51
|
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
|
52
|
Beira JV, Torres J, Paro R. Signalling crosstalk during early tumorigenesis in the absence of Polycomb silencing. PLoS Genet 2018; 14:e1007187. [PMID: 29357360 PMCID: PMC5794193 DOI: 10.1371/journal.pgen.1007187] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 02/01/2018] [Accepted: 01/04/2018] [Indexed: 12/19/2022] Open
Abstract
In response to stress and injury a coordinated activation of conserved signalling modules, such as JNK and JAK/STAT, is critical to trigger regenerative tissue restoration. While these pathways rebuild homeostasis and promote faithful organ recovery, it is intriguing that they also become activated in various tumour conditions. Therefore, it is crucial to understand how similar pathways can achieve context-dependent functional outputs, likely depending on cellular states. Compromised chromatin regulation, upon removal of the Polycomb group member polyhomeotic, leads to tumour formation with ectopic activation of JNK signalling, mediated by egr/grnd, in addition to JAK/STAT and Notch. Employing quantitative analyses, we show that blocking ectopic signalling impairs ph tumour growth. Furthermore, JAK/STAT functions in parallel to JNK, while Notch relies on JNK. Here, we reveal a signalling hierarchy in ph tumours that is distinct from the regenerative processes regulated by these pathways. Absence of ph renders a permissive state for expression of target genes, but our results suggest that both loss of repression and the presence of activators may collectively regulate gene expression during tumorigenesis. Further dissecting the effect of signalling, developmental or stress-induced factors will thus elucidate the regulation of physiological responses and the contribution of context-specific cellular states.
Collapse
Affiliation(s)
- Jorge V. Beira
- ETH Zürich, Department of Biosystems Science and Engineering, MattenstrasseBasel, Switzerland
- * E-mail: (JVB); (RP)
| | - Joana Torres
- ETH Zürich, Department of Biosystems Science and Engineering, MattenstrasseBasel, Switzerland
| | - Renato Paro
- ETH Zürich, Department of Biosystems Science and Engineering, MattenstrasseBasel, Switzerland
- Faculty of Science, University of Basel, KlingelbergstrasseBasel, Switzerland
- * E-mail: (JVB); (RP)
| |
Collapse
|
53
|
|
54
|
Endocytic Trafficking of the Notch Receptor. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1066:99-122. [DOI: 10.1007/978-3-319-89512-3_6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
55
|
Kon S. Physiological and pathological relevance of cell competition in fly to mammals. Dev Growth Differ 2017; 60:14-20. [PMID: 29250773 DOI: 10.1111/dgd.12415] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 10/24/2017] [Accepted: 10/30/2017] [Indexed: 12/20/2022]
Abstract
In multicellular organisms, incidentally emerging suboptimal cells are removed to maintain homeostasis of tissues. The unfavorable cells are excluded by a process termed cell competition whereby the resident normal cells actively eliminate the unfit cells of the identical lineage. Although the phenomenon of cell competition was originally discovered in Drosophila, a number of recent studies have provided implications of cell competition in tissue regeneration, development and oncogenesis in mammals. Here the roles of cell competition in fly to mammals are discussed.
Collapse
Affiliation(s)
- Shunsuke Kon
- Division of Molecular Oncology, Institute for Genetic Medicine, Hokkaido University Graduate School of Chemical Sciences and Engineering, Sapporo, 060-0815, Japan
| |
Collapse
|
56
|
Sun YX, Tang L, Gao J, Feng YY, Peng T, Yu YY, Yang LL, Sun Y, Zhu BJ. A role of tumor susceptibility gene 101 (TSG101) in innate immune response of crayfish Procambarus clarkii. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2017; 76:268-273. [PMID: 28673823 DOI: 10.1016/j.dci.2017.06.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 06/29/2017] [Accepted: 06/30/2017] [Indexed: 06/07/2023]
Abstract
Tumor susceptibility gene 101 (TSG101) is a multi-functional gene involved in cell growth and proliferation in vertebrates. However, its role in the innate immune response of crustaceans remains unclear. Here, a TSG101 gene was identified in crayfish Procambarus clarkii with an open reading frame of 1320 bp that encoded a predicted 48.3-kDa protein highly homologous to those in other invertebrates. TSG101 mRNA was highly expressed in stomach and hepatopancreas, and its expression was induced significantly in different tissues (hemocytes, gills and intestine) by lipopolysaccharide (LPS) and polyinosinic:polycytidylic acid (poly I: C) with various expression patterns. Recombinant TSG101 protein was expressed in Escherichia coli, and a possible protein-protein interaction between TSG101 and hepatocyte growth factor-regulated tyrosine kinase substrate (Hrs) was explored by far-western blotting. RNA interference of TSG101 affected the gene expression of members of the Toll pathway. These results suggest that TSG101 is involved in the innate immune responses of P. clarkii.
Collapse
Affiliation(s)
- Yu-Xuan Sun
- College of Life Sciences, Anhui Agricultural University, Hefei 230036, China
| | - Lin Tang
- College of Life Sciences, Anhui Agricultural University, Hefei 230036, China
| | - Jin Gao
- College of Life Sciences, Anhui Agricultural University, Hefei 230036, China
| | - Yuan-Yuan Feng
- College of Life Sciences, Anhui Agricultural University, Hefei 230036, China
| | - Tao Peng
- College of Life Sciences, Anhui Agricultural University, Hefei 230036, China
| | - Ying-Ying Yu
- College of Life Sciences, Anhui Agricultural University, Hefei 230036, China
| | - Liang-Li Yang
- College of Life Sciences, Anhui Agricultural University, Hefei 230036, China
| | - Yu Sun
- College of Life Sciences, Anhui Agricultural University, Hefei 230036, China
| | - Bao-Jian Zhu
- College of Life Sciences, Anhui Agricultural University, Hefei 230036, China.
| |
Collapse
|
57
|
Horner DS, Pasini ME, Beltrame M, Mastrodonato V, Morelli E, Vaccari T. ESCRT genes and regulation of developmental signaling. Semin Cell Dev Biol 2017; 74:29-39. [PMID: 28847745 DOI: 10.1016/j.semcdb.2017.08.038] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 08/06/2017] [Accepted: 08/18/2017] [Indexed: 11/30/2022]
Abstract
ESCRT (Endosomal Sorting Complex Required for Transport) proteins have been shown to control an increasing number of membrane-associated processes. Some of these, and prominently regulation of receptor trafficking, profoundly shape signal transduction. Evidence in fungi, plants and multiple animal models support the emerging concept that ESCRTs are main actors in coordination of signaling with the changes in cells and tissues occurring during development and homeostasis. Consistent with their pleiotropic function, ESCRTs are regulated in multiple ways to tailor signaling to developmental and homeostatic needs. ESCRT activity is crucial to correct execution of developmental programs, especially at key transitions, allowing eukaryotes to thrive and preventing appearance of congenital defects.
Collapse
Affiliation(s)
- David S Horner
- Dipartimento di Bioscienze, Universita' degli Studi di Milano, Via Celoria 26, 20133 Milano, Italy
| | - Maria E Pasini
- Dipartimento di Bioscienze, Universita' degli Studi di Milano, Via Celoria 26, 20133 Milano, Italy
| | - Monica Beltrame
- Dipartimento di Bioscienze, Universita' degli Studi di Milano, Via Celoria 26, 20133 Milano, Italy
| | - Valeria Mastrodonato
- IFOM, The FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milano, Italy
| | - Elena Morelli
- IFOM, The FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milano, Italy
| | - Thomas Vaccari
- Dipartimento di Bioscienze, Universita' degli Studi di Milano, Via Celoria 26, 20133 Milano, Italy; IFOM, The FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milano, Italy.
| |
Collapse
|
58
|
Szymanska E, Budick-Harmelin N, Miaczynska M. Endosomal "sort" of signaling control: The role of ESCRT machinery in regulation of receptor-mediated signaling pathways. Semin Cell Dev Biol 2017; 74:11-20. [PMID: 28797837 DOI: 10.1016/j.semcdb.2017.08.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Revised: 07/24/2017] [Accepted: 08/04/2017] [Indexed: 12/31/2022]
Abstract
The endosomal sorting complexes required for transport (ESCRTs) machinery consists of four protein assemblies (ESCRT-0 to -III subcomplexes) which mediate various processes of membrane remodeling in the cell. In the endocytic pathway, ESCRTs sort cargo destined for degradation into intraluminal vesicles (ILVs) of endosomes. Cargos targeted by ESCRTs include various signaling molecules, mainly internalized cell-surface receptors but also some cytosolic proteins. It is therefore expected that aberrant trafficking caused by ESCRT dysfunction affects different signaling pathways. Here we review how perturbation of ESCRT activity alters intracellular transport of membrane receptors, causing their accumulation on endocytic compartments, decreased degradation and/or altered recycling to the plasma membrane. We further describe how perturbed trafficking of receptors impacts the activity of their downstream signaling pathways, with or without changes in transcriptional responses. Finally, we present evidence that ESCRT components can also control activity and intracellular distribution of cytosolic signaling proteins (kinases, other effectors and soluble receptors). The underlying mechanisms involve sequestration of such proteins in ILVs, their sorting for degradation or towards non-lysosomal destinations, and regulating their availability in various cellular compartments. All these ESCRT-mediated processes can modulate final outputs of multiple signaling pathways.
