1
|
Martin-Blanco CA, Navarro P, Esteban-Collado J, Serra F, Almudi I, Casares F. Gill regeneration in the mayfly Cloeon uncovers new molecular pathways in insect regeneration. Open Biol 2024; 14:240118. [PMID: 39591992 PMCID: PMC11597413 DOI: 10.1098/rsob.240118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 09/23/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024] Open
Abstract
The capacity to regenerate lost organs is widespread among animals, and yet the number of species in which regeneration has been experimentally probed using molecular and functional assays is very small. This is also the case for insects, for which we still lack a complete picture of their regeneration mechanisms and the extent of their conservation. Here, we contribute to filling this gap by investigating regeneration in the mayfly Cloeon dipterum. We focus on the abdominal gills of Cloeon nymphs, which are critical for osmoregulation and gas exchange. After amputation, gills re-grow faster than they do during normal development. Direct cell count and EdU assays indicate that growth acceleration involves an uniform increase in cell proliferation throughout the gill, rather than a localized growth zone. Accordingly, transcriptomic analysis reveals an early enrichment in cell cycle-related genes. Other gene classes are also enriched in regenerating gills, including protein neddylation and other proteostatic processes. We then showed the conservation of these mechanisms by functionally testing protein neddylation, the activin signalling pathway or the mRNA-binding protein Lin28, among other genes, in Drosophila larval/pupal wing regeneration. Globally, our results contribute to elucidating regeneration mechanisms in mayflies and the conservation of mechanisms involved in regeneration across insects.
Collapse
Affiliation(s)
- Carlos A. Martin-Blanco
- CABD (Andalusian Center for Developmental Biology), CSIC/Universidad Pablo de Olavide/Junta de Andalucía, Seville41013, Spain
- Department of Genetics, Microbiology and Statistics, Universitat de Barcelona, Diagonal 643, 08028, Barcelona, Spain
| | - Pablo Navarro
- CABD (Andalusian Center for Developmental Biology), CSIC/Universidad Pablo de Olavide/Junta de Andalucía, Seville41013, Spain
| | - José Esteban-Collado
- Department of Genetics, Microbiology and Statistics, Universitat de Barcelona, Diagonal 643, 08028, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), Diagonal 643, 08028, Barcelona, Spain
| | - Florenci Serra
- Department of Genetics, Microbiology and Statistics, Universitat de Barcelona, Diagonal 643, 08028, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), Diagonal 643, 08028, Barcelona, Spain
| | - Isabel Almudi
- Department of Genetics, Microbiology and Statistics, Universitat de Barcelona, Diagonal 643, 08028, Barcelona, Spain
- Institut de Recerca de la Biodiversitat (IRBio), Universitat de Barcelona, Diagonal, 643, 08028, Barcelona, Spain
| | - Fernando Casares
- CABD (Andalusian Center for Developmental Biology), CSIC/Universidad Pablo de Olavide/Junta de Andalucía, Seville41013, Spain
| |
Collapse
|
2
|
Guo Y, Wang M, Zhu J, Li Q, Liu H, Wang Y, Hou SX. Long noncoding RNAs heat shock RNA omega nucleates TBPH and promotes intestinal stem cell differentiation upon heat shock. iScience 2024; 27:109732. [PMID: 38706862 PMCID: PMC11067334 DOI: 10.1016/j.isci.2024.109732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 03/31/2024] [Accepted: 04/09/2024] [Indexed: 05/07/2024] Open
Abstract
In Drosophila, long noncoding RNA Hsrω rapidly assembles membraneless organelle omega speckles under heat shock with unknown biological function. Here, we identified the distribution of omega speckles in multiple tissues of adult Drosophila melanogaster and found that they were selectively distributed in differentiated enterocytes but not in the intestinal stem cells of the midgut. We mimicked the high expression level of Hsrω via overexpression or intense heat shock and demonstrated that the assembly of omega speckles nucleates TBPH for the induction of ISC differentiation. Additionally, we found that heat shock stress promoted cell differentiation, which is conserved in mammalian cells through paraspeckles, resulting in large puncta of TDP-43 (a homolog of TBPH) with less mobility and the differentiation of human induced pluripotent stem cells. Overall, our findings confirm the role of Hsrω and omega speckles in the development of intestinal cells and provide new prospects for the establishment of stem cell differentiation strategies.
Collapse
Affiliation(s)
- Yinfeng Guo
- State Key Laboratory of Genetic Engineering, Department of Cell and Developmental Biology at School of Life Sciences, Institute of Metabolism and Integrative Biology, Human Phenome Institute, Department of Liver Surgery and Transplantation of Liver Cancer Institute at Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Meng Wang
- State Key Laboratory of Genetic Engineering, Department of Cell and Developmental Biology at School of Life Sciences, Institute of Metabolism and Integrative Biology, Human Phenome Institute, Department of Liver Surgery and Transplantation of Liver Cancer Institute at Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Jiaxin Zhu
- State Key Laboratory of Genetic Engineering, Department of Cell and Developmental Biology at School of Life Sciences, Institute of Metabolism and Integrative Biology, Human Phenome Institute, Department of Liver Surgery and Transplantation of Liver Cancer Institute at Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Qiaoming Li
- State Key Laboratory of Genetic Engineering, Department of Cell and Developmental Biology at School of Life Sciences, Institute of Metabolism and Integrative Biology, Human Phenome Institute, Department of Liver Surgery and Transplantation of Liver Cancer Institute at Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Haitao Liu
- State Key Laboratory of Genetic Engineering, Department of Cell and Developmental Biology at School of Life Sciences, Institute of Metabolism and Integrative Biology, Human Phenome Institute, Department of Liver Surgery and Transplantation of Liver Cancer Institute at Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Yang Wang
- State Key Laboratory of Genetic Engineering, Department of Cell and Developmental Biology at School of Life Sciences, Institute of Metabolism and Integrative Biology, Human Phenome Institute, Department of Liver Surgery and Transplantation of Liver Cancer Institute at Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Steven X. Hou
- State Key Laboratory of Genetic Engineering, Department of Cell and Developmental Biology at School of Life Sciences, Institute of Metabolism and Integrative Biology, Human Phenome Institute, Department of Liver Surgery and Transplantation of Liver Cancer Institute at Zhongshan Hospital, Fudan University, Shanghai 200438, China
| |
Collapse
|
3
|
Khanbabei A, Segura L, Petrossian C, Lemus A, Cano I, Frazier C, Halajyan A, Ca D, Loza-Coll M. Experimental validation and characterization of putative targets of Escargot and STAT, two master regulators of the intestinal stem cells in Drosophila melanogaster. Dev Biol 2024; 505:148-163. [PMID: 37952851 DOI: 10.1016/j.ydbio.2023.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 10/15/2023] [Accepted: 10/25/2023] [Indexed: 11/14/2023]
Abstract
Many organs contain adult stem cells (ASCs) to replace cells due to damage, disease, or normal tissue turnover. ASCs can divide asymmetrically, giving rise to a new copy of themselves (self-renewal) and a sister that commits to a specific cell type (differentiation). Decades of research have led to the identification of pleiotropic genes whose loss or gain of function affect diverse aspects of normal ASC biology. Genome-wide screens of these so-called genetic "master regulator" (MR) genes, have pointed to hundreds of putative targets that could serve as their downstream effectors. Here, we experimentally validate and characterize the regulation of several putative targets of Escargot (Esg) and the Signal Transducer and Activator of Transcription (Stat92E, a.k.a. STAT), two known MRs in Drosophila intestinal stem cells (ISCs). Our results indicate that regardless of bioinformatic predictions, most experimentally validated targets show a profile of gene expression that is consistent with co-regulation by both Esg and STAT, fitting a rather limited set of co-regulatory modalities. A bioinformatic analysis of proximal regulatory sequences in specific subsets of co-regulated targets identified additional transcription factors that might cooperate with Esg and STAT in modulating their transcription. Lastly, in vivo manipulations of validated targets rarely phenocopied the effects of manipulating Esg and STAT, suggesting the existence of complex genetic interactions among downstream targets of these two MR genes during ISC homeostasis.
