1
|
Klemm JW, Van Hazel C, Harris RE. Regeneration following tissue necrosis is mediated by non-apoptotic caspase activity. eLife 2025; 13:RP101114. [PMID: 40042383 PMCID: PMC11882144 DOI: 10.7554/elife.101114] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2025] Open
Abstract
Tissue necrosis is a devastating complication for many human diseases and injuries. Unfortunately, our understanding of necrosis and how it impacts surrounding healthy tissue - an essential consideration when developing effective methods to treat such injuries - has been limited by a lack of robust genetically tractable models. Our lab previously established a method to study necrosis-induced regeneration in the Drosophila wing imaginal disc, which revealed a unique phenomenon whereby cells at a distance from the injury upregulate caspase activity in a process called Necrosis-induced Apoptosis (NiA) that is vital for regeneration. Here, we have further investigated this phenomenon, showing that NiA is predominantly associated with the highly regenerative pouch region of the disc, shaped by genetic factors present in the presumptive hinge. Furthermore, we find that a proportion of NiA fail to undergo apoptosis, instead surviving effector caspase activation to persist within the tissue and stimulate reparative proliferation late in regeneration. This proliferation relies on the initiator caspase Dronc, and occurs independent of JNK, ROS or mitogens associated with the previously characterized Apoptosis-induced Proliferation (AiP) mechanism. These data reveal a new means by which non-apoptotic Dronc signaling promotes regenerative proliferation in response to necrotic damage.
Collapse
|
2
|
Galliot B, Wenger Y. Organizer formation, organizer maintenance and epithelial cell plasticity in Hydra: Role of the Wnt3/β-catenin/TCF/Sp5/Zic4 gene network. Cells Dev 2025:204002. [PMID: 39929422 DOI: 10.1016/j.cdev.2025.204002] [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: 01/12/2025] [Accepted: 02/06/2025] [Indexed: 02/14/2025]
Abstract
The experimental and conceptual knowledge in 1909 led to the discovery of the Hydra head organizer through transplantation experiments between pigmented and non-pigmented animals; a discovery followed by numerous transplantations demonstrating cross-regulation between activating and inhibiting components distributed along the body axis. This experimental work inspired mathematicians, engineers, physicists and computer scientists to develop theoretical models predicting the principles of developmental mechanisms. Today, we know that the Wnt/β-catenin/Sp5/Zic4 gene regulatory network (GRN) links organizer activity, morphogenesis and cellular identity in Hydra, with variable conformations depending on the region or epithelial layer, and varied phenotypes depending on which GRN element is misregulated. In intact animals, Wnt/β-catenin signaling acts as the head activator at the tip of the hypostome, restricted by Sp5 in the other regions of the animal. Moreover, in the tentacle ring, Sp5 and Zic4 act epistatically to support tentacle differentiation and prevent basal disc differentiation. Along the body column, Sp5 is self-repressed in the epidermis and acts as a head inhibitor along the gastrodermis. Other players modulate these activities, such as TSP and Margin/RAX apically, Notch signaling in the tentacle zone, Dkk1/2/4 and HAS-7 in the body column. In the developmental context of regeneration, cells below the amputation zone switch from repressed to locally de novo activated head organizer status, a transition driven by immediate symmetrical and asymmetrical metabolic changes that lead to gene expression regulations involving components and modulators of Wnt/β-catenin signaling, early-pulse and early-late transient both often symmetrical, together with sustained ones, specific to head regeneration.
Collapse
Affiliation(s)
- Brigitte Galliot
- Department of Genetics and Evolution, University of Geneva, Geneva, Switzerland.
| | - Yvan Wenger
- Department of Genetics and Evolution, University of Geneva, Geneva, Switzerland
| |
Collapse
|
3
|
Nagai S, Otaki JM. Wound Healing in Butterfly Pupal Wing Tissues: Real-Time In Vivo Imaging of Long-Range Cell Migration, Cluster Formation, and Calcium Oscillations. INSECTS 2025; 16:124. [PMID: 40003754 PMCID: PMC11856899 DOI: 10.3390/insects16020124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 01/21/2025] [Accepted: 01/24/2025] [Indexed: 02/27/2025]
Abstract
Insects can repair wounds and regenerate body parts in response to physical damage. Wound healing in butterfly pupal wing tissues is developmentally interesting because ectopic color patterns develop during healing, suggesting that normal and damage-induced color patterns may use similar mechanisms. Here we physiologically investigated wound healing and ectopic color pattern formation in butterfly pupal wing tissues using the blue pansy butterfly Junonia orithya. In response to physical puncture damage, various ectopic color patterns are formed around the damage site. After the wounding operation, we observed hemocytes migrating over long distances along the wing veins (lacunae) toward the damage site, where hemocytes and epidermal cells formed cellular clusters. Calcium oscillations were observed in cells at and near the damage site. Calcium oscillations were transiently affected by ruthenium red, an inhibitor of calcium transporters and channels, and ruthenium red caused various abnormalities in the scales of adult wings. These results suggest that cell migration, cluster formation, and calcium oscillations play important roles in wound healing and scale development at and near the damage site. Ectopic color patterns may develop in response to local calcium oscillations as a consequence of the evolutionary co-option of the healing process for normal development.