Collapse
Affiliation(s)
- Ewelina Szymanska
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Noga Budick-Harmelin
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland; Cell Research and Immunology Department, George S. Wise Faculty of Life Sciences, Tel Aviv University, Israel
| | - Marta Miaczynska
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland.
| |
Collapse
|
59
|
Baker NE. Mechanisms of cell competition emerging from Drosophila studies. Curr Opin Cell Biol 2017; 48:40-46. [PMID: 28600967 DOI: 10.1016/j.ceb.2017.05.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 05/15/2017] [Accepted: 05/16/2017] [Indexed: 10/19/2022]
Abstract
Cell competition was described in Drosophila as the loss from mosaic tissues of otherwise viable cells heterozygous for Ribosomal protein mutations ('Minutes'). Cell competition has now been described to occur between multiple other genotypes, such as cells differing in myc expression levels, or mutated for neoplastic tumor suppressors. Recent studies implicate innate immunity components, and possibly mechanical stress, compression and cell intercalation as a consequence of differential growth rates in competitive cell death. Competition to eliminate pre-neoplastic tumors makes use of signals and receptors also used in patterning the nervous system including Slit/Robo2 and Sas/PTP10D to recognize and extrude clones of mutant cells, at least where local epithelial cyto-architecture is favorable. Cell competition facilitates expansion of Drosophila tumors through host tissue, and in normal development may promote developmental robustness and longevity by selecting for optimal progenitor cells.
Collapse
Affiliation(s)
- Nicholas E Baker
- Department of Genetics, Department of Developmental and Molecular Biology, Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, United States.
| |
Collapse
|
60
|
Germline Proliferation Is Regulated by Somatic Endocytic Genes via JNK and BMP Signaling in Drosophila. Genetics 2017; 206:189-197. [PMID: 28315838 PMCID: PMC5419469 DOI: 10.1534/genetics.116.196535] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 03/06/2017] [Indexed: 12/14/2022] Open
Abstract
Signals derived from the microenvironment contribute greatly to tumorigenesis . The underlying mechanism requires thorough investigation. Here, we use Drosophila testis as a model system to address this question, taking the advantage of the ease to distinguish germline and somatic cells and to track the cell numbers. In an EMS mutagenesis screen, we identified Rab5, a key factor in endocytosis, for its nonautonomous role in germline proliferation. The disruption of Rab5 in somatic cyst cells, which escort the development of germline lineage, induced the overproliferation of underdifferentiated but genetically wild-type germ cells. We demonstrated that this nonautonomous effect was mediated by the transcriptional activation of Dpp [the fly homolog of bone morphogenetic protein (BMP)] by examining the Dpp-reporter expression and knocking down Dpp to block germline overgrowth. Consistently, the protein levels of Bam, the germline prodifferentiation factor normally accumulated in the absence of BMP/Dpp signaling, decreased in the overproliferating germ cells. Further, we discovered that the JNK signaling pathway operated between Rab5 and Dpp, because simultaneously inhibiting the JNK pathway and Rab5 in cyst cells prevented both dpp transcription and germline tumor growth. Additionally, we found that multiple endocytic genes, such as avl, TSG101, Vps25, or Cdc42, were required in the somatic cyst cells to restrict germline amplification. These findings indicate that when the endocytic state of the surrounding cells is impaired, genetically wild-type germ cells overgrow. This nonautonomous model of tumorigenesis provides a simple system to dissect the relation between tumor and its niche.
Collapse
|
61
|
The ligand Sas and its receptor PTP10D drive tumour-suppressive cell competition. Nature 2017; 542:246-250. [DOI: 10.1038/nature21033] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 11/29/2016] [Indexed: 01/19/2023]
|
62
|
Xie G, Chen H, Jia D, Shu Z, Palmer WH, Huang YC, Zeng X, Hou SX, Jiao R, Deng WM. The SWI/SNF Complex Protein Snr1 Is a Tumor Suppressor in Drosophila Imaginal Tissues. Cancer Res 2016; 77:862-873. [PMID: 27923836 DOI: 10.1158/0008-5472.can-16-0963] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 11/02/2016] [Accepted: 11/18/2016] [Indexed: 12/17/2022]
Abstract
Components of the SWI/SNF chromatin-remodeling complex are among the most frequently mutated genes in various human cancers, yet only SMARCB1/hSNF5, a core member of the SWI/SNF complex, is mutated in malignant rhabdoid tumors (MRT). How SMARCB1/hSNF5 functions differently from other members of the SWI/SNF complex remains unclear. Here, we use Drosophila imaginal epithelial tissues to demonstrate that Snr1, the conserved homolog of human SMARCB1/hSNF5, prevents tumorigenesis by maintaining normal endosomal trafficking-mediated signaling cascades. Removal of Snr1 resulted in neoplastic tumorigenic overgrowth in imaginal epithelial tissues, whereas depletion of any other members of the SWI/SNF complex did not induce similar phenotypes. Unlike other components of the SWI/SNF complex that were detected only in the nucleus, Snr1 was observed in both the nucleus and the cytoplasm. Aberrant regulation of multiple signaling pathways, including Notch, JNK, and JAK/STAT, was responsible for tumor progression upon snr1-depletion. Our results suggest that the cytoplasmic Snr1 may play a tumor suppressive role in Drosophila imaginal tissues, offering a foundation for understanding the pivotal role of SMARCB1/hSNF5 in suppressing MRT during early childhood. Cancer Res; 77(4); 862-73. ©2017 AACR.
Collapse
Affiliation(s)
- Gengqiang Xie
- Department of Biological Science, Florida State University, Tallahassee, Florida
| | - Hanqing Chen
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, the Chinese Academy of Sciences, Beijing, China
| | - Dongyu Jia
- Department of Biological Science, Florida State University, Tallahassee, Florida
| | - Zhiqiang Shu
- Department of Biological Science, Florida State University, Tallahassee, Florida
| | - William Hunt Palmer
- Department of Biological Science, Florida State University, Tallahassee, Florida
| | - Yi-Chun Huang
- Department of Biological Science, Florida State University, Tallahassee, Florida
| | - Xiankun Zeng
- The Basic Research Laboratory, National Cancer Institute, National Institutes of Health, Frederick, Maryland
| | - Steven X Hou
- The Basic Research Laboratory, National Cancer Institute, National Institutes of Health, Frederick, Maryland
| | - Renjie Jiao
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, the Chinese Academy of Sciences, Beijing, China. .,Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Wu-Min Deng
- Department of Biological Science, Florida State University, Tallahassee, Florida.
| |
Collapse
|
63
|
Nemetschke L, Knust E. Drosophila Crumbs prevents ectopic Notch activation in developing wings by inhibiting ligand-independent endocytosis. Development 2016; 143:4543-4553. [DOI: 10.1242/dev.141762] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 10/21/2016] [Indexed: 12/13/2022]
Abstract
Many signalling components are apically restricted in epithelial cells, and receptor localisation and abundance is key for morphogenesis and tissue homeostasis. Hence, controlling apicobasal epithelial polarity is crucial for proper signalling. Notch is a ubiquitously expressed, apically localised receptor, which performs a plethora of functions; therefore, its activity has to be tightly regulated. Here, we show that Drosophila Crumbs, an evolutionarily conserved polarity determinant, prevents Notch endocytosis in developing wings through direct interaction between the two proteins. Notch endocytosis in the absence of Crumbs results in the activation of the ligand-independent, Deltex-dependent Notch signalling pathway, and does not require the ligands Delta and Serrate or γ-secretase activity. This function of Crumbs is not due to general defects in apicobasal polarity, as localisation of other apical proteins is unaffected. Our data reveal a mechanism to explain how Crumbs directly controls localisation and trafficking of the potent Notch receptor, and adds yet another aspect of Crumbs regulation in Notch pathway activity. Furthermore, our data highlight a close link between the apical determinant Crumbs, receptor trafficking and tissue homeostasis.
Collapse
Affiliation(s)
- Linda Nemetschke
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, Dresden 01307, Germany
| | - Elisabeth Knust
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, Dresden 01307, Germany
| |
Collapse
|
64
|
Zhou W, He Q, Zhang C, He X, Cui Z, Liu F, Li W. BLOS2 negatively regulates Notch signaling during neural and hematopoietic stem and progenitor cell development. eLife 2016; 5. [PMID: 27719760 PMCID: PMC5094856 DOI: 10.7554/elife.18108] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 10/04/2016] [Indexed: 12/28/2022] Open
Abstract
Notch signaling plays a crucial role in controling the proliferation and differentiation of stem and progenitor cells during embryogenesis or organogenesis, but its regulation is incompletely understood. BLOS2, encoded by the Bloc1s2 gene, is a shared subunit of two lysosomal trafficking complexes, biogenesis of lysosome-related organelles complex-1 (BLOC-1) and BLOC-1-related complex (BORC). Bloc1s2-/- mice were embryonic lethal and exhibited defects in cortical development and hematopoiesis. Loss of BLOS2 resulted in elevated Notch signaling, which consequently increased the proliferation of neural progenitor cells and inhibited neuronal differentiation in cortices. Likewise, ablation of bloc1s2 in zebrafish or mice led to increased hematopoietic stem and progenitor cell production in the aorta-gonad-mesonephros region. BLOS2 physically interacted with Notch1 in endo-lysosomal trafficking of Notch1. Our findings suggest that BLOS2 is a novel negative player in regulating Notch signaling through lysosomal trafficking to control multiple stem and progenitor cell homeostasis in vertebrates.
