1
|
Pathway to Independence - an interview with Keaton Schuster. Development 2024; 151:dev204230. [PMID: 39140265 DOI: 10.1242/dev.204230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Abstract
Keaton Schuster completed his PhD in the lab of Rachel Smith-Bolton at the University of Illinois, USA, investigating Drosophila wing disc regeneration before joining Lionel Christiaen's lab at New York University, USA, for his postdoc studying heart regeneration in the chordate tunicate Ciona robusta (formerly Ciona intestinalis type A). Keaton is part of the second cohort of Development's Pathway to Independence Programme fellows and we spoke to him over Teams to learn more about his career to date and his future plans for starting his own group continuing to use emerging model systems to study cardiac regeneration.
Collapse
|
2
|
Bose A, Schuster K, Kodali C, Sonam S, Smith-Bolton R. The pioneer transcription factor Zelda facilitates the exit from regeneration and restoration of patterning in Drosophila. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.30.596672. [PMID: 38854062 PMCID: PMC11160785 DOI: 10.1101/2024.05.30.596672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
For a damaged tissue to regenerate, the injured site must repair the wound, proliferate, and restore the correct patterning and cell types. We found that Zelda, a pioneer transcription factor largely known for its role in embryonic zygotic genome activation, is dispensable for normal wing development but crucial for wing disc patterning during regeneration. Impairing Zelda function during disc regeneration resulted in adult wings with a plethora of cell fate errors, affecting the veins, margins, and posterior compartment identity. Using CUT&RUN, we identified and validated targets of Zelda including the cell fate genes cut, Delta and achaete, which failed to return to their normal expression patterns upon loss of Zelda. In addition, Zelda controls expression of factors previously established to preserve cell fate during regeneration like taranis and osa, which stabilizes engrailed expression during regeneration, thereby preserving posterior identity. Finally, Zelda ensures proper expression of the integrins encoded by multiple edematous wings and myospheroid during regeneration to prevent blisters in the resuting adult wing. Thus, Zelda is crucial for maintaining cell fate and structural architecture of the regenerating tissue.
Collapse
Affiliation(s)
- Anish Bose
- Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Keaton Schuster
- Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Chandril Kodali
- Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Surabhi Sonam
- Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Rachel Smith-Bolton
- Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
3
|
Abidi SNF, Hsu FTY, Smith-Bolton RK. Regenerative growth is constrained by brain tumor to ensure proper patterning in Drosophila. PLoS Genet 2023; 19:e1011103. [PMID: 38127821 PMCID: PMC10769103 DOI: 10.1371/journal.pgen.1011103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 01/05/2024] [Accepted: 12/06/2023] [Indexed: 12/23/2023] Open
Abstract
Some animals respond to injury by inducing new growth to regenerate the lost structures. This regenerative growth must be carefully controlled and constrained to prevent aberrant growth and to allow correct organization of the regenerating tissue. However, the factors that restrict regenerative growth have not been identified. Using a genetic ablation system in the Drosophila wing imaginal disc, we have identified one mechanism that constrains regenerative growth, impairment of which also leads to erroneous patterning of the final appendage. Regenerating discs with reduced levels of the RNA-regulator Brain tumor (Brat) exhibit enhanced regeneration, but produce adult wings with disrupted margins that are missing extensive tracts of sensory bristles. In these mutants, aberrantly high expression of the pro-growth factor Myc and its downstream targets likely contributes to this loss of cell-fate specification. Thus, Brat constrains the expression of pro-regeneration genes and ensures that the regenerating tissue forms the proper final structure.
Collapse
Affiliation(s)
- Syeda Nayab Fatima Abidi
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Felicity Ting-Yu Hsu
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Rachel K. Smith-Bolton
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Carle R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| |
Collapse
|
4
|
Harris RE. Regeneration enhancers: a field in development. Am J Physiol Cell Physiol 2022; 323:C1548-C1554. [PMID: 36252130 PMCID: PMC9829460 DOI: 10.1152/ajpcell.00403.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The ability to regenerate tissues and organs following damage is not equally distributed across metazoans, and even highly related species can vary considerably in their regenerative capacity. Studies of animals with high regenerative potential have shown that factors expressed during normal development are often reactivated upon damage and required for successful regeneration. As such, regenerative potential may not be dictated by the presence or absence of the necessary genes, but whether such genes are appropriately activated following injury. The identification of damage-responsive enhancers that regulate regenerative gene expression in multiple species and tissues provides possible mechanistic insight into this phenomenon. Enhancers that are reused from developmental programs, and those that are potentially unique to regeneration, have been characterized individually and at a genome-wide scale. A better understanding of the regulatory events that, direct and in some cases limit, regenerative capacity is an important step in developing new methods to manipulate and augment regeneration, particularly in tissues that do not have this ability, including those of humans.