Collapse
Affiliation(s)
- Armen Khanbabei
- Department of Biology, California State University, Northridge (CSUN), USA
| | - Lina Segura
- Department of Biology, California State University, Northridge (CSUN), USA
| | - Cynthia Petrossian
- Department of Biology, California State University, Northridge (CSUN), USA
| | - Aaron Lemus
- Department of Biology, California State University, Northridge (CSUN), USA
| | - Ithan Cano
- Department of Biology, California State University, Northridge (CSUN), USA
| | - Courtney Frazier
- Department of Biology, California State University, Northridge (CSUN), USA
| | - Armen Halajyan
- Department of Biology, California State University, Northridge (CSUN), USA
| | - Donnie Ca
- Department of Biology, California State University, Northridge (CSUN), USA
| | - Mariano Loza-Coll
- Department of Biology, California State University, Northridge (CSUN), USA.
| |
Collapse
|
4
|
Nagai H, Nagai LAE, Tasaki S, Nakato R, Umetsu D, Kuranaga E, Miura M, Nakajima Y. Nutrient-driven dedifferentiation of enteroendocrine cells promotes adaptive intestinal growth in Drosophila. Dev Cell 2023; 58:1764-1781.e10. [PMID: 37689060 DOI: 10.1016/j.devcel.2023.08.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 05/05/2023] [Accepted: 08/16/2023] [Indexed: 09/11/2023]
Abstract
Post-developmental organ resizing improves organismal fitness under constantly changing nutrient environments. Although stem cell abundance is a fundamental determinant of adaptive resizing, our understanding of its underlying mechanisms remains primarily limited to the regulation of stem cell division. Here, we demonstrate that nutrient fluctuation induces dedifferentiation in the Drosophila adult midgut to drive adaptive intestinal growth. From lineage tracing and single-cell RNA sequencing, we identify a subpopulation of enteroendocrine (EE) cells that convert into functional intestinal stem cells (ISCs) in response to dietary glucose and amino acids by activating the JAK-STAT pathway. Genetic ablation of EE-derived ISCs severely impairs ISC expansion and midgut growth despite the retention of resident ISCs, and in silico modeling further indicates that EE dedifferentiation enables an efficient increase in the midgut cell number while maintaining epithelial cell composition. Our findings identify a physiologically induced dedifferentiation that ensures ISC expansion during adaptive organ growth in concert with nutrient conditions.
Collapse
Affiliation(s)
- Hiroki Nagai
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan; Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai, Miyagi 980-0845, Japan.
| | | | - Sohei Tasaki
- Graduate School of Science, Hokkaido University, Sapporo, Hokkaido 060-0810, Japan
| | - Ryuichiro Nakato
- Institute for Quantitative Biosciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Daiki Umetsu
- Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-0845, Japan; Graduate School of Science, Osaka University, Osaka 560-0043, Japan
| | - Erina Kuranaga
- Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-0845, Japan
| | - Masayuki Miura
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Yuichiro Nakajima
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan; Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai, Miyagi 980-0845, Japan; Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-0845, Japan.
| |
Collapse
|
5
|
Zhai J, Li W, Liu X, Wang D, Zhang D, Liu Y, Liang X, Chen Z. Tiny Drosophila intestinal stem cells, big power. Cell Biol Int 2022; 47:3-14. [PMID: 36177490 DOI: 10.1002/cbin.11911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 09/12/2022] [Accepted: 09/12/2022] [Indexed: 11/12/2022]
Abstract
The signaling pathways are highly conserved between Drosophila and mammals concerning intestinal development, regeneration, and disease. The powerful genetic tools of Drosophila make it a valuable and convenient alternative to answer basic biological questions that can not be addressed using mammalian models. In this review, we discuss recent advances in how we use fly midgut to answer the following key questions: (1) How intestine stem cell niches are established; (2) which factors control asymmetric division of stem cells; (3) how intestinal cells interact with environmental factors, such as tissue damage, microbiota, and diet; (4) how to screen aging/cancer-related factors or drugs by fly intestine stem cells.
Collapse
Affiliation(s)
- Jingbo Zhai
- Medical College, Inner Mongolia Minzu University, Tongliao, China.,Key Laboratory of Zoonose Prevention and Control at Universities of Inner Mongolia Autonomous Region, Tongliao, China.,Brucellosis Prevention and Treatment Engineering Research Center of Inner Mongolia Autonomous Region, Tongliao, China
| | - Wanyang Li
- Medical College, Inner Mongolia Minzu University, Tongliao, China
| | - Xin Liu
- Medical College, Inner Mongolia Minzu University, Tongliao, China.,Key Laboratory of Zoonose Prevention and Control at Universities of Inner Mongolia Autonomous Region, Tongliao, China.,Brucellosis Prevention and Treatment Engineering Research Center of Inner Mongolia Autonomous Region, Tongliao, China
| | - Di Wang
- Medical College, Inner Mongolia Minzu University, Tongliao, China.,Key Laboratory of Zoonose Prevention and Control at Universities of Inner Mongolia Autonomous Region, Tongliao, China.,Brucellosis Prevention and Treatment Engineering Research Center of Inner Mongolia Autonomous Region, Tongliao, China
| | - Dongli Zhang
- Medical College, Inner Mongolia Minzu University, Tongliao, China.,Key Laboratory of Zoonose Prevention and Control at Universities of Inner Mongolia Autonomous Region, Tongliao, China.,Brucellosis Prevention and Treatment Engineering Research Center of Inner Mongolia Autonomous Region, Tongliao, China
| | - Yanli Liu
- Affiliated Hospital of Inner Mongolia Minzu University, Tongliao, China
| | - Xiuwen Liang
- Hulunbuir City People's Hospital, Hulunbuir City, China
| | - Zeliang Chen
- Medical College, Inner Mongolia Minzu University, Tongliao, China.,Key Laboratory of Zoonose Prevention and Control at Universities of Inner Mongolia Autonomous Region, Tongliao, China.,Brucellosis Prevention and Treatment Engineering Research Center of Inner Mongolia Autonomous Region, Tongliao, China
| |
Collapse
|
6
|
Imp interacts with Lin28 to regulate adult stem cell proliferation in the Drosophila intestine. PLoS Genet 2022; 18:e1010385. [PMID: 36070313 PMCID: PMC9484684 DOI: 10.1371/journal.pgen.1010385] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 09/19/2022] [Accepted: 08/18/2022] [Indexed: 11/30/2022] Open
Abstract
Stem cells are essential for the development and long-term maintenance of tissues and organisms. Preserving tissue homeostasis requires exquisite control of all aspects of stem cell function: cell potency, proliferation, fate decision and differentiation. RNA binding proteins (RBPs) are essential components of the regulatory network that control gene expression in stem cells to maintain self-renewal and long-term homeostasis in adult tissues. While the function of many RBPs may have been characterized in various stem cell populations, how these interact and are organized in genetic networks remains largely elusive. In this report, we show that the conserved RNA binding protein IGF2 mRNA binding protein (Imp) is expressed in intestinal stem cells (ISCs) and progenitors in the adult Drosophila midgut. We demonstrate that Imp is required cell autonomously to maintain stem cell proliferative activity under normal epithelial turnover and in response to tissue damage. Mechanistically, we show that Imp cooperates and directly interacts with Lin28, another highly conserved RBP, to regulate ISC proliferation. We found that both proteins bind to and control the InR mRNA, a critical regulator of ISC self-renewal. Altogether, our data suggests that Imp and Lin28 are part of a larger gene regulatory network controlling gene expression in ISCs and required to maintain epithelial homeostasis. Stem cells are essential to maintain healthy organs. However, dysregulation of their function is a potential major driver of diseases, including cancer and neurodegeneration, and significantly contributes to the aging process. For these reasons, numerous mechanisms control the ability of stem cells to divide and give rise to functional daughter cells. In this study, we used the Drosophila fruitfly as a genetically amenable experimental model to characterize the function of a conserved protein, the IGF2 mRNA binding protein, in the regulation of adult intestinal stem cells. We found that it is essential for stem cell proliferation under normal conditions and in response to tissue damage. We also report that it interacts with another known regulator, Lin28. Importantly, these two factors largely control stem cell biology and development in mammals, including humans, and are often dysregulated in cancer. This suggests that our work is shedding new light on the conserved mechanisms that maintain long-term stem cell function across organisms.