Collapse
Affiliation(s)
- Wenwen Zhou
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Qiuping He
- University of Chinese Academy of Sciences, Beijing, China.,State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Chunxia Zhang
- University of Chinese Academy of Sciences, Beijing, China.,State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Xin He
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Zongbin Cui
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Beijing, China
| | - Feng Liu
- University of Chinese Academy of Sciences, Beijing, China.,State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Wei Li
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.,Center for Medical Genetics, Beijing Children's Hospital, Capital Medical University, Beijing, China.,Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, China.,MOE Key Laboratory of Major Diseases in Children, Beijing, China.,Beijing Pediatric Research Institute, Beijing, China
| |
Collapse
|
65
|
Román-Fernández A, Bryant DM. Complex Polarity: Building Multicellular Tissues Through Apical Membrane Traffic. Traffic 2016; 17:1244-1261. [PMID: 27281121 DOI: 10.1111/tra.12417] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 06/06/2016] [Accepted: 06/06/2016] [Indexed: 12/20/2022]
Abstract
The formation of distinct subdomains of the cell surface is crucial for multicellular organism development. The most striking example of this is apical-basal polarization. What is much less appreciated is that underpinning an asymmetric cell surface is an equally dramatic intracellular endosome rearrangement. Here, we review the interplay between classical cell polarity proteins and membrane trafficking pathways, and discuss how this marriage gives rise to cell polarization. We focus on those mechanisms that regulate apical polarization, as this is providing a number of insights into how membrane traffic and polarity are regulated at the tissue level.
Collapse
Affiliation(s)
- Alvaro Román-Fernández
- Cancer Research UK Beatson Institute, Switchback Road, Glasgow, G61 1BD, UK.,Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - David M Bryant
- Cancer Research UK Beatson Institute, Switchback Road, Glasgow, G61 1BD, UK.,Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| |
Collapse
|
66
|
Abstract
The study of Drosophila imaginal discs has contributed to a number of discoveries in developmental and cellular biology. In addition to the elucidation of the role of tissue compartments and organ-specific master regulator genes during development, imaginal discs have also become well established as models for studying cellular interactions and complex genetic pathways. Here, we review key discoveries resulting from investigations of these epithelial precursor organs, ranging from cell fate determination and transdetermination to tissue patterning. Furthermore, the design of increasingly sophisticated genetic tools over the last decades has added value to the use of imaginal discs as model systems. As a result of tissue-specific genetic screens, several components of developmentally regulated signaling pathways were identified and epistasis revealed the levels at which they function. Discs have been widely used to assess cellular interactions in their natural tissue context, contributing to a better understanding of growth regulation, tissue regeneration, and cancer. With the continuous implementation of novel tools, imaginal discs retain significant potential as model systems to address emerging questions in biology and medicine.
Collapse
|
67
|
Regulation of Notch Signaling Through Intracellular Transport. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 323:107-27. [PMID: 26944620 DOI: 10.1016/bs.ircmb.2015.12.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The highly conserved Notch-signaling pathway performs a central role in cell differentiation, survival, and proliferation. A major mechanism by which cells modulate signaling is by controlling the intracellular transport itinerary of Notch. Indeed, Notch removal from the cell surface and its targeting to the lysosome for degradation is one way in which Notch activity is downregulated since it limits receptor exposure to ligand. In contrast, Notch-signaling capacity is maintained through repeated rounds of receptor recycling and redelivery of Notch to the cell surface from endosomal stores. This review discusses the molecular mechanisms by which Notch transit through the endosome is controlled and how various intracellular sorting decisions are thought to impact signaling activity.
Collapse
|
68
|
La Fortezza M, Schenk M, Cosolo A, Kolybaba A, Grass I, Classen AK. JAK/STAT signalling mediates cell survival in response to tissue stress. Development 2016; 143:2907-19. [DOI: 10.1242/dev.132340] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Accepted: 06/23/2016] [Indexed: 12/31/2022]
Abstract
Tissue homeostasis relies on the ability of tissues to respond to stress. Tissue regeneration and tumour models in Drosophila have shown that JNK is a prominent stress-response pathway promoting injury-induced apoptosis and compensatory proliferation. A central question remaining unanswered is how both responses are balanced by activation of a single pathway. JAK/STAT signalling, a potential JNK target, is implicated in promoting compensatory proliferation. While we observe JAK/STAT activation in imaginal discs upon damage, our data demonstrates that JAK/STAT and its downstream effector Zfh2 promote survival of JNK-signalling cells instead. The JNK component fos and the pro-apoptotic gene hid are regulated in a JAK/STAT-dependent manner. This molecular pathway restrains JNK-induced apoptosis and spatial propagation of JNK-signalling, thereby limiting the extent of tissue damage, as well as facilitating systemic and proliferative responses to injury. We find that the pro-survival function of JAK/STAT also drives tumour growth under conditions of chronic stress. Our study defines JAK/STAT function in tissue stress and illustrates how crosstalk between conserved signalling pathways establishes an intricate equilibrium between proliferation, apoptosis and survival to restore tissue homeostasis.
Collapse
Affiliation(s)
- Marco La Fortezza
- Ludwig-Maximilians-University Munich, Faculty of Biology, Grosshaderner Strasse 2-4, 82152 Planegg-Martinsried, Germany
| | - Madlin Schenk
- Ludwig-Maximilians-University Munich, Faculty of Biology, Grosshaderner Strasse 2-4, 82152 Planegg-Martinsried, Germany
| | - Andrea Cosolo
- Ludwig-Maximilians-University Munich, Faculty of Biology, Grosshaderner Strasse 2-4, 82152 Planegg-Martinsried, Germany
| | - Addie Kolybaba
- Ludwig-Maximilians-University Munich, Faculty of Biology, Grosshaderner Strasse 2-4, 82152 Planegg-Martinsried, Germany
| | - Isabelle Grass
- Ludwig-Maximilians-University Munich, Faculty of Biology, Grosshaderner Strasse 2-4, 82152 Planegg-Martinsried, Germany
| | - Anne-Kathrin Classen
- Ludwig-Maximilians-University Munich, Faculty of Biology, Grosshaderner Strasse 2-4, 82152 Planegg-Martinsried, Germany
| |
Collapse
|
69
|
|
70
|
|
71
|
BOLKENT Ş, ÖZTAY F, GEZGİNCİ OKTAYOĞLU S, SANCAR BAŞ S, KARATUĞ A. A matter of regeneration and repair: caspases as the key molecules. Turk J Biol 2016. [DOI: 10.3906/biy-1507-18] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
|
72
|
Feng D, Li Q, Yu H, Zhao X, Kong L. Comparative Transcriptome Analysis of the Pacific Oyster Crassostrea gigas Characterized by Shell Colors: Identification of Genetic Bases Potentially Involved in Pigmentation. PLoS One 2015; 10:e0145257. [PMID: 26693729 PMCID: PMC4691203 DOI: 10.1371/journal.pone.0145257] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 11/30/2015] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Shell color polymorphisms of Mollusca have contributed to development of evolutionary biology and population genetics, while the genetic bases and molecular mechanisms underlying shell pigmentation are poorly understood. The Pacific oyster (Crassostrea gigas) is one of the most important farmed oysters worldwide. Through successive family selection, four shell color variants (white, golden, black and partially pigmented) of C. gigas have been developed. To elucidate the genetic mechanisms of shell coloration in C. gigas and facilitate the selection of elite oyster lines with desired coloration patterns, differentially expressed genes (DEGs) were identified among the four shell color variants by RNA-seq. RESULTS Digital gene expression generated over fifteen million reads per sample, producing expression data for 28,027 genes. A total number of 2,645 DEGs were identified from pair-wise comparisons, of which 432, 91, 43 and 39 genes specially were up-regulated in white, black, golden and partially pigmented shell of C. gigas, respectively. Three genes of Abca1, Abca3 and Abcb1 which belong to the ATP-binding cassette (ABC) transporters super-families were significantly associated with white shell formation. A tyrosinase transcript (CGI_10008737) represented consistent up-regulated pattern with golden coloration. We proposed that white shell variant of C. gigas could employ "endocytosis" to down-regulate notch level and to prevent shell pigmentation. CONCLUSION This study discovered some potential shell coloration genes and related molecular mechanisms by the RNA-seq, which would provide foundational information to further study on shell coloration and assist in selective breeding in C. gigas.
Collapse
Affiliation(s)
- Dandan Feng
- Key Laboratory of Mariculture Ministry of Education, Ocean University of China, Qingdao, China
| | - Qi Li
- Key Laboratory of Mariculture Ministry of Education, Ocean University of China, Qingdao, China
| | - Hong Yu
- Key Laboratory of Mariculture Ministry of Education, Ocean University of China, Qingdao, China
| | - Xuelin Zhao
- Key Laboratory of Mariculture Ministry of Education, Ocean University of China, Qingdao, China
| | - Lingfeng Kong
- Key Laboratory of Mariculture Ministry of Education, Ocean University of China, Qingdao, China
| |
Collapse
|
73
|
Ellis K, Friedman C, Yedvobnick B. Drosophila domino Exhibits Genetic Interactions with a Wide Spectrum of Chromatin Protein-Encoding Loci. PLoS One 2015; 10:e0142635. [PMID: 26555684 PMCID: PMC4640824 DOI: 10.1371/journal.pone.0142635] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 10/23/2015] [Indexed: 11/18/2022] Open
Abstract
The Drosophila domino gene encodes protein of the SWI2/SNF2 family that has widespread roles in transcription, replication, recombination and DNA repair. Here, the potential relationship of Domino protein to other chromatin-associated proteins has been investigated through a genetic interaction analysis. We scored for genetic modification of a domino wing margin phenotype through coexpression of RNAi directed against a set of previously characterized and more newly characterized chromatin-encoding loci. A set of other SWI2/SNF2 loci were also assayed for interaction with domino. Our results show that the majority of tested loci exhibit synergistic enhancement or suppression of the domino wing phenotype. Therefore, depression in domino function sensitizes the wing margin to alterations in the activity of numerous chromatin components. In several cases the genetic interactions are associated with changes in the level of cell death measured across the dorsal-ventral margin of the wing imaginal disc. These results highlight the broad realms of action of many chromatin proteins and suggest significant overlap with Domino function in fundamental cell processes, including cell proliferation, cell death and cell signaling.