Collapse
Affiliation(s)
- Shuka Nagai
- The BCPH Unit of Molecular Physiology, Department of Chemistry, Biology and Marine Science, Faculty of Science, University of the Ryukyus, Nishihara, Okinawa 903-0213, Japan
- Department of Molecular Neuroscience, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Joji M. Otaki
- The BCPH Unit of Molecular Physiology, Department of Chemistry, Biology and Marine Science, Faculty of Science, University of the Ryukyus, Nishihara, Okinawa 903-0213, Japan
| |
Collapse
|
4
|
Klemm JW, Van Hazel C, Harris RE. Regeneration following tissue necrosis is mediated by non-apoptotic caspase activity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.26.605350. [PMID: 39091851 PMCID: PMC11291143 DOI: 10.1101/2024.07.26.605350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Tissue necrosis is a devastating complication for many human diseases and injuries. Unfortunately, our understanding of necrosis and how it impacts surrounding healthy tissue - an essential consideration when developing effective methods to treat such injuries - has been limited by a lack of robust genetically tractable models. Our lab previously established a method to study necrosis-induced regeneration in the Drosophila wing imaginal disc, which revealed a unique phenomenon whereby cells at a distance from the injury upregulate caspase activity in a process called Necrosis-induced Apoptosis (NiA) that is vital for regeneration. Here we have further investigated this phenomenon, showing that NiA is predominantly associated with the highly regenerative pouch region of the disc, shaped by genetic factors present in the presumptive hinge. Furthermore, we find that a proportion of NiA fail to undergo apoptosis, instead surviving effector caspase activation to persist within the tissue and stimulate reparative proliferation late in regeneration. This proliferation relies on the initiator caspase Dronc, and occurs independent of JNK, ROS or mitogens associated with the previously characterized Apoptosis-induced Proliferation (AiP) mechanism. These data reveal a new means by which non-apoptotic Dronc signaling promotes regenerative proliferation in response to necrotic damage.
Collapse
Affiliation(s)
- Jacob W Klemm
- Arizona State University, 427 E Tyler Mall LSE 229, Tempe, AZ 85287-4501
| | - Chloe Van Hazel
- Arizona State University, 427 E Tyler Mall LSE 229, Tempe, AZ 85287-4501
| | - Robin E Harris
- Arizona State University, 427 E Tyler Mall LSE 229, Tempe, AZ 85287-4501
| |
Collapse
|
5
|
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
|
6
|
Camilleri-Robles C, Amador R, Tiebe M, Teleman A, Serras F, Guigó R, Corominas M. Long non-coding RNAs involved in Drosophila development and regeneration. NAR Genom Bioinform 2024; 6:lqae091. [PMID: 39157585 PMCID: PMC11327875 DOI: 10.1093/nargab/lqae091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 07/12/2024] [Accepted: 07/19/2024] [Indexed: 08/20/2024] Open
Abstract
The discovery of functional long non-coding RNAs (lncRNAs) changed their initial concept as transcriptional noise. LncRNAs have been identified as regulators of multiple biological processes, including chromatin structure, gene expression, splicing, mRNA degradation, and translation. However, functional studies of lncRNAs are hindered by the usual lack of phenotypes upon deletion or inhibition. Here, we used Drosophila imaginal discs as a model system to identify lncRNAs involved in development and regeneration. We examined a subset of lncRNAs expressed in the wing, leg, and eye disc development. Additionally, we analyzed transcriptomic data from regenerating wing discs to profile the expression pattern of lncRNAs during tissue repair. We focused on the lncRNA CR40469, which is upregulated during regeneration. We generated CR40469 mutant flies that developed normally but showed impaired wing regeneration upon cell death induction. The ability of these mutants to regenerate was restored by the ectopic expression of CR40469. Furthermore, we found that the lncRNA CR34335 has a high degree of sequence similarity with CR40469 and can partially compensate for its function during regeneration in the absence of CR40469. Our findings point to a potential role of the lncRNA CR40469 in trans during the response to damage in the wing imaginal disc.