Collapse
Affiliation(s)
- Robin E. Harris
- School of Life Sciences, Arizona State University, Tempe, Arizona
| |
Collapse
|
5
|
Pulianmackal AJ, Kanakousaki K, Flegel K, Grushko OG, Gourley E, Rozich E, Buttitta LA. Misregulation of Nucleoporins 98 and 96 leads to defects in protein synthesis that promote hallmarks of tumorigenesis. Dis Model Mech 2022; 15:dmm049234. [PMID: 35107131 PMCID: PMC8938402 DOI: 10.1242/dmm.049234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 01/15/2022] [Indexed: 11/20/2022] Open
Abstract
Nucleoporin 98KD (Nup98) is a promiscuous translocation partner in hematological malignancies. Most disease models of Nup98 translocations involve ectopic expression of the fusion protein under study, leaving the endogenous Nup98 loci unperturbed. Overlooked in these approaches is the loss of one copy of normal Nup98 in addition to the loss of Nup96 - a second Nucleoporin encoded within the same mRNA and reading frame as Nup98 - in translocations. Nup98 and Nup96 are also mutated in a number of other cancers, suggesting that their disruption is not limited to blood cancers. We found that reducing Nup98-96 function in Drosophila melanogaster (in which the Nup98-96 shared mRNA and reading frame is conserved) de-regulates the cell cycle. We found evidence of overproliferation in tissues with reduced Nup98-96, counteracted by elevated apoptosis and aberrant signaling associated with chronic wounding. Reducing Nup98-96 function led to defects in protein synthesis that triggered JNK signaling and contributed to hallmarks of tumorigenesis when apoptosis was inhibited. We suggest that partial loss of Nup98-96 function in translocations could de-regulate protein synthesis, leading to signaling that cooperates with other mutations to promote tumorigenesis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Laura A. Buttitta
- Molecular Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
6
|
Ledru M, Clark CA, Brown J, Verghese S, Ferrara S, Goodspeed A, Su TT. Differential gene expression analysis identified determinants of cell fate plasticity during radiation-induced regeneration in Drosophila. PLoS Genet 2022; 18:e1009989. [PMID: 34990447 PMCID: PMC8769364 DOI: 10.1371/journal.pgen.1009989] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 01/19/2022] [Accepted: 12/13/2021] [Indexed: 12/28/2022] Open
Abstract
Ionizing radiation (IR) is used to treat half of all cancer patients because of its ability to kill cells. IR, however, can induce stem cell-like properties in non-stem cancer cells, potentiating tumor regrowth and reduced therapeutic success. We identified previously a subpopulation of cells in Drosophila larval wing discs that exhibit IR-induced stem cell-like properties. These cells reside in the future wing hinge, are resistant to IR-induced apoptosis, and are capable of translocating, changing fate, and participating in regenerating the pouch that suffers more IR-induced apoptosis. We used here a combination of lineage tracing, FACS-sorting of cells that change fate, genome-wide RNAseq, and functional testing of 42 genes, to identify two key changes that are required cell-autonomously for IR-induced hinge-to-pouch fate change: (1) repression of hinge determinants Wg (Drosophila Wnt1) and conserved zinc-finger transcription factor Zfh2 and (2) upregulation of three ribosome biogenesis factors. Additional data indicate a role for Myc, a transcriptional activator of ribosome biogenesis genes, in the process. These results provide a molecular understanding of IR-induced cell fate plasticity that may be leveraged to improve radiation therapy. Ionizing radiation (IR) is used to treat half of all cancer patients because of its ability to kill cells but treatment failures are common because tumors grow back (regenerate). Here, we asked which changes in the properties of cells facilitate regeneration in Drosophila (fruit flies) after exposure to radiation. We identified six genes whose products increase or decrease the regenerative potential of cells. These results help us understand how tissues regenerate after IR damage and will aid in designing better therapies that involve radiation.
Collapse
Affiliation(s)
- Michelle Ledru
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, United States of America
| | - Caitlin A. Clark
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, United States of America
| | - Jeremy Brown
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, United States of America
| | - Shilpi Verghese
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, United States of America
| | - Sarah Ferrara
- University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Andrew Goodspeed
- University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, Colorado, United States of America
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Tin Tin Su
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, United States of America
- University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, Colorado, United States of America
- * E-mail:
| |
Collapse
|
7
|
Tian Y, Smith-Bolton RK. Regulation of growth and cell fate during tissue regeneration by the two SWI/SNF chromatin-remodeling complexes of Drosophila. Genetics 2021; 217:1-16. [PMID: 33683366 DOI: 10.1093/genetics/iyaa028] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 11/10/2020] [Indexed: 11/12/2022] Open
Abstract
To regenerate, damaged tissue must heal the wound, regrow to the proper size, replace the correct cell types, and return to the normal gene-expression program. However, the mechanisms that temporally and spatially control the activation or repression of important genes during regeneration are not fully understood. To determine the role that chromatin modifiers play in regulating gene expression after tissue damage, we induced ablation in Drosophila melanogaster imaginal wing discs, and screened for chromatin regulators that are required for epithelial tissue regeneration. Here, we show that many of these genes are indeed important for promoting or constraining regeneration. Specifically, the two SWI/SNF chromatin-remodeling complexes play distinct roles in regulating different aspects of regeneration. The PBAP complex regulates regenerative growth and developmental timing, and is required for the expression of JNK signaling targets and the growth promoter Myc. By contrast, the BAP complex ensures correct patterning and cell fate by stabilizing the expression of the posterior gene engrailed. Thus, both SWI/SNF complexes are essential for proper gene expression during tissue regeneration, but they play distinct roles in regulating growth and cell fate.