Collapse
Affiliation(s)
- Kaitlyn Ellis
- Biology Department, Emory University, Atlanta, Georgia, United States of America
| | - Chloe Friedman
- Biology Department, Emory University, Atlanta, Georgia, United States of America
| | - Barry Yedvobnick
- Biology Department, Emory University, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
74
|
Lin YH, Currinn H, Pocha SM, Rothnie A, Wassmer T, Knust E. AP-2-complex-mediated endocytosis of Drosophila Crumbs regulates polarity by antagonizing Stardust. J Cell Sci 2015; 128:4538-49. [PMID: 26527400 DOI: 10.1242/jcs.174573] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 10/26/2015] [Indexed: 12/21/2022] Open
Abstract
Maintenance of epithelial polarity depends on the correct localization and levels of polarity determinants. The evolutionarily conserved transmembrane protein Crumbs is crucial for the size and identity of the apical membrane, yet little is known about the molecular mechanisms controlling the amount of Crumbs at the surface. Here, we show that Crumbs levels on the apical membrane depend on a well-balanced state of endocytosis and stabilization. The adaptor protein 2 (AP-2) complex binds to a motif in the cytoplasmic tail of Crumbs that overlaps with the binding site of Stardust, a protein known to stabilize Crumbs on the surface. Preventing endocytosis by mutation of AP-2 causes expansion of the Crumbs-positive plasma membrane domain and polarity defects, which can be partially rescued by removing one copy of crumbs. Strikingly, knocking down both AP-2 and Stardust leads to the retention of Crumbs on the membrane. This study provides evidence for a molecular mechanism, based on stabilization and endocytosis, to adjust surface levels of Crumbs, which are essential for maintaining epithelial polarity.
Collapse
Affiliation(s)
- Ya-Huei Lin
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, Dresden 01307, Germany
| | - Heather Currinn
- School of Life and Health Sciences, Aston University, Aston Triangle, Birmingham B4 7ET, UK
| | - Shirin Meher Pocha
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, Dresden 01307, Germany
| | - Alice Rothnie
- School of Life and Health Sciences, Aston University, Aston Triangle, Birmingham B4 7ET, UK
| | - Thomas Wassmer
- School of Life and Health Sciences, Aston University, Aston Triangle, Birmingham B4 7ET, UK
| | - Elisabeth Knust
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, Dresden 01307, Germany
| |
Collapse
|
75
|
Enomoto M, Vaughen J, Igaki T. Non-autonomous overgrowth by oncogenic niche cells: Cellular cooperation and competition in tumorigenesis. Cancer Sci 2015; 106:1651-8. [PMID: 26362609 PMCID: PMC4714670 DOI: 10.1111/cas.12816] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 09/02/2015] [Accepted: 09/08/2015] [Indexed: 12/23/2022] Open
Abstract
Tumor progression is classically viewed as the Darwinian evolution of subclones that sequentially acquire genetic mutations and autonomously overproliferate. However, growing evidence suggests that tumor microenvironment and subclone heterogeneity contribute to non‐autonomous tumor progression. Recent Drosophila studies revealed a common mechanism by which clones of genetically altered cells trigger non‐autonomous overgrowth. Such “oncogenic niche cells” (ONCs) do not overgrow but instead stimulate neighbor overgrowth and metastasis. Establishment of ONCs depends on competition and cooperation between heterogeneous cell populations. This review characterizes diverse ONCs identified in Drosophila and describes the genetic basis of non‐autonomous tumor progression. Similar mechanisms may contribute to mammalian cancer progression and recurrence.
Collapse
Affiliation(s)
- Masato Enomoto
- Laboratory of Genetics, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - John Vaughen
- Laboratory of Genetics, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Tatsushi Igaki
- Laboratory of Genetics, Graduate School of Biostudies, Kyoto University, Kyoto, Japan.,PRESTO, Japan Science and Technology Agency, Saitama, Japan
| |
Collapse
|
76
|
Palmer WH, Deng WM. Ligand-Independent Mechanisms of Notch Activity. Trends Cell Biol 2015; 25:697-707. [PMID: 26437585 DOI: 10.1016/j.tcb.2015.07.010] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 07/10/2015] [Accepted: 07/29/2015] [Indexed: 10/23/2022]
Abstract
Interaction between the Notch receptor and Delta-Serrate-Lag2 (DSL) ligands is generally deemed to be the starting point of the Notch signaling cascade, after which, Notch is cleaved and the intracellular domain acts as a transcriptional coactivator. By contrast, Notch protein can become activated independent of ligand stimulus through recently identified endosomal trafficking routes as well as through aberrant regulation of Notch components during Notch trafficking, ubiquitination, and degradation. In this review, we summarize genes implicated in ligand-independent Notch activity and remark on the mechanisms by which this process could occur.
Collapse
Affiliation(s)
- William Hunt Palmer
- Department of Biological Science, Florida State University, Tallahassee, FL 32306-4295, USA; Current Address: Institute of Evolutionary Biology, University of Edinburgh, Edinburgh, UK
| | - Wu-Min Deng
- Department of Biological Science, Florida State University, Tallahassee, FL 32306-4295, USA.
| |
Collapse
|
77
|
Gomez-Lamarca MJ, Snowdon LA, Seib E, Klein T, Bray SJ. Rme-8 depletion perturbs Notch recycling and predisposes to pathogenic signaling. J Cell Biol 2015; 210:303-18. [PMID: 26169355 PMCID: PMC4508892 DOI: 10.1083/jcb.201411001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 06/11/2015] [Indexed: 02/07/2023] Open
Abstract
The retromer-associated DNAJ protein Rme-8 is necessary for normal Notch recycling, and reductions in Rme-8 sensitize cells so that additional loss-of-sorting retromer or ESCRT-0 components have catastrophic effects. Notch signaling is a major regulator of cell fate, proliferation, and differentiation. Like other signaling pathways, its activity is strongly influenced by intracellular trafficking. Besides contributing to signal activation and down-regulation, differential fluxes between trafficking routes can cause aberrant Notch pathway activation. Investigating the function of the retromer-associated DNAJ protein Rme-8 in vivo, we demonstrate a critical role in regulating Notch receptor recycling. In the absence of Rme-8, Notch accumulated in enlarged tubulated Rab4-positive endosomes, and as a consequence, signaling was compromised. Strikingly, when the retromer component Vps26 was depleted at the same time, Notch no longer accumulated and instead was ectopically activated. Likewise, depletion of ESCRT-0 components Hrs or Stam in combination with Rme-8 also led to high levels of ectopic Notch activity. Together, these results highlight the importance of Rme-8 in coordinating normal endocytic recycling route and reveal that its absence predisposes toward conditions in which pathological Notch signaling can occur.
Collapse
Affiliation(s)
- Maria J Gomez-Lamarca
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, England, UK
| | - Laura A Snowdon
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, England, UK
| | - Ekatarina Seib
- Institute of Genetics, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Thomas Klein
- Institute of Genetics, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Sarah J Bray
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, England, UK
| |
Collapse
|
78
|
Li Z, Blissard G. The vacuolar protein sorting genes in insects: A comparative genome view. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2015; 62:211-225. [PMID: 25486452 DOI: 10.1016/j.ibmb.2014.11.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 11/06/2014] [Accepted: 11/21/2014] [Indexed: 06/04/2023]
Abstract
In eukaryotic cells, regulated vesicular trafficking is critical for directing protein transport and for recycling and degradation of membrane lipids and proteins. Through carefully regulated transport vesicles, the endomembrane system performs a large and important array of dynamic cellular functions while maintaining the integrity of the cellular membrane system. Genetic studies in yeast Saccharomyces cerevisiae have identified approximately 50 vacuolar protein sorting (VPS) genes involved in vesicle trafficking, and most of these genes are also characterized in mammals. The VPS proteins form distinct functional complexes, which include complexes known as ESCRT, retromer, CORVET, HOPS, GARP, and PI3K-III. Little is known about the orthologs of VPS proteins in insects. Here, with the newly annotated Manduca sexta genome, we carried out genomic comparative analysis of VPS proteins in yeast, humans, and 13 sequenced insect genomes representing the Orders Hymenoptera, Diptera, Hemiptera, Phthiraptera, Lepidoptera, and Coleoptera. Amino acid sequence alignments and domain/motif structure analyses reveal that most of the components of ESCRT, retromer, CORVET, HOPS, GARP, and PI3K-III are evolutionarily conserved across yeast, insects, and humans. However, in contrast to the VPS gene expansions observed in the human genome, only four VPS genes (VPS13, VPS16, VPS33, and VPS37) were expanded in the six insect Orders. Additionally, VPS2 was expanded only in species from Phthiraptera, Lepidoptera, and Coleoptera. These studies provide a baseline for understanding the evolution of vesicular trafficking across yeast, insect, and human genomes, and also provide a basis for further addressing specific functional roles of VPS proteins in insects.