Collapse
Affiliation(s)
- Carlos Camilleri-Robles
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia and Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Raziel Amador
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia and Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Catalonia, Spain
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology (BIST), Barcelona, Catalonia, Spain
| | - Marcel Tiebe
- German Cancer Research Center (DKFZ) Heidelberg, Division B140, 69120 Heidelberg, Germany
| | - Aurelio A Teleman
- German Cancer Research Center (DKFZ) Heidelberg, Division B140, 69120 Heidelberg, Germany
| | - Florenci Serras
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia and Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Roderic Guigó
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology (BIST), Barcelona, Catalonia, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Catalonia, Spain
| | - Montserrat Corominas
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia and Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Catalonia, Spain
| |
Collapse
|
7
|
Nagai H, Adachi Y, Nakasugi T, Takigawa E, Ui J, Makino T, Miura M, Nakajima YI. Highly regenerative species-specific genes improve age-associated features in the adult Drosophila midgut. BMC Biol 2024; 22:157. [PMID: 39090637 PMCID: PMC11295675 DOI: 10.1186/s12915-024-01956-4] [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: 07/06/2023] [Accepted: 07/09/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND The remarkable regenerative abilities observed in planarians and cnidarians are closely linked to the active proliferation of adult stem cells and the precise differentiation of their progeny, both of which typically deteriorate during aging in low regenerative animals. While regeneration-specific genes conserved in highly regenerative organisms may confer regenerative abilities and long-term maintenance of tissue homeostasis, it remains unclear whether introducing these regenerative genes into low regenerative animals can improve their regeneration and aging processes. RESULTS Here, we ectopically express highly regenerative species-specific JmjC domain-encoding genes (HRJDs) in Drosophila, a widely used low regenerative model organism. Surprisingly, HRJD expression impedes tissue regeneration in the developing wing disc but extends organismal lifespan when expressed in the intestinal stem cell lineages of the adult midgut under non-regenerative conditions. Notably, HRJDs enhance the proliferative activity of intestinal stem cells while maintaining their differentiation fidelity, ameliorating age-related decline in gut barrier functions. CONCLUSIONS These findings together suggest that the introduction of highly regenerative species-specific genes can improve stem cell functions and promote a healthy lifespan when expressed in aging animals.
Collapse
Affiliation(s)
- Hiroki Nagai
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-0033, Japan
- Institute of Science and Technology Austria, Am Campus 1, 3400, Klosterneuburg, Austria
| | - Yuya Adachi
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-0033, Japan
| | - Tenki Nakasugi
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-0033, Japan
| | - Ema Takigawa
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-0033, Japan
| | - Junichiro Ui
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-0033, Japan
| | - Takashi Makino
- Graduate School of Life Sciences, Tohoku University, 6-3 Aramaki Aza Aoba, Aoba-Ku, Sendai, 980-8578, Japan
| | - Masayuki Miura
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-0033, Japan
| | - Yu-Ichiro Nakajima
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-0033, Japan.
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
Seifert AW, Duncan EM, Zayas RM. Enduring questions in regenerative biology and the search for answers. Commun Biol 2023; 6:1139. [PMID: 37945686 PMCID: PMC10636051 DOI: 10.1038/s42003-023-05505-7] [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: 06/09/2023] [Accepted: 10/25/2023] [Indexed: 11/12/2023] Open
Abstract
The potential for basic research to uncover the inner workings of regenerative processes and produce meaningful medical therapies has inspired scientists, clinicians, and patients for hundreds of years. Decades of studies using a handful of highly regenerative model organisms have significantly advanced our knowledge of key cell types and molecular pathways involved in regeneration. However, many questions remain about how regenerative processes unfold in regeneration-competent species, how they are curtailed in non-regenerative organisms, and how they might be induced (or restored) in humans. Recent technological advances in genomics, molecular biology, computer science, bioengineering, and stem cell research hold promise to collectively provide new experimental evidence for how different organisms accomplish the process of regeneration. In theory, this new evidence should inform the design of new clinical approaches for regenerative medicine. A deeper understanding of how tissues and organs regenerate will also undoubtedly impact many adjacent scientific fields. To best apply and adapt these new technologies in ways that break long-standing barriers and answer critical questions about regeneration, we must combine the deep knowledge of developmental and evolutionary biologists with the hard-earned expertise of scientists in mechanistic and technical fields. To this end, this perspective is based on conversations from a workshop we organized at the Banbury Center, during which a diverse cross-section of the regeneration research community and experts in various technologies discussed enduring questions in regenerative biology. Here, we share the questions this group identified as significant and unanswered, i.e., known unknowns. We also describe the obstacles limiting our progress in answering these questions and how expanding the number and diversity of organisms used in regeneration research is essential for deepening our understanding of regenerative capacity. Finally, we propose that investigating these problems collaboratively across a diverse network of researchers has the potential to advance our field and produce unexpected insights into important questions in related areas of biology and medicine.
Collapse
Affiliation(s)
- Ashley W Seifert
- Department of Biology, University of Kentucky, Lexington, KY, 40506, USA.
| | - Elizabeth M Duncan
- Department of Biology, University of Kentucky, Lexington, KY, 40506, USA.
| | - Ricardo M Zayas
- Department of Biology, San Diego State University, San Diego, CA, 92182, USA.
| |
Collapse
|
11
|
Harris RE. Investigating Tissue Regeneration Using the DUAL Control Genetic Ablation System. Methods Mol Biol 2023; 2599:255-270. [PMID: 36427155 DOI: 10.1007/978-1-0716-2847-8_18] [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] [Indexed: 06/14/2023]
Abstract
Genetic ablation is a highly efficient method to study regeneration in vivo by stimulating tissue-specific cell death that subsequently induces regrowth and repair in a developing organism. This approach has been particularly successful in Drosophila, for which various temperature-based genetic ablation tools have been developed to explore the complexities of regeneration in larval imaginal discs. Here, we describe the use of a recently established ablation system called DUAL Control, which can be used to both characterize the damage response and genetically manipulate blastema cells to identify novel regulators of regeneration.