Collapse
Affiliation(s)
- Yuan Tian
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Rachel K Smith-Bolton
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.,Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
8
|
Krautz R, Khalili D, Theopold U. Tissue-autonomous immune response regulates stress signaling during hypertrophy. eLife 2020; 9:64919. [PMID: 33377870 PMCID: PMC7880693 DOI: 10.7554/elife.64919] [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: 11/15/2020] [Accepted: 12/29/2020] [Indexed: 12/19/2022] Open
Abstract
Postmitotic tissues are incapable of replacing damaged cells through proliferation, but need to rely on buffering mechanisms to prevent tissue disintegration. By constitutively activating the Ras/MAPK-pathway via RasV12-overexpression in the postmitotic salivary glands (SGs) of Drosophila larvae, we overrode the glands adaptability to growth signals and induced hypertrophy. The accompanied loss of tissue integrity, recognition by cellular immunity, and cell death are all buffered by blocking stress signaling through a genuine tissue-autonomous immune response. This novel, spatio-temporally tightly regulated mechanism relies on the inhibition of a feedback-loop in the JNK-pathway by the immune effector and antimicrobial peptide Drosomycin. While this interaction might allow growing SGs to cope with temporary stress, continuous Drosomycin expression in RasV12-glands favors unrestricted hypertrophy. These findings indicate the necessity to refine therapeutic approaches that stimulate immune responses by acknowledging their possible, detrimental effects in damaged or stressed tissues. Tissues and organs work hard to maintain balance in everything from taking up nutrients to controlling their growth. Ageing, wounding, sickness, and changes in the genetic code can all alter this balance, and cause the tissue or organ to lose some of its cells. Many tissues restore this loss by dividing their remaining cells to fill in the gaps. But some – like the salivary glands of fruit fly larvae – have lost this ability. Tissues like these rely on being able to sense and counteract problems as they arise so as to not lose their balance in the first place. The immune system and stress responses are crucial for this process. They trigger steps to correct the problem and interact with each other to find a common decision about the fate of the affected tissue. To better understand how the immune system and stress response work together, Krautz, Khalili and Theopold genetically manipulated cells in the salivary gland of fruit fly larvae. These modifications switched on signals that stimulate cells to keep growing, causing the salivary gland’s tissue to slowly lose its balance and trigger the stress and immune response. The experiments showed that while the stress response instructed the cells in the gland to die, a peptide released by the immune system called Drosomycin blocked this response and prevented the tissue from collapsing. The cells in the part of the gland not producing this immune peptide were consequently killed by the stress response. When all the cells in the salivary gland were forced to produce Drosomycin, none of the cells died and the whole tissue survived. But it also allowed the cells in the gland to grow uncontrollably, like a tumor, threatening the health of the entire organism. Mapping the interactions between immune and stress pathways could help to fine-tune treatments that can prevent tissue damage. Fruit flies share many genetic features and molecular pathways with humans. So, the next step towards these kinds of treatments would be to screen for similar mechanisms that block stress activation in damaged human tissues. But this research carries a warning: careless activation of the immune system to protect stressed tissues could lead to uncontrolled tissue growth, and might cause more harm than good.
Collapse
Affiliation(s)
- Robert Krautz
- Department of Molecular Biosciences, The Wenner-Gren Institute (MBW), Stockholm University, Stockholm, Sweden
| | - Dilan Khalili
- Department of Molecular Biosciences, The Wenner-Gren Institute (MBW), Stockholm University, Stockholm, Sweden
| | - Ulrich Theopold
- Department of Molecular Biosciences, The Wenner-Gren Institute (MBW), Stockholm University, Stockholm, Sweden
| |
Collapse
|
9
|
Bairzin JCD, Emmons-Bell M, Hariharan IK. The Hippo pathway coactivator Yorkie can reprogram cell fates and create compartment-boundary-like interactions at clone margins. SCIENCE ADVANCES 2020; 6:6/50/eabe8159. [PMID: 33298454 PMCID: PMC7725458 DOI: 10.1126/sciadv.abe8159] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 10/22/2020] [Indexed: 06/12/2023]
Abstract
During development, tissue-specific patterns of gene expression are established by transcription factors and then stably maintained via epigenetic mechanisms. Cancer cells often express genes that are inappropriate for that tissue or developmental stage. Here, we show that high activity levels of Yki, the Hippo pathway coactivator that causes overgrowth in Drosophila imaginal discs, can also disrupt cell fates by altering expression of selector genes like engrailed (en) and Ultrabithorax (Ubx). Posterior clones expressing activated Yki can down-regulate en and express an anterior selector gene, cubitus interruptus (ci). The microRNA bantam and the chromatin regulator Taranis both function downstream of Yki in promoting ci expression. The boundary between Yki-expressing posterior clones and surrounding wild-type cells acquires properties reminiscent of the anteroposterior compartment boundary; Hedgehog signaling pathway activation results in production of Dpp. Thus, at least in principle, heterotypic interactions between Yki-expressing cells and their neighbors could activate boundary-specific signaling mechanisms.
Collapse
Affiliation(s)
- Joanna C D Bairzin
- Department of Molecular and Cell Biology, 515 Weill Hall, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Maya Emmons-Bell
- Department of Molecular and Cell Biology, 515 Weill Hall, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Iswar K Hariharan
- Department of Molecular and Cell Biology, 515 Weill Hall, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
10
|
Overton IM, Sims AH, Owen JA, Heale BSE, Ford MJ, Lubbock ALR, Pairo-Castineira E, Essafi A. Functional Transcription Factor Target Networks Illuminate Control of Epithelial Remodelling. Cancers (Basel) 2020; 12:cancers12102823. [PMID: 33007944 PMCID: PMC7652213 DOI: 10.3390/cancers12102823] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 09/16/2020] [Accepted: 09/24/2020] [Indexed: 12/15/2022] Open
Abstract
Cell identity is governed by gene expression, regulated by transcription factor (TF) binding at cis-regulatory modules. Decoding the relationship between TF binding patterns and gene regulation is nontrivial, remaining a fundamental limitation in understanding cell decision-making. We developed the NetNC software to predict functionally active regulation of TF targets; demonstrated on nine datasets for the TFs Snail, Twist, and modENCODE Highly Occupied Target (HOT) regions. Snail and Twist are canonical drivers of epithelial to mesenchymal transition (EMT), a cell programme important in development, tumour progression and fibrosis. Predicted "neutral" (non-functional) TF binding always accounted for the majority (50% to 95%) of candidate target genes from statistically significant peaks and HOT regions had higher functional binding than most of the Snail and Twist datasets examined. Our results illuminated conserved gene networks that control epithelial plasticity in development and disease. We identified new gene functions and network modules including crosstalk with notch signalling and regulation of chromatin organisation, evidencing networks that reshape Waddington's epigenetic landscape during epithelial remodelling. Expression of orthologous functional TF targets discriminated breast cancer molecular subtypes and predicted novel tumour biology, with implications for precision medicine. Predicted invasion roles were validated using a tractable cell model, supporting our approach.