Collapse
Affiliation(s)
- Zhaofei Li
- State Key Laboratory of Crop Stress Biology for Arid Areas, Key Laboratory of Northwest Loess Plateau Crop Pest Management of Ministry of Agriculture, College of Plant Protection, Northwest A&F University, Taicheng Road, Yangling, Shaanxi 712100, China.
| | - Gary Blissard
- Boyce Thompson Institute, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
79
|
Juan T, Fürthauer M. [The ESCRT complex: from endosomal transport to the development of multicellular organisms]. Biol Aujourdhui 2015; 209:111-124. [PMID: 26115716 DOI: 10.1051/jbio/2015009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Indexed: 06/04/2023]
Abstract
Since its discovery more than 50 years ago, the endo-lysosomal system has emerged as a central integrator of different cellular activities. This vesicular trafficking apparatus governs processes as diverse as the transduction of stimuli by growth factor receptors, the recycling and secretion of signaling molecules and the regulation of cellular homeostasis through autophagy. Accordingly, dysfunctions of the vesicular transport machinery have been linked to a growing number of pathologies. In this review we take the "Endosomal Sorting Complex Required for Transport" (ESCRT) as an example to illustrate the multiple functions of an evolutionarily conserved endosomal transport machinery. We describe the major concepts that have emerged from the study of this machinery at the level of the development and the physiology of multi-cellular organisms. In particular, we highlight the essential contributions of ESCRT proteins on the regulation of three biological processes: the endocytic regulation of cell signaling, autophagy and its role in neuronal morphogenesis and finally the biogenesis and function of extracellular vesicles.
Collapse
|
80
|
Legent K, Liu HH, Treisman JE. Drosophila Vps4 promotes Epidermal growth factor receptor signaling independently of its role in receptor degradation. Development 2015; 142:1480-91. [PMID: 25790850 DOI: 10.1242/dev.117960] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 02/20/2015] [Indexed: 12/12/2022]
Abstract
Endocytic trafficking of signaling receptors is an important mechanism for limiting signal duration. Components of the Endosomal Sorting Complexes Required for Transport (ESCRT), which target ubiquitylated receptors to intra-lumenal vesicles (ILVs) of multivesicular bodies, are thought to terminate signaling by the epidermal growth factor receptor (EGFR) and direct it for lysosomal degradation. In a genetic screen for mutations that affect Drosophila eye development, we identified an allele of Vacuolar protein sorting 4 (Vps4), which encodes an AAA ATPase that interacts with the ESCRT-III complex to drive the final step of ILV formation. Photoreceptors are largely absent from Vps4 mutant clones in the eye disc, and even when cell death is genetically prevented, the mutant R8 photoreceptors that develop fail to recruit surrounding cells to differentiate as R1-R7 photoreceptors. This recruitment requires EGFR signaling, suggesting that loss of Vps4 disrupts the EGFR pathway. In imaginal disc cells mutant for Vps4, EGFR and other receptors accumulate in endosomes and EGFR target genes are not expressed; epistasis experiments place the function of Vps4 at the level of the receptor. Surprisingly, Vps4 is required for EGFR signaling even in the absence of Shibire, the Dynamin that internalizes EGFR from the plasma membrane. In ovarian follicle cells, in contrast, Vps4 does not affect EGFR signaling, although it is still essential for receptor degradation. Taken together, these findings indicate that Vps4 can promote EGFR activity through an endocytosis-independent mechanism.
Collapse
Affiliation(s)
- Kevin Legent
- Kimmel Center for Biology and Medicine of the Skirball Institute and Department of Cell Biology, NYU School of Medicine, 540 First Avenue, New York, NY 10016, USA
| | - Hui Hua Liu
- Kimmel Center for Biology and Medicine of the Skirball Institute and Department of Cell Biology, NYU School of Medicine, 540 First Avenue, New York, NY 10016, USA
| | - Jessica E Treisman
- Kimmel Center for Biology and Medicine of the Skirball Institute and Department of Cell Biology, NYU School of Medicine, 540 First Avenue, New York, NY 10016, USA
| |
Collapse
|
81
|
Patel M, Antala B, Shrivastava N. In silico screening of alleged miRNAs associated with cell competition: an emerging cellular event in cancer. ACTA ACUST UNITED AC 2015; 20:798-815. [DOI: 10.1515/cmble-2015-0046] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Accepted: 10/20/2015] [Indexed: 01/13/2023]
Abstract
AbstractCell competition is identified as a crucial phenomenon for cancer and organ development. There is a possibility that microRNAs (miRNAs) may play an important role in the regulation of expression of genes involved in cell competition. In silico screening of miRNAs is an effort to abridge, economize and expedite the experimental approaches to identification of potential miRNAs involved in cell competition, as no study has reported involvement of miRNAs in cell competition to date. In this study, we used multiple screening steps as follows: (i) selection of cell competition related genes of Drosophila through a literature survey; (ii) homology study of selected cell competition related genes; (iii) identification of miRNAs that target conserved cell competitionrelated genes through prediction tools; (iv) sequence conservation analysis of identified miRNAs with human genome; (v) identification of conserved cell competition miRNAs using their expression profiles and exploration of roles of their homologous human miRNAs. This study led to the identification of nine potential cell competition miRNAs in the Drosophila genome. Importantly, eighteen human homologs of these nine potential Drosophila miRNAs are well reported for their involvement in different types of cancers. This confirms their probable involvement in cell competition as well, because cell competition is well justified for its involvement in cancer initiation and maintenance.
Collapse
|
82
|
Shen J, Liu Y, Song Y, Li L, Duan C, Zhou Y, Ke K. CHMP4B, ESCRT-III associating protein, associated with neuronal apoptosis following intracerebral hemorrhage. Brain Res 2014; 1597:1-13. [PMID: 25478783 DOI: 10.1016/j.brainres.2014.11.043] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 11/19/2014] [Accepted: 11/20/2014] [Indexed: 12/17/2022]
Abstract
Charged multivesicular body protein (CHMP) represents a family of small helical proteins that contain an N-terminal basically charged region and a smaller C-terminal acidic region, which are highly conserved in all eukaryotes. CHMP4B, a core component of the endosomal sorting complex required for transport (ESCRT)-III, is requisite for endosomal sorting and other biological processes. Here, we demonstrate that CHMP4B may be involved in neuronal apoptosis in the processes of intracerebral hemorrhage (ICH). From the results of Western blot, immunohistochemistry and immunofluorescence, we obtained a significant up-regulation of CHMP4B in neurons adjacent to the hematoma following ICH. Increasing CHMP4B level was found to be accompanied by the up-regulation of Fas receptor (Fas), Fas ligand (FasL), active caspase-8, and active caspase-3. Besides, CHMP4B co-localized well with Fas and active caspase-3 in neurons, indicating its potential role in neuronal apoptosis. What's more, our in vitro study, using CHMP4B RNA interference in PC12 cells, further confirmed that CHMP4B might exert its pro-apoptotic function on neuronal apoptosis through extrinsic pathway. Thus, CHMP4B may play a role in promoting the brain damage following ICH.
Collapse
Affiliation(s)
- Jiabing Shen
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China; Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong, Jiangsu Province 226001, China
| | - Yonghua Liu
- Department of Pathogen Biology, Medical College, Nantong University, Nantong 226001, Jiangsu Province, China; Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong, Jiangsu Province 226001, China
| | - Yan Song
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China; Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong, Jiangsu Province 226001, China
| | - Lei Li
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Chengwei Duan
- The Jiangsu Province Key Laboratory of Neuroregeneration, Nantong University, Nantong, China; Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong, Jiangsu Province 226001, China
| | - Ying Zhou
- Department of Pathogen Biology, Medical College, Nantong University, Nantong 226001, Jiangsu Province, China; Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong, Jiangsu Province 226001, China
| | - Kaifu Ke
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China; Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong, Jiangsu Province 226001, China.
| |
Collapse
|
83
|
Takino K, Ohsawa S, Igaki T. Loss of Rab5 drives non-autonomous cell proliferation through TNF and Ras signaling in Drosophila. Dev Biol 2014; 395:19-28. [PMID: 25224221 DOI: 10.1016/j.ydbio.2014.09.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 08/26/2014] [Accepted: 09/05/2014] [Indexed: 01/16/2023]
Abstract
Deregulation of the endocytic machinery has been implicated in human cancers. However, the mechanism by which endocytic defects drive cancer development remains to be clarified. Here, we find through a genetic screen in Drosophila that loss of Rab5, a protein required for early endocytic trafficking, drives non-autonomous cell proliferation in imaginal epithelium. Our genetic data indicate that dysfunction of Rab5 leads to cell-autonomous accumulation of Eiger (a TNF homolog) and EGF receptor (EGFR), which causes activation of downstream JNK and Ras signaling, respectively. JNK signaling and its downstream component Cdc42 cooperate with Ras signaling to induce upregulation of a secreted growth factor Upd (an IL-6 homolog) through inactivation of the Hippo pathway. Such non-autonomous tissue growth triggered by Rab5 defect could contribute to epithelial homeostasis as well as cancer development within heterogeneous tumor microenvironment.