Collapse
Affiliation(s)
- R E Harris
- School of Life Sciences, Arizona State University, Tempe, AZ, USA.
| |
Collapse
|
12
|
Crucianelli C, Jaiswal J, Vijayakumar Maya A, Nogay L, Cosolo A, Grass I, Classen AK. Distinct signaling signatures drive compensatory proliferation via S-phase acceleration. PLoS Genet 2022; 18:e1010516. [PMID: 36520882 PMCID: PMC9799308 DOI: 10.1371/journal.pgen.1010516] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 12/29/2022] [Accepted: 11/08/2022] [Indexed: 12/23/2022] Open
Abstract
Regeneration relies on cell proliferation to restore damaged tissues. Multiple signaling pathways activated by local or paracrine cues have been identified to promote regenerative proliferation. How different types of tissue damage may activate distinct signaling pathways and how these differences converge on regenerative proliferation is less well defined. To better understand how tissue damage and proliferative signals are integrated during regeneration, we investigate models of compensatory proliferation in Drosophila imaginal discs. We find that compensatory proliferation is associated with a unique cell cycle profile, which is characterized by short G1 and G2 phases and, surprisingly, by acceleration of the S-phase. S-phase acceleration can be induced by two distinct signaling signatures, aligning with inflammatory and non-inflammatory tissue damage. Specifically, non-autonomous activation of JAK/STAT and Myc in response to inflammatory damage, or local activation of Ras/ERK and Hippo/Yki in response to elevated cell death, promote accelerated nucleotide incorporation during S-phase. This previously unappreciated convergence of different damaging insults on the same regenerative cell cycle program reconciles previous conflicting observations on proliferative signaling in different tissue regeneration and tumor models.
Collapse
Affiliation(s)
- Carlo Crucianelli
- Hilde-Mangold-Haus, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Janhvi Jaiswal
- Hilde-Mangold-Haus, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
| | - Ananthakrishnan Vijayakumar Maya
- Hilde-Mangold-Haus, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- International Max Planck Research School for Immunobiology, Epigenetics, and Metabolism, Freiburg, Germany
| | - Liyne Nogay
- Hilde-Mangold-Haus, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- International Max Planck Research School for Immunobiology, Epigenetics, and Metabolism, Freiburg, Germany
| | - Andrea Cosolo
- Hilde-Mangold-Haus, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Isabelle Grass
- Hilde-Mangold-Haus, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Anne-Kathrin Classen
- Hilde-Mangold-Haus, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany
- CIBSS Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| |
Collapse
|
13
|
Worley MI, Hariharan IK. Imaginal Disc Regeneration: Something Old, Something New. Cold Spring Harb Perspect Biol 2022; 14:a040733. [PMID: 34872971 PMCID: PMC9620854 DOI: 10.1101/cshperspect.a040733] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Imaginal discs are simple epithelial sacs found in Drosophila larvae, which generate adult structures including wings and legs. The first studies of imaginal disc regeneration involved technically challenging transplantation experiments. Yet despite the difficulty, many aspects of regeneration including wound healing, blastema formation, and the repatterning of regenerated tissue were characterized. An important discovery was the phenomenon of transdetermination, where a small group of cells in regenerating tissue collectively switch fate ("collective cell reprogramming"). The development of genetic tissue-ablation systems over the last 12 years has energized this field, by making experiments less technically challenging, more reproducible, and by incorporating additional genetic analysis. Recent progress includes defining mechanistic links between early responses to wounding and the signaling pathways that drive proliferation, uncovering a role for localized silencing of damage-responsive enhancers to limit regenerative capacity as tissues mature, and identifying genes that maintain cellular plasticity within acceptable limits during regeneration.
Collapse
Affiliation(s)
- Melanie I Worley
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720-3200, USA
| | - Iswar K Hariharan
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720-3200, USA
| |
Collapse
|
14
|
Serras F. The sooner, the better: ROS, kinases and nutrients at the onset of the damage response in Drosophila. Front Cell Dev Biol 2022; 10:1047823. [PMID: 36353511 PMCID: PMC9637634 DOI: 10.3389/fcell.2022.1047823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 10/11/2022] [Indexed: 11/20/2022] Open
Abstract
One of the main topics in regeneration biology is the nature of the early signals that trigger the damage response. Recent advances in Drosophila point to the MAP3 kinase Ask1 as a molecular hub that integrates several signals at the onset of regeneration. It has been discovered that reactive oxygen species (ROS) produced in damaged imaginal discs and gut epithelia will activate the MAP3 kinase Ask1. Severely damaged and apoptotic cells produce an enormous amount of ROS, which ensures their elimination by activating Ask1 and in turn the pro-apoptotic function of JNK. However, this creates an oxidative stress environment with beneficial effects that is sensed by neighboring healthy cells. This environment, in addition to the Pi3K/Akt nutrient sensing pathway, can be integrated into Ask1 to launch regeneration. Ultimately the activity of Ask1 depends on these and other inputs and modulates its signaling to achieve moderate levels of p38 and low JNK signaling and thus promote survival and regeneration. This model based on the dual function of Ask1 for early response to damage is discussed here.