Collapse
Affiliation(s)
- Ian M. Overton
- MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK; (A.H.S.); (B.S.E.H.); (M.J.F.); (A.L.R.L.); (E.P.-C.); (A.E.)
- Department of Systems Biology, Harvard University, Boston, MA 02115, USA;
- Centre for Synthetic and Systems Biology (SynthSys), University of Edinburgh, Edinburgh EH9 3BF, UK
- Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast BT9 7AE, UK
- Correspondence:
| | - Andrew H. Sims
- MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK; (A.H.S.); (B.S.E.H.); (M.J.F.); (A.L.R.L.); (E.P.-C.); (A.E.)
| | - Jeremy A. Owen
- Department of Systems Biology, Harvard University, Boston, MA 02115, USA;
- Department of Physics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Bret S. E. Heale
- MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK; (A.H.S.); (B.S.E.H.); (M.J.F.); (A.L.R.L.); (E.P.-C.); (A.E.)
| | - Matthew J. Ford
- MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK; (A.H.S.); (B.S.E.H.); (M.J.F.); (A.L.R.L.); (E.P.-C.); (A.E.)
| | - Alexander L. R. Lubbock
- MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK; (A.H.S.); (B.S.E.H.); (M.J.F.); (A.L.R.L.); (E.P.-C.); (A.E.)
| | - Erola Pairo-Castineira
- MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK; (A.H.S.); (B.S.E.H.); (M.J.F.); (A.L.R.L.); (E.P.-C.); (A.E.)
| | - Abdelkader Essafi
- MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK; (A.H.S.); (B.S.E.H.); (M.J.F.); (A.L.R.L.); (E.P.-C.); (A.E.)
| |
Collapse
|
11
|
Dutta P, Nath S, Li J, Li WX. Drosophila SERTAD domain protein Taranis is required in somatic cells for maintenance of male germline stem cells. Dev Dyn 2020; 250:237-248. [PMID: 32969117 DOI: 10.1002/dvdy.255] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/15/2020] [Accepted: 09/21/2020] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND Polycomb proteins are essential for maintaining stem cell identity across different stem cell niches. However, how they function to maintain stem cell niches is not fully understood. RESULTS Here we show that the SERTAD protein Taranis (Tara), which is a Polycomb-trithorax group protein, is expressed in the adult testis niche and plays a role in its maintenance in Drosophila. We found that tara is expressed in early cyst cells, likely including somatic cyst stem cells (CySCs) of Drosophila male testis tip region, which houses both germline and somatic cyst stem cells along with the hub cells, forming the stem cell niche. Consistent with its expression, we found that, while loss of tara in germline cells only had minimal effects, tara knockdown in all cells or only in somatic cells of the niche reduced the number of not only somatic cells, but also germline stem cells (GSCs). We further found that Tara might antagonize Notch signaling in CySCs to maintain the stem cell niche. CONCLUSIONS Our studies suggest that Tara might function in somatic CySCs for GSC maintenance in the Drosophila testis.
Collapse
Affiliation(s)
- Pranabananda Dutta
- Department of Medicine, University of California San Diego, La Jolla, California, USA.,Division of Cancer Research and Training, Department of Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, California, USA
| | - Soma Nath
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Jinghong Li
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Willis X Li
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
12
|
Abstract
Regeneration is the process by which organisms replace lost or damaged tissue, and regenerative capacity can vary greatly among species, tissues and life stages. Tissue regeneration shares certain hallmarks of embryonic development, in that lineage-specific factors can be repurposed upon injury to initiate morphogenesis; however, many differences exist between regeneration and embryogenesis. Recent studies of regenerating tissues in laboratory model organisms - such as acoel worms, frogs, fish and mice - have revealed that chromatin structure, dedicated enhancers and transcriptional networks are regulated in a context-specific manner to control key gene expression programmes. A deeper mechanistic understanding of the gene regulatory networks of regeneration pathways might ultimately enable their targeted reactivation as a means to treat human injuries and degenerative diseases. In this Review, we consider the regeneration of body parts across a range of tissues and species to explore common themes and potentially exploitable elements.
Collapse
Affiliation(s)
- Joseph A Goldman
- Department of Biological Chemistry and Pharmacology, Ohio State University, Columbus, OH, USA.
| | - Kenneth D Poss
- Regeneration Next, Duke University, Durham, NC, USA.
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
13
|
Abstract
Drosophila melanogaster has historically been a workhorse model organism for studying developmental biology. In addition, Drosophila is an excellent model for studying how damaged tissues and organs can regenerate. Recently, new precision approaches that enable both highly targeted injury and genetic manipulation have accelerated progress in this field. Here, we highlight these techniques and review examples of recently discovered mechanisms that regulate regeneration in Drosophila larval and adult tissues. We also discuss how, by applying these powerful approaches, studies of Drosophila can continue to guide the future of regeneration research.