Collapse
Affiliation(s)
- Kyoko Takino
- Laboratory of Genetics, Graduate School of Biostudies, Kyoto University, Yoshida-Konoecho-cho, Sakyo-ku, Kyoto 606-8501, Japan; Division of Genetics, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Shizue Ohsawa
- Laboratory of Genetics, Graduate School of Biostudies, Kyoto University, Yoshida-Konoecho-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Tatsushi Igaki
- Laboratory of Genetics, Graduate School of Biostudies, Kyoto University, Yoshida-Konoecho-cho, Sakyo-ku, Kyoto 606-8501, Japan; PRESTO, Japan Science and Technology Agency (JST), 4-1-8 Honcho Kawaguchi, Saitama 332-0012, Japan.
| |
Collapse
|
84
|
Parsons LM, Portela M, Grzeschik NA, Richardson HE. Lgl regulates Notch signaling via endocytosis, independently of the apical aPKC-Par6-Baz polarity complex. Curr Biol 2014; 24:2073-2084. [PMID: 25220057 DOI: 10.1016/j.cub.2014.07.075] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 07/01/2014] [Accepted: 07/28/2014] [Indexed: 12/17/2022]
Abstract
BACKGROUND The Drosophila melanogaster junctional neoplastic tumor suppressor, Lethal-2-giant larvae (Lgl), is a regulator of apicobasal cell polarity and tissue growth. We have previously shown in the developing Drosophila eye epithelium that, without affecting cell polarity, depletion of Lgl results in ectopic cell proliferation and blockage of developmental cell death due to deregulation of the Hippo signaling pathway. RESULTS Here, we show that Notch signaling is increased in lgl-depleted eye tissue, independently of Lgl's function in apicobasal cell polarity. The upregulation of Notch signaling is ligand dependent and correlates with accumulation of cleaved Notch. Concomitant with higher cleaved Notch levels in lgl- tissue, early endosomes (Avalanche [Avl+]), recycling endosomes (Rab11+), early multivesicular bodies (Hrs+), and acidified vesicles, but not late endosomal markers (Car+ and Rab7+), accumulate. Colocalization studies revealed that Lgl associates with early to late endosomes and lysosomes. Upregulation of Notch signaling in lgl- tissue requires dynamin- and Rab5-mediated endocytosis and vesicle acidification but is independent of Hrs/Stam or Rab11 activity. Furthermore, Lgl regulates Notch signaling independently of the aPKC-Par6-Baz apical polarity complex. CONCLUSIONS Altogether, our data show that Lgl regulates endocytosis to restrict vesicle acidification and prevent ectopic ligand-dependent Notch signaling. This Lgl function is independent of the aPKC-Par6-Baz polarity complex and uncovers a novel attenuation mechanism of ligand-activated Notch signaling during Drosophila eye development.
Collapse
Affiliation(s)
- Linda M Parsons
- Cell Cycle and Development Laboratory, Research Division, Peter MacCallum Cancer Centre, 7 St. Andrew's Place, East Melbourne, Melbourne, VIC 3002, Australia; Department of Genetics, University of Melbourne, 1-100 Grattan Street, Parkville, Melbourne, VIC 3010, Australia
| | - Marta Portela
- Cell Cycle and Development Laboratory, Research Division, Peter MacCallum Cancer Centre, 7 St. Andrew's Place, East Melbourne, Melbourne, VIC 3002, Australia
| | - Nicola A Grzeschik
- Cell Cycle and Development Laboratory, Research Division, Peter MacCallum Cancer Centre, 7 St. Andrew's Place, East Melbourne, Melbourne, VIC 3002, Australia
| | - Helena E Richardson
- Cell Cycle and Development Laboratory, Research Division, Peter MacCallum Cancer Centre, 7 St. Andrew's Place, East Melbourne, Melbourne, VIC 3002, Australia; Sir Peter MacCallum Department of Oncology, Peter MacCallum Cancer Centre, 7 St. Andrew's Place, East Melbourne, Melbourne, VIC 3002, Australia; Department of Biochemistry and Molecular Biology, University of Melbourne, 1-100 Grattan Street, Parkville, Melbourne, VIC 3010, Australia; Department of Anatomy and Neuroscience, University of Melbourne, 1-100 Grattan Street, Parkville, Melbourne, VIC 3010, Australia.
| |
Collapse
|
85
|
de Vreede G, Schoenfeld JD, Windler SL, Morrison H, Lu H, Bilder D. The Scribble module regulates retromer-dependent endocytic trafficking during epithelial polarization. Development 2014; 141:2796-802. [PMID: 25005475 DOI: 10.1242/dev.105403] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Scribble (Scrib) module proteins are major regulators of cell polarity, but how they influence membrane traffic is not known. Endocytosis is also a key regulator of polarity through roles that remain unclear. Here we link Scrib to a specific arm of the endocytic trafficking system. Drosophila mutants that block AP-2-dependent endocytosis share many phenotypes with Scrib module mutants, but Scrib module mutants show intact internalization and endolysosomal transport. However, defective traffic of retromer pathway cargo is seen, and retromer components show strong genetic interactions with the Scrib module. The Scrib module is required for proper retromer localization to endosomes and promotes appropriate cargo sorting into the retromer pathway via both aPKC-dependent and -independent mechanisms. We propose that the Scrib module regulates epithelial polarity by influencing endocytic itineraries of Crumbs and other retromer-dependent cargo.
Collapse
Affiliation(s)
- Geert de Vreede
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720-3200, USA
| | - Joshua D Schoenfeld
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720-3200, USA
| | - Sarah L Windler
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720-3200, USA
| | - Holly Morrison
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720-3200, USA
| | - Han Lu
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720-3200, USA
| | - David Bilder
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720-3200, USA
| |
Collapse
|
86
|
Penzo-Méndez AI, Stanger BZ. Cell competition in vertebrate organ size regulation. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2014; 3:419-27. [PMID: 25176591 DOI: 10.1002/wdev.148] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 06/25/2014] [Accepted: 06/30/2014] [Indexed: 01/24/2023]
Abstract
The study of animal organ size determination has provided evidence of the existence of organ-intrinsic mechanisms that 'sense' and adjust organ growth. Cell competition, a form of cell interaction that equalizes cell population growth, has been proposed to play a role in organ size regulation. Cell competition involves a cell-context dependent response triggered by perceived differences in cell growth and/or proliferation rates, resulting in apoptosis in growth-disadvantaged cells and compensatory expansion of the more 'fit' cells. The mechanisms that allow cells to compare growth are not yet understood, but a number of genes and pathways have been implicated in cell competition. These include Myc, the members of the Hippo, JAK/STAT and WNT signaling pathways, and the Dlg/Lgl/Scrib and the Crb/Std/PatJ membrane protein complexes. Cell competition was initially characterized in the Drosophila imaginal disc, but several recent studies have shown that cell competition occurs in mouse embryonic stem cells and in the embryonic epiblast, where it plays a role in the regulation of early embryo size. In addition, competition-like behavior has been described in the adult mouse liver and the hematopoietic stem cell compartment. These data indicate that cell competition plays a more universal role in organ size regulation. In addition, as some authors have suggested that similar types of competitive behavior may operate in during tumorigenesis, there may be additional practical reasons for understanding this fundamental process of intercellular communication.
Collapse
Affiliation(s)
- Alfredo I Penzo-Méndez
- Gastroenterology Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Cell and Developmental Biology, Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | |
Collapse
|
87
|
Ohsawa S, Takemoto D, Igaki T. Dissecting tumour heterogeneity in flies: genetic basis of interclonal oncogenic cooperation. J Biochem 2014; 156:129-36. [PMID: 25012819 DOI: 10.1093/jb/mvu045] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Cancers develop through sequential acquisition of oncogenic mutations, indicating a crucial role of genetic alterations in tumour progression. However, accumulating evidence suggests that cancers also progress towards malignancy through cell-cell interactions within heterogeneous tumour tissue. Recent studies have indicated that tumour heterogeneity not only contributes to drug resistance and tumour recurrence but also plays an active role in promoting tumour progression. Especially, genetic studies in Drosophila have discovered novel types of tumour progression through cell-cell interactions and have dissected the underlying mechanisms. This review focuses on describing recent findings obtained from Drosophila genetics that provide genetic basis of interclonal oncogenic cooperation in heterogeneous tumour tissue.
Collapse
Affiliation(s)
- Shizue Ohsawa
- Laboratory of Genetics, Graduate School of Biostudies, Kyoto University, Yoshida-Konoecho-cho, Sakyo-ku, Kyoto 606-8501, Japan; and PRESTO, Japan Science and Technology Agency (JST), 4-1-8 Honcho Kawaguchi, Saitama 332-0012, Japan
| | - Daisaku Takemoto
- Laboratory of Genetics, Graduate School of Biostudies, Kyoto University, Yoshida-Konoecho-cho, Sakyo-ku, Kyoto 606-8501, Japan; and PRESTO, Japan Science and Technology Agency (JST), 4-1-8 Honcho Kawaguchi, Saitama 332-0012, Japan
| | - Tatsushi Igaki
- Laboratory of Genetics, Graduate School of Biostudies, Kyoto University, Yoshida-Konoecho-cho, Sakyo-ku, Kyoto 606-8501, Japan; and PRESTO, Japan Science and Technology Agency (JST), 4-1-8 Honcho Kawaguchi, Saitama 332-0012, Japan Laboratory of Genetics, Graduate School of Biostudies, Kyoto University, Yoshida-Konoecho-cho, Sakyo-ku, Kyoto 606-8501, Japan; and PRESTO, Japan Science and Technology Agency (JST), 4-1-8 Honcho Kawaguchi, Saitama 332-0012, Japan
| |
Collapse
|
88
|
Mattissek C, Teis D. The role of the endosomal sorting complexes required for transport (ESCRT) in tumorigenesis. Mol Membr Biol 2014; 31:111-9. [PMID: 24641493 PMCID: PMC4059258 DOI: 10.3109/09687688.2014.894210] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 01/30/2014] [Accepted: 02/07/2014] [Indexed: 11/30/2022]
Abstract
The endosomal sorting complexes required for transport (ESCRT) are needed for three distinct cellular functions in higher eukaryotes: (i) Multivesicular body formation for the degradation of transmembrane proteins in lysosomes, (ii) midbody abscission during cytokinesis and (iii) retroviral budding. Not surprisingly, loss of ESCRT function has severe consequences, which include the failure to down-regulate growth factor receptors leading to deregulated mitogenic signaling. While it is clear that the function of the ESCRT machinery is important for embryonic development, its role in cancer is more controversial. Various experimental approaches in different model organisms arrive at partially divergent conclusions regarding the contribution of ESCRTs to tumorigenesis. Therefore the aim of this review is to provide an overview on different model systems used to study the role of the ESCRT machinery in cancer development, to highlight common grounds and present certain controversies in the field.