Collapse
Affiliation(s)
- Florenci Serras
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, and Institute of Biomedicine of the University of Barcelona, University of Barcelona, Barcelona, Spain
| |
Collapse
|
15
|
Avalos PN, Forsthoefel DJ. An Emerging Frontier in Intercellular Communication: Extracellular Vesicles in Regeneration. Front Cell Dev Biol 2022; 10:849905. [PMID: 35646926 PMCID: PMC9130466 DOI: 10.3389/fcell.2022.849905] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 03/28/2022] [Indexed: 12/12/2022] Open
Abstract
Regeneration requires cellular proliferation, differentiation, and other processes that are regulated by secreted cues originating from cells in the local environment. Recent studies suggest that signaling by extracellular vesicles (EVs), another mode of paracrine communication, may also play a significant role in coordinating cellular behaviors during regeneration. EVs are nanoparticles composed of a lipid bilayer enclosing proteins, nucleic acids, lipids, and other metabolites, and are secreted by most cell types. Upon EV uptake by target cells, EV cargo can influence diverse cellular behaviors during regeneration, including cell survival, immune responses, extracellular matrix remodeling, proliferation, migration, and differentiation. In this review, we briefly introduce the history of EV research and EV biogenesis. Then, we review current understanding of how EVs regulate cellular behaviors during regeneration derived from numerous studies of stem cell-derived EVs in mammalian injury models. Finally, we discuss the potential of other established and emerging research organisms to expand our mechanistic knowledge of basic EV biology, how injury modulates EV biogenesis, cellular sources of EVs in vivo, and the roles of EVs in organisms with greater regenerative capacity.
Collapse
Affiliation(s)
- Priscilla N. Avalos
- Department of Cell Biology, College of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - David J. Forsthoefel
- Department of Cell Biology, College of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| |
Collapse
|
16
|
Kumar N, Huizar FJ, Farfán-Pira KJ, Brodskiy PA, Soundarrajan DK, Nahmad M, Zartman JJ. MAPPER: An Open-Source, High-Dimensional Image Analysis Pipeline Unmasks Differential Regulation of Drosophila Wing Features. Front Genet 2022; 13:869719. [PMID: 35480325 PMCID: PMC9035675 DOI: 10.3389/fgene.2022.869719] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 03/03/2022] [Indexed: 11/13/2022] Open
Abstract
Phenomics requires quantification of large volumes of image data, necessitating high throughput image processing approaches. Existing image processing pipelines for Drosophila wings, a powerful genetic model for studying the underlying genetics for a broad range of cellular and developmental processes, are limited in speed, precision, and functional versatility. To expand on the utility of the wing as a phenotypic screening system, we developed MAPPER, an automated machine learning-based pipeline that quantifies high-dimensional phenotypic signatures, with each dimension quantifying a unique morphological feature of the Drosophila wing. MAPPER magnifies the power of Drosophila phenomics by rapidly quantifying subtle phenotypic differences in sample populations. We benchmarked MAPPER's accuracy and precision in replicating manual measurements to demonstrate its widespread utility. The morphological features extracted using MAPPER reveal variable sexual dimorphism across Drosophila species and unique underlying sex-specific differences in morphogen signaling in male and female wings. Moreover, the length of the proximal-distal axis across the species and sexes shows a conserved scaling relationship with respect to the wing size. In sum, MAPPER is an open-source tool for rapid, high-dimensional analysis of large imaging datasets. These high-content phenomic capabilities enable rigorous and systematic identification of genotype-to-phenotype relationships in a broad range of screening and drug testing applications and amplify the potential power of multimodal genomic approaches.
Collapse
Affiliation(s)
- Nilay Kumar
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, United States
| | - Francisco J. Huizar
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, United States
| | - Keity J. Farfán-Pira
- Department of Physiology, Biophysics, and Neurosciences, Center for Research and Advanced Studies of the National Polytechnical Institute (Cinvestav), Mexico City, Mexico
| | - Pavel A. Brodskiy
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, United States
| | - Dharsan K. Soundarrajan
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, United States
| | - Marcos Nahmad
- Department of Physiology, Biophysics, and Neurosciences, Center for Research and Advanced Studies of the National Polytechnical Institute (Cinvestav), Mexico City, Mexico
| | - Jeremiah J. Zartman
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, United States
| |
Collapse
|
17
|
Paci G, Mao Y. Forced into shape: Mechanical forces in Drosophila development and homeostasis. Semin Cell Dev Biol 2021; 120:160-170. [PMID: 34092509 PMCID: PMC8681862 DOI: 10.1016/j.semcdb.2021.05.026] [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: 03/04/2021] [Revised: 05/19/2021] [Accepted: 05/20/2021] [Indexed: 12/03/2022]
Abstract
Mechanical forces play a central role in shaping tissues during development and maintaining epithelial integrity in homeostasis. In this review, we discuss the roles of mechanical forces in Drosophila development and homeostasis, starting from the interplay of mechanics with cell growth and division. We then discuss several examples of morphogenetic processes where complex 3D structures are shaped by mechanical forces, followed by a closer look at patterning processes. We also review the role of forces in homeostatic processes, including cell elimination and wound healing. Finally, we look at the interplay of mechanics and developmental robustness and discuss open questions in the field, as well as novel approaches that will help tackle them in the future.