Collapse
Affiliation(s)
- Donald T Fox
- Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
- Regeneration Next, Duke University, Durham, NC 27710, USA
| | - Erez Cohen
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
- Regeneration Next, Duke University, Durham, NC 27710, USA
| | - Rachel Smith-Bolton
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
14
|
Chromatin dynamics in regeneration epithelia: Lessons from Drosophila imaginal discs. Semin Cell Dev Biol 2020; 97:55-62. [DOI: 10.1016/j.semcdb.2019.04.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 04/29/2019] [Indexed: 12/21/2022]
|
15
|
Lee LR, Wengier DL, Bergmann DC. Cell-type-specific transcriptome and histone modification dynamics during cellular reprogramming in the Arabidopsis stomatal lineage. Proc Natl Acad Sci U S A 2019; 116:21914-21924. [PMID: 31594845 PMCID: PMC6815143 DOI: 10.1073/pnas.1911400116] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Plant cells maintain remarkable developmental plasticity, allowing them to clonally reproduce and to repair tissues following wounding; yet plant cells normally stably maintain consistent identities. Although this capacity was recognized long ago, our mechanistic understanding of the establishment, maintenance, and erasure of cellular identities in plants remains limited. Here, we develop a cell-type-specific reprogramming system that can be probed at the genome-wide scale for alterations in gene expression and histone modifications. We show that relationships among H3K27me3, H3K4me3, and gene expression in single cell types mirror trends from complex tissue, and that H3K27me3 dynamics regulate guard cell identity. Further, upon initiation of reprogramming, guard cells induce H3K27me3-mediated repression of a regulator of wound-induced callus formation, suggesting that cells in intact tissues may have mechanisms to sense and resist inappropriate dedifferentiation. The matched ChIP-sequencing (seq) and RNA-seq datasets created for this analysis also serve as a resource enabling inquiries into the dynamic and global-scale distribution of histone modifications in single cell types in plants.
Collapse
Affiliation(s)
- Laura R Lee
- Department of Biology, Stanford University, Stanford, CA 94305
| | - Diego L Wengier
- Department of Biology, Stanford University, Stanford, CA 94305
- HHMI, Stanford University, Stanford, CA 94305
| | - Dominique C Bergmann
- Department of Biology, Stanford University, Stanford, CA 94305;
- HHMI, Stanford University, Stanford, CA 94305
| |
Collapse
|
16
|
Verghese S, Su TT. Ionizing radiation induces stem cell-like properties in a caspase-dependent manner in Drosophila. PLoS Genet 2018; 14:e1007659. [PMID: 30462636 PMCID: PMC6248896 DOI: 10.1371/journal.pgen.1007659] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 08/27/2018] [Indexed: 11/18/2022] Open
Abstract
Cancer treatments including ionizing radiation (IR) can induce cancer stem cell-like properties in non-stem cancer cells, an outcome that can interfere with therapeutic success. Yet, we understand little about what consequences of IR induces stem cell like properties and why some cancer cells show this response but not others. In previous studies, we identified a pool of epithelial cells in Drosophila larval wing discs that display IR-induced stem cell-like properties. These cells are resistant to killing by IR and, after radiation damage, change fate and translocate to regenerate parts of the disc that suffered more cell death. Here, we report the identification of two new pools of cells with IR-induced regenerative capability. We addressed how IR exposure results in the induction of stem cell-like behavior, and found a requirement for IR-induced caspase activity and for Zfh2, a transcription factor and an effector in the JAK/STAT pathway. Unexpectedly, the requirement for caspase activity was cell-autonomous within cell populations that display regenerative behavior. We propose a model in which the requirement for caspase activity and Zfh2 can be explained by apoptotic and non-apoptotic functions of caspases in the induction of stem cell-like behavior. Ionizing Radiation (IR), alone or in combination with other therapies, is used to treat an estimated half of all cancer patients. Yet, we understand little about why some tumors cells respond to treatment while others grow back (regenerate). We identified specific pools of cells within a Drosophila organ that are capable of regeneration after damage by IR. We also identified what it is about IR damage that allows these cells to regenerate. These results help us understand how tissues regenerate after IR damage and will aid in designing better therapies that involve radiation.
Collapse
Affiliation(s)
- Shilpi Verghese
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO, United States of America
| | - Tin Tin Su
- University of Colorado Comprehensive Cancer Center, Anschutz Medical Campus, Aurora, CO, United States of America
- * E-mail:
| |
Collapse
|
17
|
Drosophila as a Model System to Study Cell Signaling in Organ Regeneration. BIOMED RESEARCH INTERNATIONAL 2018; 2018:7359267. [PMID: 29750169 PMCID: PMC5884440 DOI: 10.1155/2018/7359267] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 02/06/2018] [Indexed: 12/22/2022]
Abstract
Regeneration is a fascinating phenomenon that allows organisms to replace or repair damaged organs or tissues. This ability occurs to varying extents among metazoans. The rebuilding of the damaged structure depends on regenerative proliferation that must be accompanied by proper cell fate respecification and patterning. These cellular processes are regulated by the action of different signaling pathways that are activated in response to the damage. The imaginal discs of Drosophila melanogaster have the ability to regenerate and have been extensively used as a model system to study regeneration. Drosophila provides an opportunity to use powerful genetic tools to address fundamental problems about the genetic mechanisms involved in organ regeneration. Different studies in Drosophila have helped to elucidate the genes and signaling pathways that initiate regeneration, promote regenerative growth, and induce cell fate respecification. Here we review the signaling networks involved in regulating the variety of cellular responses that are required for discs regeneration.
Collapse
|
18
|
Worley MI, Alexander LA, Hariharan IK. CtBP impedes JNK- and Upd/STAT-driven cell fate misspecifications in regenerating Drosophila imaginal discs. eLife 2018; 7:30391. [PMID: 29372681 PMCID: PMC5823544 DOI: 10.7554/elife.30391] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 01/19/2018] [Indexed: 12/27/2022] Open
Abstract
Regeneration following tissue damage often necessitates a mechanism for cellular re-programming, so that surviving cells can give rise to all cell types originally found in the damaged tissue. This process, if unchecked, can also generate cell types that are inappropriate for a given location. We conducted a screen for genes that negatively regulate the frequency of notum-to-wing transformations following genetic ablation and regeneration of the wing pouch, from which we identified mutations in the transcriptional co-repressor C-terminal Binding Protein (CtBP). When CtBP function is reduced, ablation of the pouch can activate the JNK/AP-1 and JAK/STAT pathways in the notum to destabilize cell fates. Ectopic expression of Wingless and Dilp8 precede the formation of the ectopic pouch, which is subsequently generated by recruitment of both anterior and posterior cells near the compartment boundary. Thus, CtBP stabilizes cell fates following damage by opposing the destabilizing effects of the JNK/AP-1 and JAK/STAT pathways.