Collapse
|
7
|
Dillon N, Cocanougher B, Sood C, Yuan X, Kohn AB, Moroz LL, Siegrist SE, Zlatic M, Doe CQ. Single cell RNA-seq analysis reveals temporally-regulated and quiescence-regulated gene expression in Drosophila larval neuroblasts. Neural Dev 2022; 17:7. [PMID: 36002894 PMCID: PMC9404614 DOI: 10.1186/s13064-022-00163-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/19/2022] [Indexed: 12/12/2022] Open
Abstract
The mechanisms that generate neural diversity during development remains largely unknown. Here, we use scRNA-seq methodology to discover new features of the Drosophila larval CNS across several key developmental timepoints. We identify multiple progenitor subtypes - both stem cell-like neuroblasts and intermediate progenitors - that change gene expression across larval development, and report on new candidate markers for each class of progenitors. We identify a pool of quiescent neuroblasts in newly hatched larvae and show that they are transcriptionally primed to respond to the insulin signaling pathway to exit from quiescence, including relevant pathway components in the adjacent glial signaling cell type. We identify candidate "temporal transcription factors" (TTFs) that are expressed at different times in progenitor lineages. Our work identifies many cell type specific genes that are candidates for functional roles, and generates new insight into the differentiation trajectory of larval neurons.
Collapse
Affiliation(s)
- Noah Dillon
- Institute of Neuroscience, Howard Hughes Medical Institute, University of Oregon, OR, 97403, Eugene, USA
| | - Ben Cocanougher
- Department of Zoology, University of Cambridge, Cambridge, UK
| | - Chhavi Sood
- Department of Biology, University of Virginia, VA, 22904, Charlottesville, USA
| | - Xin Yuan
- Department of Biology, University of Virginia, VA, 22904, Charlottesville, USA
| | - Andrea B Kohn
- Whitney Laboratory for Marine Biosciences, University of Florida, FL, 32080, St. Augustine, USA
| | - Leonid L Moroz
- Whitney Laboratory for Marine Biosciences, University of Florida, FL, 32080, St. Augustine, USA
| | - Sarah E Siegrist
- Department of Biology, University of Virginia, VA, 22904, Charlottesville, USA
| | - Marta Zlatic
- MRC Laboratory of Molecular Biology, Dept of Zoology, University of Cambridge, Cambridge, UK
- Janelia Research Campus, VA, Ashburn, USA
| | - Chris Q Doe
- Institute of Neuroscience, Howard Hughes Medical Institute, University of Oregon, OR, 97403, Eugene, USA.
| |
Collapse
|
8
|
Ariyapala IS, Buddika K, Hundley HA, Calvi BR, Sokol NS. The RNA binding protein Swm is critical for Drosophila melanogaster intestinal progenitor cell maintenance. Genetics 2022; 222:6619166. [PMID: 35762963 DOI: 10.1093/genetics/iyac099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 05/09/2022] [Indexed: 11/13/2022] Open
Abstract
The regulation of stem cell survival, self-renewal, and differentiation is critical for the maintenance of tissue homeostasis. Although the involvement of signaling pathways and transcriptional control mechanisms in stem cell regulation have been extensively investigated, the role of post-transcriptional control is still poorly understood. Here we show that the nuclear activity of the RNA-binding protein Second Mitotic Wave Missing (Swm) is critical for Drosophila melanogaster intestinal stem cells (ISCs) and their daughter cells, enteroblasts (EBs), to maintain their progenitor cell properties and functions. Loss of swm causes ISCs and EBs to stop dividing and instead detach from the basement membrane, resulting in severe progenitor cell loss. swm loss is further characterized by nuclear accumulation of poly(A)+ RNA in progenitor cells. Swm associates with transcripts involved in epithelial cell maintenance and adhesion, and the loss of swm, while not generally affecting the levels of these Swm-bound mRNAs, leads to elevated expression of proteins encoded by some of them, including the fly ortholog of Filamin. Taken together, this study indicates a nuclear role for Swm in adult stem cell maintenance, raising the possibility that nuclear post-transcriptional regulation of mRNAs encoding cell adhesion proteins ensures proper attachment of progenitor cells.
Collapse
Affiliation(s)
| | - Kasun Buddika
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Heather A Hundley
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN 47405, USA
| | - Brian R Calvi
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Nicholas S Sokol
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| |
Collapse
|
9
|
Emerging Roles of RNA-Binding Proteins in Neurodevelopment. J Dev Biol 2022; 10:jdb10020023. [PMID: 35735914 PMCID: PMC9224834 DOI: 10.3390/jdb10020023] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/02/2022] [Accepted: 06/08/2022] [Indexed: 02/06/2023] Open
Abstract
Diverse cell types in the central nervous system (CNS) are generated by a relatively small pool of neural stem cells during early development. Spatial and temporal regulation of stem cell behavior relies on precise coordination of gene expression. Well-studied mechanisms include hormone signaling, transcription factor activity, and chromatin remodeling processes. Much less is known about downstream RNA-dependent mechanisms including posttranscriptional regulation, nuclear export, alternative splicing, and transcript stability. These important functions are carried out by RNA-binding proteins (RBPs). Recent work has begun to explore how RBPs contribute to stem cell function and homeostasis, including their role in metabolism, transport, epigenetic regulation, and turnover of target transcripts. Additional layers of complexity are provided by the different target recognition mechanisms of each RBP as well as the posttranslational modifications of the RBPs themselves that alter function. Altogether, these functions allow RBPs to influence various aspects of RNA metabolism to regulate numerous cellular processes. Here we compile advances in RNA biology that have added to our still limited understanding of the role of RBPs in neurodevelopment.
Collapse
|
10
|
Abstract
Adult tissues in Metazoa dynamically remodel their structures in response to environmental challenges including sudden injury, pathogen infection, and nutritional fluctuation, while maintaining quiescence under homoeostatic conditions. This characteristic, hereafter referred to as adult tissue plasticity, can prevent tissue dysfunction and improve the fitness of organisms in continuous and/or severe change of environments. With its relatively simple tissue structures and genetic tools, studies using the fruit fly Drosophila melanogaster have provided insights into molecular mechanisms that control cellular responses, particularly during regeneration and nutrient adaptation. In this review, we present the current understanding of cellular mechanisms, stem cell proliferation, polyploidization, and cell fate plasticity, all of which enable adult tissue plasticity in various Drosophila adult organs including the midgut, the brain, and the gonad, and discuss the organismal strategy in response to environmental changes and future directions of the research.
