences, No. 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China [2] College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Xingting Guo
- 1] National Institute of Biological Sciences, No. 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China [2] College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Rongwen Xi
- National Institute of Biological Sciences, No. 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China
| |
Collapse
|
39
|
Conserved mechanisms of tumorigenesis in the Drosophila adult midgut. PLoS One 2014; 9:e88413. [PMID: 24516653 PMCID: PMC3916428 DOI: 10.1371/journal.pone.0088413] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 01/06/2014] [Indexed: 11/19/2022] Open
Abstract
Whereas the series of genetic events leading to colorectal cancer (CRC) have been well established, the precise functions that these alterations play in tumor progression and how they disrupt intestinal homeostasis remain poorly characterized. Activation of the Wnt/Wg signaling pathway by a mutation in the gene APC is the most common trigger for CRC, inducing benign lesions that progress to carcinomas due to the accumulation of other genetic alterations. Among those, Ras mutations drive tumour progression in CRC, as well as in most epithelial cancers. As mammalian and Drosophila's intestines share many similarities, we decided to explore the alterations induced in the Drosophila midgut by the combined activation of the Wnt signaling pathway with gain of function of Ras signaling in the intestinal stem cells. Here we show that compound Apc-Ras clones, but not clones bearing the individual mutations, expand as aggressive intestinal tumor-like outgrowths. These lesions reproduce many of the human CRC hallmarks such as increased proliferation, blockade of cell differentiation and cell polarity and disrupted organ architecture. This process is followed by expression of tumoral markers present in human lesions. Finally, a metabolic behavioral assay shows that these flies suffer a progressive deterioration in intestinal homeostasis, providing a simple readout that could be used in screens for tumor modifiers or therapeutic compounds. Taken together, our results illustrate the conservation of the mechanisms of CRC tumorigenesis in Drosophila, providing an excellent model system to unravel the events that, upon mutation in Apc and Ras, lead to CRC initiation and progression.
Collapse
|