Collapse
Affiliation(s)
- Melanie I Worley
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Larissa A Alexander
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Iswar K Hariharan
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| |
Collapse
|
19
|
Abstract
Understanding how and why animals regenerate complex tissues has the potential to transform regenerative medicine. Here we present an overview of genetic approaches that have recently been applied to dissect mechanisms of regeneration. We describe new advances that relate to central objectives of regeneration biologists researching different tissues and species, focusing mainly on vertebrates. These objectives include defining the cellular sources and key cell behaviors in regenerating tissue, elucidating molecular triggers and brakes for regeneration, and defining the earliest events that control the presence of these molecular factors.
Collapse
Affiliation(s)
- Chen-Hui Chen
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan;
| | - Kenneth D Poss
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina 27710, USA;
- Regeneration Next, Duke University, Durham, North Carolina 27710, USA
| |
Collapse
|
20
|
STAT, Wingless, and Nurf-38 determine the accuracy of regeneration after radiation damage in Drosophila. PLoS Genet 2017; 13:e1007055. [PMID: 29028797 PMCID: PMC5656321 DOI: 10.1371/journal.pgen.1007055] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 10/25/2017] [Accepted: 10/04/2017] [Indexed: 01/01/2023] Open
Abstract
We report here a study of regeneration in Drosophila larval wing imaginal discs after damage by ionizing radiation. We detected faithful regeneration that restored a wing disc and abnormal regeneration that produced an extra wing disc. We describe a sequence of changes in cell number, location and fate that occur to produce an ectopic disc. We identified a group of cells that not only participate in ectopic disc formation but also recruit others to do so. STAT92E (Drosophila STAT3/5) and Nurf-38, which encodes a member of the Nucleosome Remodeling Factor complex, oppose each other in these cells to modulate the frequency of ectopic disc growth. The picture that emerges is one in which activities like STAT increase after radiation damage and fulfill essential roles in rebuilding the tissue. But such activities must be kept in check so that one and only one wing disc is regenerated. Accuracy in regeneration ensures that the original structures are restored, no more and no less. Prior studies in the wing primordia of Drosophila melanogaster larvae that have been damaged by high energy radiation show that regeneration occurs to restore the original structure. We report here that, in the same experimental system, abnormal regeneration can also occur to produce extra wing structures. We describe a series of cell rearrangements and fate changes that underlie abnormal regeneration, and identify genes responsible for these events. Modulation of such genes have the potential to mitigate abnormal regeneration that occurs after radiation damage to produce such side effects as ulcers and fibrosis.
Collapse
|
21
|
Hariharan IK, Serras F. Imaginal disc regeneration takes flight. Curr Opin Cell Biol 2017; 48:10-16. [PMID: 28376317 PMCID: PMC5591769 DOI: 10.1016/j.ceb.2017.03.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 03/07/2017] [Accepted: 03/10/2017] [Indexed: 12/20/2022]
Abstract
Drosophila imaginal discs, the larval precursors of adult structures such as the wing and leg, are capable of regenerating after damage. During the course of regeneration, discs can sometimes generate structures that are appropriate for a different type of disc, a phenomenon termed transdetermination. Until recently, these phenomena were studied by physically fragmenting discs and then transplanting them into the abdomens of adult female flies. This field has experienced a renaissance following the development of genetic ablation systems that can damage precisely defined regions of the disc without the need for surgery. Together with more traditional approaches, these newer methods have generated many novel insights into wound healing, the mechanisms that drive regenerative growth, plasticity during regeneration and systemic effects of tissue damage and regeneration.
Collapse
Affiliation(s)
- Iswar K Hariharan
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720-3200, USA.
| | - Florenci Serras
- Departament de Genètica, Facultat de Biologia and Institute de Biomedicina (IBUB), Universitat de Barcelona, Diagonal 645, 08028 Barcelona, Spain.
| |
Collapse
|
22
|
Khan SJ, Abidi SNF, Skinner A, Tian Y, Smith-Bolton RK. The Drosophila Duox maturation factor is a key component of a positive feedback loop that sustains regeneration signaling. PLoS Genet 2017; 13:e1006937. [PMID: 28753614 PMCID: PMC5550008 DOI: 10.1371/journal.pgen.1006937] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 08/09/2017] [Accepted: 07/20/2017] [Indexed: 12/21/2022] Open
Abstract
Regenerating tissue must initiate the signaling that drives regenerative growth, and sustain that signaling long enough for regeneration to complete. How these key signals are sustained is unclear. To gain a comprehensive view of the changes in gene expression that occur during regeneration, we performed whole-genome mRNAseq of actively regenerating tissue from damaged Drosophila wing imaginal discs. We used genetic tools to ablate the wing primordium to induce regeneration, and carried out transcriptional profiling of the regeneration blastema by fluorescently labeling and sorting the blastema cells, thus identifying differentially expressed genes. Importantly, by using genetic mutants of several of these differentially expressed genes we have confirmed that they have roles in regeneration. Using this approach, we show that high expression of the gene moladietz (mol), which encodes the Duox-maturation factor NIP, is required during regeneration to produce reactive oxygen species (ROS), which in turn sustain JNK signaling during regeneration. We also show that JNK signaling upregulates mol expression, thereby activating a positive feedback signal that ensures the prolonged JNK activation required for regenerative growth. Thus, by whole-genome transcriptional profiling of regenerating tissue we have identified a positive feedback loop that regulates the extent of regenerative growth. Regenerating tissue must initiate the signaling that drives regenerative growth, and then sustain that signaling long enough for regeneration to complete. Drosophila imaginal discs, the epithelial structures in the larva that will form the adult animal during metamorphosis, have been an important model system for tissue repair and regeneration for over 60 years. Here we show that damage-induced JNK signaling leads to the upregulation of a gene called moladietz, which encodes a co-factor for an enzyme, NADPH dual oxidase (Duox), that generates reactive oxygen species (ROS), a key tissue-damage signal. High expression of moladietz induces continuous production of ROS in the regenerating tissue. The sustained production of ROS then continues to activate JNK signaling throughout the course of regeneration, ensuring maximal tissue regrowth.