Collapse
Affiliation(s)
- Hiroki Nagai
- Graduate School of Pharmaceutical Sciences, the University of Tokyo, Tokyo, Japan
| | - Masayuki Miura
- Graduate School of Pharmaceutical Sciences, the University of Tokyo, Tokyo, Japan
| | - Yu-Ichiro Nakajima
- Graduate School of Pharmaceutical Sciences, the University of Tokyo, Tokyo, Japan
| |
Collapse
|
11
|
Buddika K, Huang YT, Ariyapala IS, Butrum-Griffith A, Norrell SA, O'Connor AM, Patel VK, Rector SA, Slovan M, Sokolowski M, Kato Y, Nakamura A, Sokol NS. Coordinated repression of pro-differentiation genes via P-bodies and transcription maintains Drosophila intestinal stem cell identity. Curr Biol 2022; 32:386-397.e6. [PMID: 34875230 PMCID: PMC8792327 DOI: 10.1016/j.cub.2021.11.032] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 09/17/2021] [Accepted: 11/11/2021] [Indexed: 01/26/2023]
Abstract
The role of processing bodies (P-bodies), key sites of post-transcriptional control, in adult stem cells remains poorly understood. Here, we report that adult Drosophila intestinal stem cells, but not surrounding differentiated cells such as absorptive enterocytes (ECs), harbor P-bodies that contain Drosophila orthologs of mammalian P-body components DDX6, EDC3, EDC4, and LSM14A/B. A targeted RNAi screen in intestinal progenitor cells identified 39 previously known and 64 novel P-body regulators, including Patr-1, a gene necessary for P-body assembly. Loss of Patr-1-dependent P-bodies leads to a loss of stem cells that is associated with inappropriate expression of EC-fate gene nubbin. Transcriptomic analysis of progenitor cells identifies a cadre of such weakly transcribed pro-differentiation transcripts that are elevated after P-body loss. Altogether, this study identifies a P-body-dependent repression activity that coordinates with known transcriptional repression programs to maintain a population of in vivo stem cells in a state primed for differentiation.
Collapse
Affiliation(s)
- Kasun Buddika
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Yi-Ting Huang
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | | | | | - Sam A Norrell
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Alex M O'Connor
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Viraj K Patel
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Samuel A Rector
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Mark Slovan
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | | | - Yasuko Kato
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | - Akira Nakamura
- Department of Germline Development, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Kumamoto 860-0811, Japan
| | - Nicholas S Sokol
- Department of Biology, Indiana University, Bloomington, IN 47405, USA.
| |
Collapse
|
12
|
Wang J, Liu Q, Gong Y, Jin LH. Anchor maintains gut homeostasis by restricting the JNK and Notch pathways in Drosophila. JOURNAL OF INSECT PHYSIOLOGY 2021; 134:104309. [PMID: 34496279 DOI: 10.1016/j.jinsphys.2021.104309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 09/01/2021] [Accepted: 09/02/2021] [Indexed: 06/13/2023]
Abstract
The adult Drosophila intestinal epithelium must be tightly regulated to maintain regeneration and homeostasis. The dysregulation of the regenerative capacity is frequently associated with intestinal diseases such as inflammation and tumorigenesis. Here, we showed that the G protein-coupled receptor Anchor maintains Drosophila adult midgut homeostasis by restricting Jun-N-terminal kinase (JNK) and Notch pathway activity. anchor inactivation resulted in aberrant JNK pathway activation, which led to excessive enteroblast (EB) production and premature enterocyte (EC) differentiation. In addition, increased Notch levels promoted premature EC differentiation following the loss of anchor. This defect induced by the loss of anchor ultimately caused sensitivity to stress or environmental challenge in adult flies. Taken together, our results demonstrate that the activity of anchor is essential to coordinate stem cell differentiation and proliferation to maintain intestinal homeostasis.
Collapse
Affiliation(s)
- Jiewei Wang
- Department of Genetics, College of Life Sciences, Northeast Forestry University, No. 26 Hexing Road, Xiangfang District, Harbin 150040, China
| | - Qiang Liu
- Department of Cell Biology and Genetics, Shenyang Medical College, 146 Huanghe North Street, Shenyang 110034, China
| | - Yuan Gong
- Department of Genetics, College of Life Sciences, Northeast Forestry University, No. 26 Hexing Road, Xiangfang District, Harbin 150040, China
| | - Li Hua Jin
- Department of Genetics, College of Life Sciences, Northeast Forestry University, No. 26 Hexing Road, Xiangfang District, Harbin 150040, China.
| |
Collapse
|
13
|
Mukherjee S, Paricio N, Sokol NS. A stress-responsive miRNA regulates BMP signaling to maintain tissue homeostasis. Proc Natl Acad Sci U S A 2021; 118:e2022583118. [PMID: 34016750 PMCID: PMC8166057 DOI: 10.1073/pnas.2022583118] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Adult organisms must sense and adapt to environmental fluctuations. In high-turnover tissues such as the intestine, these adaptive responses require rapid changes in gene expression that, in turn, likely involve posttranscriptional gene control. However, intestinal-tissue-specific microRNA (miRNA)-mediated regulatory pathways remain unexplored. Here, we report the role of an intestinal-specific miRNA, miR-958, that non-cell autonomously regulates stem cell numbers during tissue homeostasis and regeneration in the Drosophila adult midgut. We identify its downstream target cabut, the Drosophila ortholog of mammalian KLF10/11 transcription factors, which mediates this miR-958 function by promoting paracrine enterocyte-to-stem-cell bone morphogenetic protein (BMP) signaling. We also show that mature miR-958 levels transiently decrease in response to stress and that this decrease is required for proper stem cell expansion during tissue regeneration. In summary, we have identified a posttranscriptional mechanism that modulates BMP signaling activity within Drosophila adult intestinal tissue during both normal homeostasis and tissue regeneration to regulate intestinal stem cell numbers.
Collapse
Affiliation(s)
| | - Nuria Paricio
- Departamento de Genética, Facultad de Ciencies Biológicas and Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina, University of Valencia, 46100 Burjasot, Spain
| | - Nicholas S Sokol
- Department of Biology, Indiana University, Bloomington, IN 47405;
| |
Collapse
|
14
|
Takemura M, Bowden N, Lu YS, Nakato E, O'Connor MB, Nakato H. Drosophila MOV10 regulates the termination of midgut regeneration. Genetics 2021; 218:6156853. [PMID: 33693718 DOI: 10.1093/genetics/iyab031] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 02/23/2021] [Indexed: 12/22/2022] Open
Abstract
The molecular mechanisms by which stem cell proliferation is precisely controlled during the course of regeneration are poorly understood. Namely, how a damaged tissue senses when to terminate the regeneration process, inactivates stem cell mitotic activity, and organizes ECM integrity remain fundamental unanswered questions. The Drosophila midgut intestinal stem cell (ISC) offers an excellent model system to study the molecular basis for stem cell inactivation. Here, we show that a novel gene, CG6967 or dMOV10, is induced at the termination stage of midgut regeneration, and shows an inhibitory effect on ISC proliferation. dMOV10 encodes a putative component of the microRNA (miRNA) gene silencing complex (miRISC). Our data, along with previous studies on the mammalian MOV10, suggest that dMOV10 is not a core member of miRISC, but modulates miRISC activity as an additional component. Further analyses identified direct target mRNAs of dMOV10-containing miRISC, including Daughter against Dpp (Dad), a known inhibitor of BMP/TGF-β signaling. We show that RNAi knockdown of Dad significantly impaired ISC division during regeneration. We also identified six miRNAs that are induced at the termination stage and their potential target transcripts. One of these miRNAs, mir-1, is required for proper termination of ISC division at the end of regeneration. We propose that miRNA-mediated gene regulation contributes to the precise control of Drosophila midgut regeneration.