Collapse
Affiliation(s)
- Claudia Mattissek
- Division of Cell Biology, Biocenter, Innsbruck Medical University
InnsbruckAustria
| | - David Teis
- Division of Cell Biology, Biocenter, Innsbruck Medical University
InnsbruckAustria
| |
Collapse
|
89
|
Abstract
A conventional view of development is that cells cooperate to build an organism. However, based on studies of Drosophila, it has been known for years that viable cells can be eliminated by their neighbours through a process termed cell competition. New studies in mammals have revealed that this process is universal and that many factors and mechanisms are conserved. During cell competition, cells with lower translation rates or those with lower levels of proteins involved in signal transduction, polarity and cellular growth can survive in a homogenous environment but are killed when surrounded by cells of higher fitness. Here, we discuss recent advances in the field as well as the mechanistic steps involved in this phenomenon, which have shed light on how and why cell competition exists in developing and adult organisms.
Collapse
Affiliation(s)
- Marc Amoyel
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, 550 First Avenue, MSB 497B, New York, NY 10016, USA
| | | |
Collapse
|
90
|
Dominguez M. Oncogenic programmes and Notch activity: an 'organized crime'? Semin Cell Dev Biol 2014; 28:78-85. [PMID: 24780858 DOI: 10.1016/j.semcdb.2014.04.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 04/02/2014] [Indexed: 10/25/2022]
Abstract
The inappropriate Notch signalling can influence virtually all aspect of cancer, including tumour-cell growth, survival, apoptosis, angiogenesis, invasion and metastasis, although it does not do this alone. Hence, elucidating the partners of Notch that are active in cancer is now the focus of much intense research activity. The genetic toolkits available, coupled to the small size and short life of the fruit fly Drosophila melanogaster, makes this an inexpensive and effective animal model, suited to large-scale cancer gene discovery studies. The fly eye is not only a non-vital organ but its stereotyped size and disposition also means it is easy to screen for mutations that cause tumours and metastases and provides ample opportunities to test cancer theories and to unravel unanticipated nexus between Notch and other cancer genes, or to discover unforeseen Notch's partners in cancer. These studies suggest that Notch's oncogenic capacity is brought about not simply by increasing signal strength but through partnerships, whereby oncogenes gain more by cooperating than acting individually, as in a ring 'organized crime'.
Collapse
|
91
|
Berns N, Woichansky I, Friedrichsen S, Kraft N, Riechmann V. A genome-scale in vivo RNAi analysis of epithelial development in Drosophila identifies new proliferation domains outside of the stem cell niche. J Cell Sci 2014; 127:2736-48. [PMID: 24762813 DOI: 10.1242/jcs.144519] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The Drosophila oogenesis system provides an excellent model to study the development of epithelial tissues. Here, we report the first genome-scale in vivo RNA interference (RNAi) screen for genes controlling epithelial development. By directly analysing cell and tissue architecture we identified 1125 genes, which we assigned to seven different functions in epithelial formation and homeostasis. We validated the significance of our screen by generating mutants for Vps60, a component of the endosomal sorting complexes required for transport (ESCRT) machinery. This analysis provided new insights into spatiotemporal control of cell proliferation in the follicular epithelium. Previous studies have identified signals controlling divisions in the follicle stem cell niche. However, 99% of cell divisions occur outside of the niche and it is unclear how these divisions are controlled. Our data distinguish two new domains outside of the stem cell niche where there are differing controls on proliferation. One domain abuts the niche and is characterised by ESCRT, Notch and JAK/STAT-mediated control of proliferation. Adjacent to this domain, another domain is defined by loss of the impact of ESCRT on cell division. Thus, during development epithelial cells pass through a variety of microenvironments that exert different modes of proliferation control. The switch between these modes might reflect a decrease in the 'stemness' of epithelial cells over time.
Collapse
Affiliation(s)
- Nicola Berns
- Heidelberg University, Medical Faculty Mannheim, Department of Cell and Molecular Biology and German Cancer Research Center (DKFZ), Division of Signaling and Functional Genomics, Ludolf-Krehl-Strasse 13-17, D-68167 Mannheim, Germany
| | - Innokenty Woichansky
- Heidelberg University, Medical Faculty Mannheim, Department of Cell and Molecular Biology and German Cancer Research Center (DKFZ), Division of Signaling and Functional Genomics, Ludolf-Krehl-Strasse 13-17, D-68167 Mannheim, Germany
| | - Steffen Friedrichsen
- Heidelberg University, Medical Faculty Mannheim, Department of Cell and Molecular Biology and German Cancer Research Center (DKFZ), Division of Signaling and Functional Genomics, Ludolf-Krehl-Strasse 13-17, D-68167 Mannheim, Germany
| | - Nadine Kraft
- Heidelberg University, Medical Faculty Mannheim, Department of Cell and Molecular Biology and German Cancer Research Center (DKFZ), Division of Signaling and Functional Genomics, Ludolf-Krehl-Strasse 13-17, D-68167 Mannheim, Germany
| | - Veit Riechmann
- Heidelberg University, Medical Faculty Mannheim, Department of Cell and Molecular Biology and German Cancer Research Center (DKFZ), Division of Signaling and Functional Genomics, Ludolf-Krehl-Strasse 13-17, D-68167 Mannheim, Germany
| |
Collapse
|
92
|
How to take autophagy and endocytosis up a notch. BIOMED RESEARCH INTERNATIONAL 2014; 2014:960803. [PMID: 24860831 PMCID: PMC4016896 DOI: 10.1155/2014/960803] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 03/12/2014] [Indexed: 11/23/2022]
Abstract
The interconnection of the endocytic and autophagosomal trafficking routes has been recognized more than two decades ago with both pathways using a set of identical effector proteins and sharing the same ultimate lysosomal destination. More recent data sheds light onto how other pathways are intertwined into this network, and how degradation via the endosomal/autophagosomal system may affect signaling pathways in multicellular organisms. Here, we briefly review the common features of autophagy and endocytosis and discuss how other players enter this mix with particular respect to the Notch signaling pathway.
Collapse
|
93
|
Abstract
Cancer was seen for a long time as a strictly cell-autonomous process in which oncogenes and tumor-suppressor mutations drive clonal cell expansions. Research in the past decade, however, paints a more integrative picture of communication and interplay between neighboring cells in tissues. It is increasingly clear as well that tumors, far from being homogenous lumps of cells, consist of different cell types that function together as complex tissue-level communities. The repertoire of interactive cell behaviors and the quantity of cellular players involved call for a social cell biology that investigates these interactions. Research into this social cell biology is critical for understanding development of normal and tumoral tissues. Such complex social cell biology interactions can be parsed in Drosophila. Techniques in Drosophila for analysis of gene function and clonal behavior allow us to generate tumors and dissect their complex interactive biology with cellular resolution. Here, we review recent Drosophila research aimed at understanding tissue-level biology and social cell interactions in tumors, highlighting the principles these studies reveal.
Collapse
|
94
|
ESCRT-0 is not required for ectopic Notch activation and tumor suppression in Drosophila. PLoS One 2014; 9:e93987. [PMID: 24718108 PMCID: PMC3981749 DOI: 10.1371/journal.pone.0093987] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 03/10/2014] [Indexed: 12/05/2022] Open
Abstract
Multivesicular endosome (MVE) sorting depends on proteins of the Endosomal Sorting Complex Required for Transport (ESCRT) family. These are organized in four complexes (ESCRT-0, -I, -II, -III) that act in a sequential fashion to deliver ubiquitylated cargoes into the internal luminal vesicles (ILVs) of the MVE. Drosophila genes encoding ESCRT-I, -II, -III components function in sorting signaling receptors, including Notch and the JAK/STAT signaling receptor Domeless. Loss of ESCRT-I, -II, -III in Drosophila epithelia causes altered signaling and cell polarity, suggesting that ESCRTs genes are tumor suppressors. However, the nature of the tumor suppressive function of ESCRTs, and whether tumor suppression is linked to receptor sorting is unclear. Unexpectedly, a null mutant in Hrs, encoding one of the components of the ESCRT-0 complex, which acts upstream of ESCRT-I, -II, -III in MVE sorting is dispensable for tumor suppression. Here, we report that two Drosophila epithelia lacking activity of Stam, the other known components of the ESCRT-0 complex, or of both Hrs and Stam, accumulate the signaling receptors Notch and Dome in endosomes. However, mutant tissue surprisingly maintains normal apico-basal polarity and proliferation control and does not display ectopic Notch signaling activation, unlike cells that lack ESCRT-I, -II, -III activity. Overall, our in vivo data confirm previous evidence indicating that the ESCRT-0 complex plays no crucial role in regulation of tumor suppression, and suggest re-evaluation of the relationship of signaling modulation in endosomes and tumorigenesis.