Collapse
Affiliation(s)
- Sumbul Jawed Khan
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL, United States of America
| | - Syeda Nayab Fatima Abidi
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL, United States of America
| | - Andrea Skinner
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL, United States of America
| | - Yuan Tian
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL, United States of America
| | - Rachel K. Smith-Bolton
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL, United States of America
- * E-mail:
| |
Collapse
|
23
|
Dutta P, Li WX. The SERTAD protein Taranis plays a role in Polycomb-mediated gene repression. PLoS One 2017; 12:e0180026. [PMID: 28665982 PMCID: PMC5493352 DOI: 10.1371/journal.pone.0180026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 06/08/2017] [Indexed: 11/19/2022] Open
Abstract
The Polycomb group (PcG) proteins have been implicated in epigenetic transcriptional repression in development, stem cell maintenance and in cancer. The chromodomain protein Polycomb (Pc) is a key member of the PcG. Pc binds to the histone mark, trimethylated histone 3 lysine 27 (H3K27me3), to initiate transcriptional repression. How PcG proteins are recruited to target loci is not fully understood. Here we show that the Drosophila SERTA domain protein Taranis (Tara) is involved in transcriptional regulation of Pc target genes. Embryos lacking Tara exhibit a partial homeotic transformation of cuticular the segments, a phenotype associated with the loss of Pc function. Moreover, Drosophila embryos homozygous for a tara hypomorphic allele also misexpress engrailed, a Pc-regulated gene, and this phenotype is associated with the loss of Pc binding to the cis response element in the engrailed enhancer. In relation to that, Pc recruitment is reduced on the salivary gland polytene chromosomes and specifically at the engrailed locus. These results suggest that Tara might be required for positioning Pc to a subset of its target genes.
Collapse
Affiliation(s)
- Pranabananda Dutta
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, New York, United States of America
- * E-mail:
| | - Willis X. Li
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, New York, United States of America
| |
Collapse
|
24
|
Cap-n-Collar Promotes Tissue Regeneration by Regulating ROS and JNK Signaling in the Drosophila melanogaster Wing Imaginal Disc. Genetics 2017; 206:1505-1520. [PMID: 28512185 DOI: 10.1534/genetics.116.196832] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 05/10/2017] [Indexed: 01/03/2023] Open
Abstract
Regeneration is a complex process that requires an organism to recognize and repair tissue damage, as well as grow and pattern new tissue. Here, we describe a genetic screen to identify novel regulators of regeneration. We ablated the Drosophila melanogaster larval wing primordium by inducing apoptosis in a spatially and temporally controlled manner and allowed the tissue to regenerate and repattern. To identify genes that regulate regeneration, we carried out a dominant-modifier screen by assessing the amount and quality of regeneration in adult wings heterozygous for isogenic deficiencies. We have identified 31 regions on the right arm of the third chromosome that modify the regenerative response. Interestingly, we observed several distinct phenotypes: mutants that regenerated poorly, mutants that regenerated faster or better than wild-type, and mutants that regenerated imperfectly and had patterning defects. We mapped one deficiency region to cap-n-collar (cnc), the Drosophila Nrf2 ortholog, which is required for regeneration. Cnc regulates reactive oxygen species levels in the regenerating epithelium, and affects c-Jun N-terminal protein kinase (JNK) signaling, growth, debris localization, and pupariation timing. Here, we present the results of our screen and propose a model wherein Cnc regulates regeneration by maintaining an optimal level of reactive oxygen species to promote JNK signaling.
Collapse
|
25
|
Losada-Perez M, Harrison N, Hidalgo A. Molecular mechanism of central nervous system repair by the Drosophila NG2 homologue kon-tiki. J Cell Biol 2016; 214:587-601. [PMID: 27551055 PMCID: PMC5004445 DOI: 10.1083/jcb.201603054] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 07/25/2016] [Indexed: 11/22/2022] Open
Abstract
Glial cells help central nervous system injury repair, but this is limited by the failure of newly produced glial cells to differentiate. Here, Losada-Perez et al. identify the NG2-dependent mechanism modulating glial proliferation and differentiation after damage to promote repair, in the central nervous system of Drosophila. Neuron glia antigen 2 (NG2)–positive glia are repair cells that proliferate upon central nervous system (CNS) damage, promoting functional recovery. However, repair is limited because of the failure of the newly produced glial cells to differentiate. It is a key goal to discover how to regulate NG2 to enable glial proliferation and differentiation conducive to repair. Drosophila has an NG2 homologue called kon-tiki (kon), of unknown CNS function. We show that kon promotes repair and identify the underlying mechanism. Crush injury up-regulates kon expression downstream of Notch. Kon in turn induces glial proliferation and initiates glial differentiation by activating glial genes and prospero (pros). Two negative feedback loops with Notch and Pros allow Kon to drive the homeostatic regulation required for repair. By modulating Kon levels in glia, we could prevent or promote CNS repair. Thus, the functional links between Kon, Notch, and Pros are essential for, and can drive, repair. Analogous mechanisms could promote CNS repair in mammals.