Collapse
Affiliation(s)
- Giulia Paci
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK; Institute for the Physics of Living Systems, University College London, Gower Street, London WC1E 6BT, UK
| | - Yanlan Mao
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK; Institute for the Physics of Living Systems, University College London, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
18
|
Cao X, Rojas M, Pastor-Pareja JC. Intrinsic and damage-induced JAK/STAT signaling regulate developmental timing by the Drosophila prothoracic gland. Dis Model Mech 2021; 15:273570. [PMID: 34842272 PMCID: PMC8807578 DOI: 10.1242/dmm.049160] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 11/15/2021] [Indexed: 11/20/2022] Open
Abstract
Development involves tightly paced, reproducible sequences of events, yet it must adjust to conditions external to it, such as resource availability and organismal damage. A major mediator of damage-induced immune responses in vertebrates and insects is JAK/STAT signaling. At the same time, JAK/STAT activation by the Drosophila Upd cytokines is pleiotropically involved in normal development of multiple organs. Whether inflammatory and developmental JAK/STAT roles intersect is unknown. Here, we show that JAK/STAT is active during development of the prothoracic gland (PG), which controls metamorphosis onset through ecdysone production. Reducing JAK/STAT signaling decreased PG size and advanced metamorphosis. Conversely, JAK/STAT hyperactivation by overexpression of pathway components or SUMOylation loss caused PG hypertrophy and metamorphosis delay. Tissue damage and tumors, known to secrete Upd cytokines, also activated JAK/STAT in the PG and delayed metamorphosis, at least in part by inducing expression of the JAK/STAT target Apontic. JAK/STAT damage signaling, therefore, regulates metamorphosis onset by co-opting its developmental role in the PG. Our findings in Drosophila provide insights on how systemic effects of damage and cancer can interfere with hormonally controlled development and developmental transitions. Summary: Damage signaling from tumors mediated by JAK/STAT-activating Upd cytokines delays the Drosophila larva–pupa transition through co-option of a JAK/STAT developmental role in the prothoracic gland.
Collapse
Affiliation(s)
- Xueya Cao
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Marta Rojas
- School of Medicine, Tsinghua University, Beijing, China
| | - José Carlos Pastor-Pareja
- School of Life Sciences, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Beijing, China
| |
Collapse
|
19
|
Klemm J, Stinchfield MJ, Harris RE. Necrosis-induced apoptosis promotes regeneration in Drosophila wing imaginal discs. Genetics 2021; 219:6365941. [PMID: 34740246 PMCID: PMC8570793 DOI: 10.1093/genetics/iyab144] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 08/26/2021] [Indexed: 01/13/2023] Open
Abstract
Regeneration is a complex process that requires a coordinated genetic response to tissue loss. Signals from dying cells are crucial to this process and are best understood in the context of regeneration following programmed cell death, like apoptosis. Conversely, regeneration following unregulated forms of death, such as necrosis, have yet to be fully explored. Here, we have developed a method to investigate regeneration following necrosis using the Drosophila wing imaginal disc. We show that necrosis stimulates regeneration at an equivalent level to that of apoptosis-mediated cell death and activates a similar response at the wound edge involving localized JNK signaling. Unexpectedly, however, necrosis also results in significant apoptosis far from the site of ablation, which we have termed necrosis-induced apoptosis (NiA). This apoptosis occurs independent of changes at the wound edge and importantly does not rely on JNK signaling. Furthermore, we find that blocking NiA limits proliferation and subsequently inhibits regeneration, suggesting that tissues damaged by necrosis can activate programmed cell death at a distance from the injury to promote regeneration.
Collapse
Affiliation(s)
- Jacob Klemm
- School of Life Sciences, Arizona State University, Tempe, AZ 85728, USA
| | | | - Robin E Harris
- School of Life Sciences, Arizona State University, Tempe, AZ 85728, USA
| |
Collapse
|
20
|
He D, Yan L, Zhang J, Li F, Wu Y, Su L, Chen P, Wu M, Choi J, Tong H. Sargassum fusiforme polysaccharide attenuates high-sugar-induced lipid accumulation in HepG2 cells and Drosophila melanogaster larvae. Food Sci Nutr 2021; 9:5590-5599. [PMID: 34646529 PMCID: PMC8498055 DOI: 10.1002/fsn3.2521] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 07/31/2021] [Accepted: 08/03/2021] [Indexed: 12/22/2022] Open
Abstract
Lipid accumulation is a major factor in the development of non-alcoholic fatty liver disease (NAFLD). Currently, there is a lack of intervention or therapeutic drugs against NAFLD. In this study, we investigated the ability of Sargassum fusiforme polysaccharide (SFPS) to reduce lipid accumulation induced by high sugar in HepG2 cells and Drosophila melanogaster larvae. The results indicated that SFPS significantly (p < .01) decreased the accumulation of lipid droplets in high sugar-induced HepG2 cells. Furthermore, SFPS also suppressed the expression of Srebp and Fas (genes involved in lipogenesis) and increased the expression of PPARɑ and Cpt1 (genes that participated in fatty acid β-oxidation) in these cells. SFPS markedly reduced the content of triglyceride of the third instar larvae developed from D. melanogaster eggs reared on the high-sucrose diet. The expression of the Srebp and Fas genes in the larvae was also inhibited whereas the expression of two genes involved in the β-oxidation of fatty acids, Acox57D-d and Fabp, was increased in the larval fat body (a functional homolog of the human liver). We also found that SFPS ameliorated developmental abnormalities induced by the high-sucrose diet. These results of this study suggest that SFPS could potentially be used as a therapeutic agent for the prevention and treatment of NAFLD.