Collapse
Affiliation(s)
- Masahiko Takemura
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Nanako Bowden
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Yi-Si Lu
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Eriko Nakato
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Michael B O'Connor
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Hiroshi Nakato
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
15
|
Buddika K, Xu J, Ariyapala IS, Sokol NS. I-KCKT allows dissection-free RNA profiling of adult Drosophila intestinal progenitor cells. Development 2021; 148:dev196568. [PMID: 33246929 PMCID: PMC7803463 DOI: 10.1242/dev.196568] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/19/2021] [Indexed: 12/13/2022]
Abstract
The adult Drosophila intestinal epithelium is a model system for stem cell biology, but its utility is limited by current biochemical methods that lack cell type resolution. Here, we describe a new proximity-based profiling method that relies upon a GAL4 driver, termed intestinal-kickout-GAL4 (I-KCKT-GAL4), that is exclusively expressed in intestinal progenitor cells. This method uses UV crosslinked whole animal frozen powder as its starting material to immunoprecipitate the RNA cargoes of transgenic epitope-tagged RNA binding proteins driven by I-KCKT-GAL4 When applied to the general mRNA-binder, poly(A)-binding protein, the RNA profile obtained by this method identifies 98.8% of transcripts found after progenitor cell sorting, and has low background noise despite being derived from whole animal lysate. We also mapped the targets of the more selective RNA binder, Fragile X mental retardation protein (FMRP), using enhanced crosslinking and immunoprecipitation (eCLIP), and report for the first time its binding motif in Drosophila cells. This method will therefore enable the RNA profiling of wild-type and mutant intestinal progenitor cells from intact flies exposed to normal and altered environments, as well as the identification of RNA-protein interactions crucial for stem cell function.
Collapse
Affiliation(s)
- Kasun Buddika
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Jingjing Xu
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | | | - Nicholas S Sokol
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| |
Collapse
|
16
|
Izumi H, Kaneko Y, Nakagawara A. The Role of MYCN in Symmetric vs. Asymmetric Cell Division of Human Neuroblastoma Cells. Front Oncol 2020; 10:570815. [PMID: 33194665 PMCID: PMC7609879 DOI: 10.3389/fonc.2020.570815] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 09/21/2020] [Indexed: 12/13/2022] Open
Abstract
Asymmetric cell division (ACD) is an important physiological event in the development of various organisms and maintenance of tissue homeostasis. ACD produces two different cells in a single cell division: a stem/progenitor cell and differentiated cell. Although the balance between self-renewal and differentiation is precisely controlled, disruptions to ACD and/or enhancements in the self-renewal division (symmetric cell division: SCD) of stem cells resulted in the formation of tumors in Drosophila neuroblasts. ACD is now regarded as one of the characteristics of human cancer stem cells, and is a driving force for cancer cell heterogeneity. We recently reported that MYCN controls the balance between SCD and ACD in human neuroblastoma cells. In this mini-review, we discuss the mechanisms underlying MYCN-mediated cell division fate.
Collapse
Affiliation(s)
- Hideki Izumi
- Laboratory of Molecular Medicine, Life Sciences Institute, Saga-Ken Medical Centre Koseikan, Saga, Japan
| | - Yasuhiko Kaneko
- Research Institute for Clinical Oncology, Saitama Cancer Center, Saitama, Japan
| | | |
Collapse
|
17
|
Joly A, Rousset R. Tissue Adaptation to Environmental Cues by Symmetric and Asymmetric Division Modes of Intestinal Stem Cells. Int J Mol Sci 2020; 21:ijms21176362. [PMID: 32887329 PMCID: PMC7504256 DOI: 10.3390/ijms21176362] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 08/28/2020] [Accepted: 08/30/2020] [Indexed: 12/20/2022] Open
Abstract
Tissues must adapt to the different external stimuli so that organisms can survive in their environments. The intestine is a vital organ involved in food processing and absorption, as well as in innate immune response. Its adaptation to environmental cues such as diet and biotic/abiotic stress involves regulation of the proliferative rate and a switch of division mode (asymmetric versus symmetric) of intestinal stem cells (ISC). In this review, we outline the current comprehension of the physiological and molecular mechanisms implicated in stem cell division modes in the adult Drosophila midgut. We present the signaling pathways and polarity cues that control the mitotic spindle orientation, which is the terminal determinant ensuring execution of the division mode. We review these events during gut homeostasis, as well as during its response to nutrient availability, bacterial infection, chemical damage, and aging. JNK signaling acts as a central player, being involved in each of these conditions as a direct regulator of spindle orientation. The studies of the mechanisms regulating ISC divisions allow a better understanding of how adult stem cells integrate different signals to control tissue plasticity, and of how various diseases, notably cancers, arise from their alterations.
Collapse
|
18
|
Funk MC, Zhou J, Boutros M. Ageing, metabolism and the intestine. EMBO Rep 2020; 21:e50047. [PMID: 32567155 PMCID: PMC7332987 DOI: 10.15252/embr.202050047] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 04/18/2020] [Accepted: 05/29/2020] [Indexed: 12/14/2022] Open
Abstract
The intestinal epithelium serves as a dynamic barrier to the environment and integrates a variety of signals, including those from metabolites, commensal microbiota, immune responses and stressors upon ageing. The intestine is constantly challenged and requires a high renewal rate to replace damaged cells in order to maintain its barrier function. Essential for its renewal capacity are intestinal stem cells, which constantly give rise to progenitor cells that differentiate into the multiple cell types present in the epithelium. Here, we review the current state of research of how metabolism and ageing control intestinal stem cell function and epithelial homeostasis. We focus on recent insights gained from model organisms that indicate how changes in metabolic signalling during ageing are a major driver for the loss of stem cell plasticity and epithelial homeostasis, ultimately affecting the resilience of an organism and limiting its lifespan. We compare findings made in mouse and Drosophila and discuss differences and commonalities in the underlying signalling pathways and mechanisms in the context of ageing.
Collapse
Affiliation(s)
- Maja C Funk
- Division Signaling and Functional Genomics, German Cancer Research Center (DKFZ), Heidelberg University, Heidelberg, Germany
| | - Jun Zhou
- Division Signaling and Functional Genomics, German Cancer Research Center (DKFZ), Heidelberg University, Heidelberg, Germany
| | - Michael Boutros
- Division Signaling and Functional Genomics, German Cancer Research Center (DKFZ), Heidelberg University, Heidelberg, Germany
| |
Collapse
|
19
|
Buddika K, Ariyapala IS, Hazuga MA, Riffert D, Sokol NS. Canonical nucleators are dispensable for stress granule assembly in Drosophila intestinal progenitors. J Cell Sci 2020; 133:jcs243451. [PMID: 32265270 PMCID: PMC7325430 DOI: 10.1242/jcs.243451] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 03/26/2020] [Indexed: 12/18/2022] Open
Abstract
Stressed cells downregulate translation initiation and assemble membrane-less foci termed stress granules (SGs). Although SGs have been extensively characterized in cultured cells, the existence of such structures in stressed adult stem cell pools remains poorly characterized. Here, we report that the Drosophila orthologs of the mammalian SG components AGO1, ATX2, CAPRIN, eIF4E, FMRP, G3BP, LIN-28, PABP and TIAR are enriched in adult fly intestinal progenitor cells, where they accumulate in small cytoplasmic messenger ribonucleoprotein complexes (mRNPs). Treatment with sodium arsenite or rapamycin reorganized these mRNPs into large cytoplasmic granules. Formation of these intestinal progenitor stress granules (IPSGs) depended on polysome disassembly, led to translational downregulation and was reversible. Although the canonical SG nucleators ATX2 and G3BP were sufficient for IPSG formation in the absence of stress, neither of them, nor TIAR, either individually or collectively, were required for stress-induced IPSG formation. This work therefore finds that IPSGs do not assemble via a canonical mechanism, raising the possibility that other stem cell populations employ a similar stress-response mechanism.