Collapse
Affiliation(s)
- Maria Losada-Perez
- NeuroDevelopment Lab, School of Biosciences, University of Birmingham, Birmingham B15 2TT, England, UK
| | - Neale Harrison
- NeuroDevelopment Lab, School of Biosciences, University of Birmingham, Birmingham B15 2TT, England, UK
| | - Alicia Hidalgo
- NeuroDevelopment Lab, School of Biosciences, University of Birmingham, Birmingham B15 2TT, England, UK
| |
Collapse
|
26
|
Abstract
Living organisms experience tissue damage from both, the surrounding environment and from inside their bodies. Tissue repair/regeneration is triggered by local tissue injury to restore an injured, or lost, part of the body. Tissue damage results in a series of responses, not only locally but also systemically in distant tissues. In our recent publication, we established a "dual system" that induces spatiotemporal tissue damage simultaneously with gene manipulation in surrounding tissues. With this system, we demonstrated that appropriate regulation of methionine metabolism in the fat body is required for tissue repair in Drosophila wing discs, thus highlighting the importance of systemic damage response (SDR) in tissue repair. This "Extra View" aims to discuss our recent reports that propose methionine metabolism to be an essential part of SDR, together with related topics in several model organisms.
Collapse
Affiliation(s)
- Soshiro Kashio
- a Department of Genetics , Graduate School of Pharmaceutical Sciences, The University of Tokyo , Bunkyo-ku, Tokyo , Japan
| | - Fumiaki Obata
- a Department of Genetics , Graduate School of Pharmaceutical Sciences, The University of Tokyo , Bunkyo-ku, Tokyo , Japan.,b The Francis Crick Institute , The Ridgeway, Mill Hill, London , United Kingdom
| | - Masayuki Miura
- a Department of Genetics , Graduate School of Pharmaceutical Sciences, The University of Tokyo , Bunkyo-ku, Tokyo , Japan.,c Agency for Medical Research and Development-Core Research for Evolutional Medical Science and Technology (AMED-CREST), Japan Agency for Medical Research and Development , Chiyoda-ku, Tokyo , Japan
| |
Collapse
|
27
|
Morata G, Herrera SC. Cell reprogramming during regeneration in Drosophila: transgression of compartment boundaries. Curr Opin Genet Dev 2016; 40:11-16. [PMID: 27266970 DOI: 10.1016/j.gde.2016.05.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 04/11/2016] [Accepted: 05/19/2016] [Indexed: 10/21/2022]
Abstract
We discuss recent work about cellular reprogramming during regeneration of the imaginal discs of Drosophila. These contain various lineage blocks, compartments, which express distinct genetic programmes. It has been found that after massive damage to a compartment cells from a neighbour compartment can transgress the compartment border and contribute to its regeneration. The transgressing cells are genetically reprogrammed and acquire a new identity, a process facilitated by up regulation of the JNK pathway and transient loss of epigenetic control by the Pc-G and trx-G genes. The final acquisition of the new identity appears to be mediated by induction by neighbour cells, a phenomenon akin the Community Effect described for the specification of amphibian muscle cells.
Collapse
Affiliation(s)
- Ginés Morata
- Centro de Biología Molecular, CSIC-UAM, Madrid, Spain
| | | |
Collapse
|
28
|
Abstract
Recent work has strengthened Drosophila imaginal discs as a model system for regeneration studies. Evidence is accumulating that oxidative stress drives the cellular responses for repair and regeneration. Drosophila imaginal discs generate a burst of reactive oxygen species (ROS) upon damage that is necessary for the activation of the Jun N-terminal kinase (JNK) and p38 MAP kinase signaling pathways. Moreover, these pathways are pivotal in the activation of regenerative growth. A hypothetical mechanism of how the ROS are initiated, and how repair and regeneration is activated is discussed here.
Collapse
Affiliation(s)
- Florenci Serras
- a Institute of Biomedicine of the University of Barcelona (IBUB) , Barcelona , Spain.,b Departament de Genètica, Facultat de Biologia , Universitat de Barcelona , Barcelona , Spain
| |
Collapse
|
29
|
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
|
30
|
Khan SJ, Abidi SNF, Tian Y, Skinner A, Smith-Bolton RK. A rapid, gentle and scalable method for dissociation and fluorescent sorting of imaginal disc cells for mRNA sequencing. Fly (Austin) 2016; 10:73-80. [PMID: 27057746 DOI: 10.1080/19336934.2016.1173296] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Dissociation of imaginal disc cells has been carried out previously to enable flow cytometry and cell sorting to analyze cell cycle progression, cell size, gene expression, and other aspects of imaginal tissues. However, the lengthy dissociation protocols employed may alter gene expression, cell behavior and overall viability. Here we describe a new rapid and gentle method of dissociating the cells of wing imaginal discs that significantly enhances cell viability and reduces the likelihood of gene expression changes. Furthermore, this method is scalable, enabling collection of large amounts of sample for high-throughput experiments without the need for data-distorting amplifications.
Collapse
Affiliation(s)
- Sumbul Jawed Khan
- a Department of Cell and Developmental Biology , University of Illinois at Urbana-Champaign , Urbana , IL , USA
| | - Syeda Nayab Fatima Abidi
- a Department of Cell and Developmental Biology , University of Illinois at Urbana-Champaign , Urbana , IL , USA
| | - Yuan Tian
- a Department of Cell and Developmental Biology , University of Illinois at Urbana-Champaign , Urbana , IL , USA
| | - Andrea Skinner
- a Department of Cell and Developmental Biology , University of Illinois at Urbana-Champaign , Urbana , IL , USA
| | - Rachel K Smith-Bolton
- a Department of Cell and Developmental Biology , University of Illinois at Urbana-Champaign , Urbana , IL , USA
| |
Collapse
|
31
|
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
|