Collapse
Affiliation(s)
- Dan He
- College of Life and Environmental ScienceWenzhou UniversityWenzhouChina
- Department of Biotechnology and BioengineeringChonnam National UniversityGwangjuSouth Korea
| | - Liping Yan
- College of Life and Environmental ScienceWenzhou UniversityWenzhouChina
| | - Jiaqi Zhang
- College of Life and Environmental ScienceWenzhou UniversityWenzhouChina
| | - Fang Li
- College of Life and Environmental ScienceWenzhou UniversityWenzhouChina
| | - Yu Wu
- College of Life and Environmental ScienceWenzhou UniversityWenzhouChina
| | - Laijin Su
- College of Life and Environmental ScienceWenzhou UniversityWenzhouChina
| | - Peichao Chen
- College of Life and Environmental ScienceWenzhou UniversityWenzhouChina
| | - Mingjiang Wu
- College of Life and Environmental ScienceWenzhou UniversityWenzhouChina
| | - Jong‐il Choi
- Department of Biotechnology and BioengineeringChonnam National UniversityGwangjuSouth Korea
| | - Haibin Tong
- College of Life and Environmental ScienceWenzhou UniversityWenzhouChina
| |
Collapse
|
21
|
Gavory G, Baril C, Laberge G, Bidla G, Koonpaew S, Sonea T, Sauvageau G, Therrien M. A genetic screen in Drosophila uncovers the multifaceted properties of the NUP98-HOXA9 oncogene. PLoS Genet 2021; 17:e1009730. [PMID: 34383740 PMCID: PMC8384169 DOI: 10.1371/journal.pgen.1009730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 08/24/2021] [Accepted: 07/20/2021] [Indexed: 11/19/2022] Open
Abstract
Acute myeloid leukemia (AML) underlies the uncontrolled accumulation of immature myeloid blasts. Several cytogenetic abnormalities have been associated with AML. Among these is the NUP98-HOXA9 (NA9) translocation that fuses the Phe-Gly repeats of nucleoporin NUP98 to the homeodomain of the transcription factor HOXA9. The mechanisms enabling NA9-induced leukemia are poorly understood. Here, we conducted a genetic screen in Drosophila for modifiers of NA9. The screen uncovered 29 complementation groups, including genes with mammalian homologs known to impinge on NA9 activity. Markedly, the modifiers encompassed a diversity of functional categories, suggesting that NA9 perturbs multiple intracellular events. Unexpectedly, we discovered that NA9 promotes cell fate transdetermination and that this phenomenon is greatly influenced by NA9 modifiers involved in epigenetic regulation. Together, our work reveals a network of genes functionally connected to NA9 that not only provides insights into its mechanism of action, but also represents potential therapeutic targets. Acute myeloid leukemia or AML is a cancer of blood cells. Despite significant progress in recent years, a majority of afflicted individuals still succumbs to the disease. A variety of genetic defects have been associated to AML. Among these are chromosomal translocations, which entail the fusion of two genes, leading to the production of cancer-inducing chimeric proteins. A representative example is the NUP98-HOXA9 oncoprotein, which results from the fusion of the NUP98 and HOXA9 genes. The mechanism of action of NUP98-HOXA9 remains poorly understood. Given the evolutionarily conservation of NUP98 and HOXA9 as well as basic cellular processes across multicellular organisms, we took advantage of Drosophila fruit flies as a genetic tool to identify genes that impinge on the activity of human NUP98-HOXA9. Surprisingly, this approach identified a relatively large spectrum of conserved genes that engaged in functional interplay with NUP98-HOXA9, which indicated the pervasive effects that this oncogene has on basic cellular events. While some genes have been previously linked to NUP98-HOXA9, thus validating our experimental approach, several others are novel and as such represent potentially new avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Gwenaëlle Gavory
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
| | - Caroline Baril
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
| | - Gino Laberge
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
| | - Gawa Bidla
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
| | - Surapong Koonpaew
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
| | - Thomas Sonea
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
| | - Guy Sauvageau
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada.,Département de médecine, Université de Montréal, Montréal, Canada
| | - Marc Therrien
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada.,Département de pathologie et de biologie cellulaire, Université de Montréal, Montréal, Canada
| |
Collapse
|
22
|
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: 26] [Impact Index Per Article: 6.5] [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
|
23
|
Cohen E, Peterson NG, Sawyer JK, Fox DT. Accelerated cell cycles enable organ regeneration under developmental time constraints in the Drosophila hindgut. Dev Cell 2021; 56:2059-2072.e3. [PMID: 34019841 PMCID: PMC8319103 DOI: 10.1016/j.devcel.2021.04.029] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/10/2021] [Accepted: 04/28/2021] [Indexed: 12/22/2022]
Abstract
Individual organ development must be temporally coordinated with development of the rest of the organism. As a result, cell division cycles in a developing organ occur on a relatively fixed timescale. Despite this, many developing organs can regenerate cells lost to injury. How organs regenerate within the time constraints of organism development remains unclear. Here, we show that the developing Drosophila hindgut regenerates by accelerating the mitotic cell cycle. This process is achieved by decreasing G1 length and requires the JAK/STAT ligand unpaired-3. Mitotic capacity is then terminated by the steroid hormone ecdysone receptor and the Sox transcription factor Dichaete. These two factors converge on regulation of a hindgut-specific enhancer of fizzy-related, a negative regulator of mitotic cyclins. Our findings reveal how the cell-cycle machinery and cytokine signaling can be adapted to accomplish developmental organ regeneration.