Collapse
Affiliation(s)
- Kasun Buddika
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | | | - Mary A Hazuga
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Derek Riffert
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Nicholas S Sokol
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| |
Collapse
|
20
|
Sreejith P, Jang W, To V, Hun Jo Y, Biteau B, Kim C. Lin28 is a critical factor in the function and aging of Drosophila testis stem cell niche. Aging (Albany NY) 2020; 11:855-873. [PMID: 30713156 PMCID: PMC6382437 DOI: 10.18632/aging.101765] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 01/05/2019] [Indexed: 12/21/2022]
Abstract
Age-related decline in stem cell function is observed in many tissues from invertebrates to humans. While cell intrinsic alterations impair stem cells, aging of the stem cell niche also significantly contributes to the loss of tissue homeostasis associated with reduced regenerative capacity. Hub cells, which constitute the stem cell niche in the Drosophila testis, exhibit age-associated decline in number and activities, yet underlying mechanisms are not fully understood. Here we show that Lin28, a highly conserved RNA binding protein, is expressed in hub cells and its expression dramatically declines in old testis. lin28 mutant testes exhibit hub cell loss and defective hub architecture, recapitulating the normal aging process. Importantly, maintained expression of Lin28 prolongs hub integrity and function in aged testes, suggesting that Lin28 decline is a driver of hub cell aging. Mechanistically, the level of unpaired (upd), a stem cell self-renewal factor, is reduced in lin28 mutant testis and Lin28 protein directly binds and stabilizes upd transcripts, in a let-7 independent manner. Altogether, our results suggest that Lin28 acts to protect upd transcripts in hub cells, and reduction of Lin28 in old testis leads to decreased upd levels, hub cell aging and loss of the stem cell niche.
Collapse
Affiliation(s)
- Perinthottathil Sreejith
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea.,Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Wijeong Jang
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Van To
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Yong Hun Jo
- Division of Plant Biotechnology, Institute of Environmentally-Friendly Agriculture (IEFA), College of Agriculture and Life Sciences, Chonnam National University, Gwangju 500-757, Republic of Korea
| | - Benoit Biteau
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Changsoo Kim
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| |
Collapse
|
21
|
Lee M, Nguyen TMT, Kim K. In-depth study of lin-28 suggests selectively conserved let-7 independent mechanism in Drosophila. Gene 2019; 687:64-72. [DOI: 10.1016/j.gene.2018.11.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 11/03/2018] [Accepted: 11/07/2018] [Indexed: 12/13/2022]
|
22
|
Parham LR, Williams PA, Chatterji P, Whelan KA, Hamilton KE. RNA regulons are essential in intestinal homeostasis. Am J Physiol Gastrointest Liver Physiol 2019; 316:G197-G204. [PMID: 30520692 PMCID: PMC6383383 DOI: 10.1152/ajpgi.00403.2017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Intestinal epithelial cells are among the most rapidly proliferating cell types in the human body. There are several different subtypes of epithelial cells, each with unique functional roles in responding to the ever-changing environment. The epithelium's ability for rapid and customized responses to environmental changes requires multitiered levels of gene regulation. An emerging paradigm in gastrointestinal epithelial cells is the regulation of functionally related mRNA families, or regulons, via RNA-binding proteins (RBPs). RBPs represent a rapid and efficient mechanism to regulate gene expression and cell function. In this review, we will provide an overview of intestinal epithelial RBPs and how they contribute specifically to intestinal epithelial stem cell dynamics. In addition, we will highlight key gaps in knowledge in the global understanding of RBPs in gastrointestinal physiology as an opportunity for future studies.
Collapse
Affiliation(s)
- Louis R. Parham
- 1Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Patrick A. Williams
- 1Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Priya Chatterji
- 2Division of Gastroenterology, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kelly A. Whelan
- 3Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania,4Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Kathryn E. Hamilton
- 1Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| |
Collapse
|
23
|
Martin JL, Sanders EN, Moreno-Roman P, Jaramillo Koyama LA, Balachandra S, Du X, O'Brien LE. Long-term live imaging of the Drosophila adult midgut reveals real-time dynamics of division, differentiation and loss. eLife 2018; 7:36248. [PMID: 30427308 PMCID: PMC6277200 DOI: 10.7554/elife.36248] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 11/12/2018] [Indexed: 12/18/2022] Open
Abstract
Organ renewal is governed by the dynamics of cell division, differentiation and loss. To study these dynamics in real time, we present a platform for extended live imaging of the adult Drosophila midgut, a premier genetic model for stem-cell-based organs. A window cut into a living animal allows the midgut to be imaged while intact and physiologically functioning. This approach prolongs imaging sessions to 12–16 hr and yields movies that document cell and tissue dynamics at vivid spatiotemporal resolution. By applying a pipeline for movie processing and analysis, we uncover new and intriguing cell behaviors: that mitotic stem cells dynamically re-orient, that daughter cells use slow kinetics of Notch activation to reach a fate-specifying threshold, and that enterocytes extrude via ratcheted constriction of a junctional ring. By enabling real-time study of midgut phenomena that were previously inaccessible, our platform opens a new realm for dynamic understanding of adult organ renewal.
Collapse
Affiliation(s)
- Judy Lisette Martin
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, United States
| | - Erin Nicole Sanders
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, United States.,Department of Developmental Biology, Stanford University School of Medicine, Stanford, United States
| | - Paola Moreno-Roman
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, United States.,Department of Biology, Stanford University, Stanford, United States
| | - Leslie Ann Jaramillo Koyama
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, United States.,Department of Developmental Biology, Stanford University School of Medicine, Stanford, United States
| | - Shruthi Balachandra
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, United States
| | - XinXin Du
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, United States
| | - Lucy Erin O'Brien
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, United States
| |
Collapse
|
24
|
Dietrich C, Singh M, Kumar N, Singh SR. The Emerging Roles of microRNAs in Stem Cell Aging. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1056:11-26. [PMID: 29754172 DOI: 10.1007/978-3-319-74470-4_2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Aging is the continuous loss of tissue and organ function over time. MicroRNAs (miRNAs) are thought to play a vital role in this process. miRNAs are endogenous small noncoding RNAs that control the expression of target mRNA. They are involved in many biological processes such as developmental timing, differentiation, cell death, stem cell proliferation and differentiation, immune response, aging and cancer. Accumulating studies in recent years suggest that miRNAs play crucial roles in stem cell division and differentiation. In the present chapter, we present a brief overview of these studies and discuss their contributions toward our understanding of the importance of miRNAs in normal and aged stem cell function in various model systems.
Collapse
Affiliation(s)
- Catharine Dietrich
- Stem Cell Regulation and Animal Aging Section, Basic Research Laboratory, National Cancer Institute, Frederick, MD, USA
| | - Manish Singh
- Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD, USA
| | - Nishant Kumar
- Hospitalist Division, Department of Medicine, Inova Fairfax Medical Campus, Falls Church, VA, USA
| | - Shree Ram Singh
- Stem Cell Regulation and Animal Aging Section, Basic Research Laboratory, National Cancer Institute, Frederick, MD, USA.
| |
Collapse
|
25
|
Miguel-Aliaga I, Jasper H, Lemaitre B. Anatomy and Physiology of the Digestive Tract of Drosophila melanogaster. Genetics 2018; 210:357-396. [PMID: 30287514 PMCID: PMC6216580 DOI: 10.1534/genetics.118.300224] [Citation(s) in RCA: 288] [Impact Index Per Article: 41.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 07/26/2018] [Indexed: 12/15/2022] Open
Abstract
The gastrointestinal tract has recently come to the forefront of multiple research fields. It is now recognized as a major source of signals modulating food intake, insulin secretion and energy balance. It is also a key player in immunity and, through its interaction with microbiota, can shape our physiology and behavior in complex and sometimes unexpected ways. The insect intestine had remained, by comparison, relatively unexplored until the identification of adult somatic stem cells in the Drosophila intestine over a decade ago. Since then, a growing scientific community has exploited the genetic amenability of this insect organ in powerful and creative ways. By doing so, we have shed light on a broad range of biological questions revolving around stem cells and their niches, interorgan signaling and immunity. Despite their relatively recent discovery, some of the mechanisms active in the intestine of flies have already been shown to be more widely applicable to other gastrointestinal systems, and may therefore become relevant in the context of human pathologies such as gastrointestinal cancers, aging, or obesity. This review summarizes our current knowledge of both the formation and function of the Drosophila melanogaster digestive tract, with a major focus on its main digestive/absorptive portion: the strikingly adaptable adult midgut.