Collapse
|
95
|
Amoyel M, Anderson AM, Bach EA. JAK/STAT pathway dysregulation in tumors: a Drosophila perspective. Semin Cell Dev Biol 2014; 28:96-103. [PMID: 24685611 PMCID: PMC4037387 DOI: 10.1016/j.semcdb.2014.03.023] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Revised: 03/17/2014] [Accepted: 03/20/2014] [Indexed: 12/18/2022]
Abstract
Sustained activation of the JAK/STAT pathway is causal to human cancers. This pathway is less complex in Drosophila, and its dysregulation has been linked to several tumor models in this organism. Here, we discuss models of metastatic epithelial and hematopoietic tumors that are causally linked to dysregulation of JAK/STAT signaling in Drosophila. First, we focus on cancer models in imaginal discs where ectopic expression of the JAK/STAT pathway ligand Unpaired downstream of distinct tumor suppressors has emerged as an unexpected mediator of neoplastic transformation. We also discuss the collaboration between STAT and oncogenic Ras in epithelial transformation. Second, we examine hematopoietic tumors, where mutations that cause hyperactive JAK/STAT signaling are necessary and sufficient for "fly leukemia". We highlight the important contributions that genetic screens in Drosophila have made to understanding the JAK/STAT pathway, its developmental roles, and how its function is co-opted during tumorigenesis.
Collapse
Affiliation(s)
- Marc Amoyel
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, 550 First Avenue, MSB 497B, New York, NY 10016, USA
| | - Abigail M Anderson
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, 550 First Avenue, MSB 497B, New York, NY 10016, USA
| | - Erika A Bach
- The Helen L. and Martin S. Kimmel Center for Stem Cell Biology, New York University School of Medicine.
| |
Collapse
|
96
|
Leitch CC, Lodh S, Prieto-Echagüe V, Badano JL, Zaghloul NA. Basal body proteins regulate Notch signaling through endosomal trafficking. J Cell Sci 2014; 127:2407-19. [PMID: 24681783 DOI: 10.1242/jcs.130344] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Proteins associated with primary cilia and basal bodies mediate numerous signaling pathways, but little is known about their role in Notch signaling. Here, we report that loss of the Bardet-Biedl syndrome proteins BBS1 or BBS4 produces increased Notch-directed transcription in a zebrafish reporter line and in human cell lines. Pathway overactivation is accompanied by reduced localization of Notch receptor at both the plasma membrane and the cilium. In Drosophila mutants, overactivation of Notch can result from receptor accumulation in endosomes, and recent studies implicate ciliary proteins in endosomal trafficking, suggesting a possible mechanism by which overactivation occurs in BBS mutants. Consistent with this, we observe genetic interaction of BBS1 and BBS4 with the endosomal sorting complexes required for transport (ESCRT) gene TSG101 and accumulation of receptor in late endosomes, reduced endosomal recycling and reduced receptor degradation in lysosomes. We observe similar defects with disruption of BBS3. Loss of another basal body protein, ALMS1, also enhances Notch activation and the accumulation of receptor in late endosomes, but does not disrupt recycling. These findings suggest a role for these proteins in the regulation of Notch through endosomal trafficking of the receptor.
Collapse
Affiliation(s)
- Carmen C Leitch
- Department of Medicine, Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Sukanya Lodh
- Department of Medicine, Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | - Jose L Badano
- Institut Pasteur de Montevideo, CP11400 Montevideo, Uruguay
| | - Norann A Zaghloul
- Department of Medicine, Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
97
|
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
|
98
|
Ignesti M, Barraco M, Nallamothu G, Woolworth JA, Duchi S, Gargiulo G, Cavaliere V, Hsu T. Notch signaling during development requires the function of awd, the Drosophila homolog of human metastasis suppressor gene Nm23. BMC Biol 2014; 12:12. [PMID: 24528630 PMCID: PMC3937027 DOI: 10.1186/1741-7007-12-12] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 02/10/2014] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND The Drosophila abnormal wing discs (awd) belongs to a highly conserved family of genes implicated in metastasis suppression, metabolic homeostasis and epithelial morphogenesis. The cellular function of the mammalian members of this family, the Nm23 proteins, has not yet been clearly defined. Previous awd genetic analyses unraveled its endocytic role that is required for proper internalization of receptors controlling different signaling pathways. In this study, we analyzed the role of Awd in controlling Notch signaling during development. RESULTS To study the awd gene function we used genetic mosaic approaches to obtain cells homozygous for a loss of function allele. In awd mutant follicle cells and wing disc cells, Notch accumulates in enlarged early endosomes, resulting in defective Notch signaling. Our results demonstrate that awd function is required before γ-secretase mediated cleavage since over-expression of the constitutively active form of the Notch receptor in awd mutant follicle cells allows rescue of the signaling. By using markers of different endosomal compartments we show that Notch receptor accumulates in early endosomes in awd mutant follicle cells. A trafficking assay in living wing discs also shows that Notch accumulates in early endosomes. Importantly, constitutively active Rab5 cannot rescue the awd phenotype, suggesting that awd is required for Rab5 function in early endosome maturation. CONCLUSIONS In this report we demonstrate that awd is essential for Notch signaling via its endocytic role. In addition, we identify the endocytic step at which Awd function is required for Notch signaling and we obtain evidence indicating that Awd is necessary for Rab5 function. These findings provide new insights into the developmental and pathophysiological function of this important gene family.
Collapse
Affiliation(s)
- Marilena Ignesti
- Dipartimento di Farmacia e Biotecnologie, Alma Mater Studiorum Università di Bologna, Via Selmi, 3, Bologna 40126, Italy
| | - Marilena Barraco
- Dipartimento di Farmacia e Biotecnologie, Alma Mater Studiorum Università di Bologna, Via Selmi, 3, Bologna 40126, Italy
- Present address: Institute of Hematology “L. e A. Seràgnoli”, University of Bologna, Bologna, Italy
| | - Gouthami Nallamothu
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | - Julie A Woolworth
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | - Serena Duchi
- Dipartimento di Farmacia e Biotecnologie, Alma Mater Studiorum Università di Bologna, Via Selmi, 3, Bologna 40126, Italy
- Present address: Bone Regeneration Laboratory, Research Institute Codivilla-Putti, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Giuseppe Gargiulo
- Dipartimento di Farmacia e Biotecnologie, Alma Mater Studiorum Università di Bologna, Via Selmi, 3, Bologna 40126, Italy
| | - Valeria Cavaliere
- Dipartimento di Farmacia e Biotecnologie, Alma Mater Studiorum Università di Bologna, Via Selmi, 3, Bologna 40126, Italy
| | - Tien Hsu
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts 02118, USA
- Graduate Institute of Systems Biology and Bioinformatics, National Central University, Jhongli, Taiwan
| |
Collapse
|
99
|
Aoyama N, Yamakawa T, Sasamura T, Yoshida Y, Ohori M, Okubo H, Iida E, Sasaki N, Ueda R, Matsuno K. Loss- and gain-of-function analyses of vacuolar protein sorting 2 in Notch signaling of Drosophila melanogaster. Genes Genet Syst 2014; 88:45-57. [PMID: 23676709 DOI: 10.1266/ggs.88.45] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Notch signaling is an evolutionarily conserved mechanism that controls many cell-fate specifications through local cell-cell interactions. The core mechanisms of Notch activation and its subsequent intracellular signaling are well understood. Various cellular functions are required for the activation and regulation of Notch signaling. Among them, the endocytosis of Notch and its ligands is important for the activation and suppression of Notch signaling. The endosomal sorting complex required for transport (ESCRT) proteins are required to sort ubiquitinated membrane proteins, such as Notch, into early endosomes. A loss-of-function allele of vacuolar protein sorting 2 (vps2), which encodes a component of ESCRT-III, has been reported. However, this vps2 mutant still produces the N-terminal half of the protein, and its phenotypes were studied in only a few organs. Here, we generated the first null mutant allele of Drosophila vps2, designated vps2², to better understand the function of this gene. In Drosophila wing imaginal discs homozygous for the vps2² allele, early endosomes and multivesicular bodies (MVBs) were enlarged, and Notch and Delta accumulated inside them. As reported for the previous vps2 mutant, the epithelium grew excessively under this condition. We further studied the roles of vps2 by RNA interference-knockdown. These experiments revealed that a partial reduction of vps2 attenuated Notch signaling; in contrast, the loss-of-function vps2 mutant is reported to up-regulate the Notch signaling in eye imaginal disc cells. These results suggest that Notch signaling can be up- or down-regulated, depending on the level of vps2 expression. Finally, we found that vps2 overexpression also resulted in early-endosome enlargement and the accumulation of Notch and Delta. In these cells, a portion of the Vps2 protein was detected in MVBs and colocalized with Notch. These data indicate that the expression of vps2 must be precisely regulated to maintain the normal structure of early endosomes.
Collapse
Affiliation(s)
- Naoki Aoyama
- Department of Biological Science and Technology, Tokyo University of Science, 2641 Yamazaki,Noda, Chiba 278-8510, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Tipping M, Perrimon N. Drosophila as a model for context-dependent tumorigenesis. J Cell Physiol 2013; 229:27-33. [PMID: 23836429 DOI: 10.1002/jcp.24427] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 06/21/2013] [Indexed: 01/06/2023]
Abstract
Drosophila can exhibit classic hallmarks of cancer, such as evasion of apoptosis, sustained proliferation, metastasis, prolonged survival, genome instability, and metabolic reprogramming, when cancer-related genes are perturbed. In the last two decades, studies in flies have identified several tumor suppressor and oncogenes. However, the greatest strength of the fly lies in its ability to model cancer hallmarks in a variety of tissue types, which enables the study of context-dependent tumorigenesis. We review the organs and tissues that have been used to model tumor formation, and propose new strategies to maximize the potential of Drosophila in cancer research.
Collapse
Affiliation(s)
- Marla Tipping
- Department of Genetics, Harvard Medical School, Boston, Massachusetts
| | | |
Collapse
|