Collapse
Affiliation(s)
- Irene Miguel-Aliaga
- Medical Research Council London Institute of Medical Sciences, Imperial College London, W12 0NN, United Kingdom
| | - Heinrich Jasper
- Buck Institute for Research on Aging, Novato, California 94945-1400
- Immunology Discovery, Genentech, Inc., San Francisco, California 94080
| | - Bruno Lemaitre
- Global Health Institute, School of Life Sciences, École polytechnique fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| |
Collapse
|
26
|
Luhur A, Buddika K, Ariyapala IS, Chen S, Sokol NS. Opposing Post-transcriptional Control of InR by FMRP and LIN-28 Adjusts Stem Cell-Based Tissue Growth. Cell Rep 2018; 21:2671-2677. [PMID: 29212015 DOI: 10.1016/j.celrep.2017.11.039] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 10/13/2017] [Accepted: 11/10/2017] [Indexed: 01/01/2023] Open
Abstract
Although the intrinsic mechanisms that control whether stem cells divide symmetrically or asymmetrically underlie tissue growth and homeostasis, they remain poorly defined. We report that the RNA-binding protein fragile X mental retardation protein (FMRP) limits the symmetric division, and resulting expansion, of the stem cell population during adaptive intestinal growth in Drosophila. The elevated insulin sensitivity that FMRP-deficient progenitor cells display contributes to their accelerated expansion, which is suppressed by the depletion of insulin-signaling components. This FMRP activity is mediated solely via a second conserved RNA-binding protein, LIN-28, known to boost insulin signaling in stem cells. Via LIN-28, FMRP controls progenitor cell behavior by post-transcriptionally repressing the level of insulin receptor (InR). This study identifies the stem cell-based mechanism by which FMRP controls tissue adaptation, and it raises the possibility that defective adaptive growth underlies the accelerated growth, gastrointestinal, and other symptoms that affect fragile X syndrome patients.
Collapse
Affiliation(s)
- Arthur Luhur
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Kasun Buddika
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | | | - Shengyao Chen
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | | |
Collapse
|
27
|
A role for Lin-28 in growth and metamorphosis in Drosophila melanogaster. Mech Dev 2018; 154:107-115. [PMID: 29908237 DOI: 10.1016/j.mod.2018.06.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 06/04/2018] [Accepted: 06/12/2018] [Indexed: 12/25/2022]
Abstract
Insect metamorphosis has been a classic model to understand the role of hormones in growth and timing of developmental transitions. In addition to hormones, transitions in some species are regulated by genetic programs, such as the heterochronic gene network discovered in C. elegans. However, the functional link between hormones and heterochronic genes is not clear. The heterochronic gene lin-28 is involved in the maintenance of stem cells, growth and developmental timing in vertebrates. In this work, we used gain-of-function and loss-of-function experiments to study the role of Lin-28 in larval growth and the timing of metamorphosis of Drosophila melanogaster. During the late third instar stage, Lin-28 is mainly expressed in neurons of the central nervous system and in the intestine. Loss-of-function lin-28 mutant larvae are smaller and the larval-to-pupal transition is accelerated. This faster transition correlates with increased levels of ecdysone direct target genes such as Broad-Complex (BR-C) and Ecdysone Receptor (EcR). Overexpression of Lin-28 does not affect the timing of pupariation but most animals are not able to eclose, suggesting defects in metamorphosis. Overexpression of human Lin-28 results in delayed pupariation and the death of animals during metamorphosis. Altogether, these results suggest that Lin-28 is involved in the control of growth during larval development and in the timing and progression of metamorphosis.
Collapse
|
28
|
The zinc finger protein CG12744 is essential for differentiation and regeneration after infection in the adult Drosophila midgut. Exp Cell Res 2017; 361:225-235. [DOI: 10.1016/j.yexcr.2017.10.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Revised: 10/21/2017] [Accepted: 10/23/2017] [Indexed: 11/19/2022]
|
29
|
Gervais L, Bardin AJ. Tissue homeostasis and aging: new insight from the fly intestine. Curr Opin Cell Biol 2017; 48:97-105. [DOI: 10.1016/j.ceb.2017.06.005] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Revised: 05/29/2017] [Accepted: 06/23/2017] [Indexed: 12/12/2022]
|
30
|
Liu Q, Jin LH. Tissue-resident stem cell activity: a view from the adult Drosophila gastrointestinal tract. Cell Commun Signal 2017; 15:33. [PMID: 28923062 PMCID: PMC5604405 DOI: 10.1186/s12964-017-0184-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 07/12/2017] [Indexed: 12/11/2022] Open
Abstract
The gastrointestinal tract serves as a fast-renewing model for unraveling the multifaceted molecular mechanisms underlying remarkably rapid cell renewal, which is exclusively fueled by a small number of long-lived stem cells and their progeny. Stem cell activity is the best-characterized aspect of mucosal homeostasis in mitotically active tissues, and the dysregulation of regenerative capacity is a hallmark of epithelial immune defects. This dysregulation is frequently associated with pathologies ranging from chronic enteritis to malignancies in humans. Application of the adult Drosophila gastrointestinal tract model in current and future studies to analyze the immuno-physiological aspects of epithelial defense strategies, including stem cell behavior and re-epithelialization, will be necessary to improve our general understanding of stem cell participation in epithelial turnover. In this review, which describes exciting observations obtained from the adult Drosophila gastrointestinal tract, we summarize a remarkable series of recent findings in the literature to decipher the molecular mechanisms through which stem cells respond to nonsterile environments.
Collapse
Affiliation(s)
- Qiang Liu
- Department of Genetics, College of Life Sciences, Northeast Forestry University, No.26 Hexing Road Xiangfang District, Harbin, 150040, China
| | - Li Hua Jin
- Department of Genetics, College of Life Sciences, Northeast Forestry University, No.26 Hexing Road Xiangfang District, Harbin, 150040, China.
| |
Collapse
|
31
|
Luhur A, Sokol N. Starving for more: Nutrient sensing by LIN-28 in adult intestinal progenitor cells. Fly (Austin) 2016; 9:173-7. [PMID: 26934725 DOI: 10.1080/19336934.2016.1158366] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
In this Extra View, we extend our recent work on the protein LIN-28 and its role in adult stem cell divisions. LIN-28 is an mRNA- and microRNA-binding protein that is conserved from worms to humans. When expressed ectopically, it promotes the reprogramming of differentiated vertebrate cells into pluripotent stem cells as well as the regeneration of vertebrate tissues after injury. However, its endogenous function in stem cell populations is less clear. We recently reported that LIN-28 is specifically expressed in progenitor cells in the adult Drosophila intestine and enhances insulin signaling within this population. Loss of lin-28 alters the division patterns of these progenitor cells, limiting the growth of the intestinal epithelium that is ordinarily caused by feeding. Thus, LIN-28 is part of an uncharacterized circuit used to remodel a tissue in response to environmental cues like nutrition. Here, we extend this analysis by reporting that the levels of LIN-28 in progenitor cells are sensitive to nutrient availability. In addition, we speculate about the role of LIN-28 in the translational control of target mRNAs such as Insulin Receptor (InR) and how such translational control may be an important mechanism that underlies the stem cell dynamics needed for tissue homeostasis and growth.
Collapse
Affiliation(s)
- Arthur Luhur
- a Department of Biology ; Indiana University ; Bloomington , IN USA
| | - Nicholas Sokol
- a Department of Biology ; Indiana University ; Bloomington , IN USA
| |
Collapse
|
32
|
Jiang H, Tian A, Jiang J. Intestinal stem cell response to injury: lessons from Drosophila. Cell Mol Life Sci 2016; 73:3337-49. [PMID: 27137186 PMCID: PMC4998060 DOI: 10.1007/s00018-016-2235-9] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 04/13/2016] [Accepted: 04/21/2016] [Indexed: 12/14/2022]
Abstract
Many adult tissues and organs are maintained by resident stem cells that are activated in response to injury but the mechanisms that regulate stem cell activity during regeneration are still poorly understood. An emerging system to study such problem is the Drosophila adult midgut. Recent studies have identified both intrinsic factors and extrinsic niche signals that control the proliferation, self-renewal, and lineage differentiation of Drosophila adult intestinal stem cells (ISCs). These findings set up the stage to interrogate how niche signals are regulated and how they are integrated with cell-intrinsic factors to control ISC activity during normal homeostasis and regeneration. Here we review the current understanding of the mechanisms that control ISC self-renewal, proliferation, and lineage differentiation in Drosophila adult midgut with a focus on the niche signaling network that governs ISC activity in response to injury.
Collapse
Affiliation(s)
- Huaqi Jiang
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390, USA
| | - Aiguo Tian
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390, USA
| | - Jin Jiang
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390, USA.
- Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390, USA.
| |
Collapse
|
33
|
Chawla G, Deosthale P, Childress S, Wu YC, Sokol NS. A let-7-to-miR-125 MicroRNA Switch Regulates Neuronal Integrity and Lifespan in Drosophila. PLoS Genet 2016; 12:e1006247. [PMID: 27508495 PMCID: PMC4979967 DOI: 10.1371/journal.pgen.1006247] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 07/15/2016] [Indexed: 12/21/2022] Open
Abstract
Messenger RNAs (mRNAs) often contain binding sites for multiple, different microRNAs (miRNAs). However, the biological significance of this feature is unclear, since such co-targeting miRNAs could function coordinately, independently, or redundantly with one another. Here, we show that two co-transcribed Drosophila miRNAs, let-7 and miR-125, non-redundantly regulate a common target, the transcription factor Chronologically Inappropriate Morphogenesis (Chinmo). We first characterize novel adult phenotypes associated with loss of both let-7 and miR-125, which are derived from a common, polycistronic transcript that also encodes a third miRNA, miR-100. Consistent with the coordinate upregulation of all three miRNAs in aging flies, these phenotypes include brain degeneration and shortened lifespan. However, transgenic rescue analysis reveal separable roles for these miRNAs: adult miR-125 but not let-7 mutant phenotypes are associated with ectopic Chinmo expression in adult brains and are suppressed by chinmo reduction. In contrast, let-7 is predominantly responsible for regulating chinmo during nervous system formation. These results indicate that let-7 and miR-125 function during two distinct stages, development and adulthood, rather than acting at the same time. These different activities are facilitated by an increased rate of processing of let-7 during development and a lower rate of decay of the accumulated miR-125 in the adult nervous system. Thus, this work not only establishes a key role for the highly conserved miR-125 in aging. It also demonstrates that two co-transcribed miRNAs function independently during distinct stages to regulate a common target, raising the possibility that such biphasic control may be a general feature of clustered miRNAs. Deregulation of mRNAs that are targeted by multiple miRNAs is a common feature of a number of diseased states including neurodegenerative disorders. The currently accepted model is that the combined action of all binding miRNAs ensures target repression. Here, we show that two co-expressed miRNAs exert distinct outcomes on a common target. While miR-125 extends lifespan by repressing its target, chinmo, in adult brains, let-7 downregulates Chinmo in developing animals. Our results indicate that differential processing and turnover rates of let-7 and miR-125 contribute to this switch in miRNA activity. This study has identified the physiological relevance of the targeting of a single mRNA by multiple miRNAs in a scenario where each miRNA exerts a distinct and non-overlapping outcome.
Collapse
Affiliation(s)
- Geetanjali Chawla
- Department of Biology, Indiana University, Bloomington, Bloomington, Indiana, United States of America
- * E-mail: (GC); (NSS)
| | - Padmini Deosthale
- Department of Biology, Indiana University, Bloomington, Bloomington, Indiana, United States of America
| | - Sue Childress
- Medical Sciences Program, Indiana University, Bloomington, Bloomington, Indiana, United States of America
| | - Yen-chi Wu
- Department of Biology, Indiana University, Bloomington, Bloomington, Indiana, United States of America
| | - Nicholas S. Sokol
- Department of Biology, Indiana University, Bloomington, Bloomington, Indiana, United States of America
- * E-mail: (GC); (NSS)
| |
Collapse
|
34
|
Faunes F, Larraín J. Conservation in the involvement of heterochronic genes and hormones during developmental transitions. Dev Biol 2016; 416:3-17. [DOI: 10.1016/j.ydbio.2016.06.013] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 06/03/2016] [Accepted: 06/09/2016] [Indexed: 01/26/2023]
|
35
|
Narbonne-Reveau K, Lanet E, Dillard C, Foppolo S, Chen CH, Parrinello H, Rialle S, Sokol NS, Maurange C. Neural stem cell-encoded temporal patterning delineates an early window of malignant susceptibility in Drosophila. eLife 2016; 5:e13463. [PMID: 27296804 PMCID: PMC4907696 DOI: 10.7554/elife.13463] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 05/26/2016] [Indexed: 12/30/2022] Open
Abstract
Pediatric neural tumors are often initiated during early development and can undergo very rapid transformation. However, the molecular basis of this early malignant susceptibility remains unknown. During Drosophila development, neural stem cells (NSCs) divide asymmetrically and generate intermediate progenitors that rapidly differentiate in neurons. Upon gene inactivation, these progeny can dedifferentiate and generate malignant tumors. Here, we find that intermediate progenitors are prone to malignancy only when born during an early window of development while expressing the transcription factor Chinmo, and the mRNA-binding proteins Imp/IGF2BP and Lin-28. These genes compose an oncogenic module that is coopted upon dedifferentiation of early-born intermediate progenitors to drive unlimited tumor growth. In late larvae, temporal transcription factor progression in NSCs silences the module, thereby limiting mitotic potential and terminating the window of malignant susceptibility. Thus, this study identifies the gene regulatory network that confers malignant potential to neural tumors with early developmental origins.
Collapse
Affiliation(s)
| | - Elodie Lanet
- Aix Marseille Univ, CNRS, IBDM, Marseille, France
| | | | | | - Ching-Huan Chen
- Department of Biology, Indiana University, Bloomington, United States
| | - Hugues Parrinello
- MGX-Montpellier GenomiX, Institut de Génomique Fonctionnelle, Montpellier, France
| | - Stéphanie Rialle
- MGX-Montpellier GenomiX, Institut de Génomique Fonctionnelle, Montpellier, France
| | - Nicholas S Sokol
- Department of Biology, Indiana University, Bloomington, United States
| | | |
Collapse
|