Collapse
Affiliation(s)
- Erez Cohen
- Department of Cell Biology, Duke University School of Medicine, Durham, USA
| | - Nora G Peterson
- Department of Cell Biology, Duke University School of Medicine, Durham, USA
| | - Jessica K Sawyer
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, USA
| | - Donald T Fox
- Department of Cell Biology, Duke University School of Medicine, Durham, USA; Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, USA; Regeneration Next Initiative, Duke University School of Medicine, Durham, USA.
| |
Collapse
|
24
|
Herrera SC, Bach EA. The Emerging Roles of JNK Signaling in Drosophila Stem Cell Homeostasis. Int J Mol Sci 2021; 22:ijms22115519. [PMID: 34073743 PMCID: PMC8197226 DOI: 10.3390/ijms22115519] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/20/2021] [Accepted: 05/21/2021] [Indexed: 12/11/2022] Open
Abstract
The Jun N-terminal kinase (JNK) pathway is an evolutionary conserved kinase cascade best known for its roles during stress-induced apoptosis and tumor progression. Recent findings, however, have identified new roles for this pleiotropic pathway in stem cells during regenerative responses and in cellular plasticity. Here, we provide an overview of recent findings about the new roles of JNK signaling in stem cell biology using two well-established Drosophila models: the testis and the intestine. We highlight the pathway’s roles in processes such as proliferation, death, self-renewal and reprogramming, and discuss the known parallels between flies and mammals.
Collapse
Affiliation(s)
- Salvador C. Herrera
- Centro Andaluz de Biología del Desarrollo, CSIC/Universidad Pablo de Olavide/JA, Carretera de Utrera km 1, 41018 Sevilla, Spain
- Correspondence: (S.C.H.); (E.A.B.)
| | - Erika A. Bach
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, USA
- Helen L. and Martin S. Kimmel Center for Stem Cell Biology, New York University Grossman School of Medicine, New York, NY 10016, USA
- Correspondence: (S.C.H.); (E.A.B.)
| |
Collapse
|
25
|
Esteban-Collado J, Corominas M, Serras F. Nutrition and PI3K/Akt signaling are required for p38-dependent regeneration. Development 2021; 148:258580. [PMID: 33913483 DOI: 10.1242/dev.197087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 03/29/2021] [Indexed: 12/22/2022]
Abstract
Regeneration after damage requires early signals to trigger the tissue repair machinery. Reactive oxygen species (ROS) act as early signals that are sensed by the MAP3 kinase Ask1, which in turn activates by phosphorylation the MAP kinases p38 and JNK. The sustained or high activation of these kinases can result in apoptosis, whereas short or low activation can promote regeneration. Using the Ask1-dependent regeneration program, we demonstrate in Drosophila wing that PI3K/Akt signaling is necessary for Ask1 to activate p38, but not JNK. In addition, nutrient restriction or mutations that target Ser83 of the Drosophila Ask1 protein, a PI3K/Akt-sensitive residue, block regeneration. However, these effects can be reversed by the ectopic activation of p38, but not of JNK. Our results demonstrate that Ask1 controls the activation of p38 through Ser83, and that the phosphorylation of p38 during regeneration is nutrient sensitive. This mechanism is important for discriminating between p38 and JNK in the cells involved in tissue repair and regenerative growth.
Collapse
Affiliation(s)
- José Esteban-Collado
- Department of Genetics, Microbiology and Statistics, School of Biology, University of Barcelona and Institute of Biomedicine of the University of Barcelona (IBUB), Diagonal 643, 08028 Barcelona, Spain
| | - Montserrat Corominas
- Department of Genetics, Microbiology and Statistics, School of Biology, University of Barcelona and Institute of Biomedicine of the University of Barcelona (IBUB), Diagonal 643, 08028 Barcelona, Spain
| | - Florenci Serras
- Department of Genetics, Microbiology and Statistics, School of Biology, University of Barcelona and Institute of Biomedicine of the University of Barcelona (IBUB), Diagonal 643, 08028 Barcelona, Spain
| |
Collapse
|
26
|
Xu Y, Wei W, Lin G, Yan S, Zhang J, Shen J, Wang D. The Ras/MAPK pathway is required for regenerative growth of wing discs in the black cutworm Agrotis ypsilon. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2021; 131:103552. [PMID: 33577967 DOI: 10.1016/j.ibmb.2021.103552] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 02/01/2021] [Accepted: 02/02/2021] [Indexed: 06/12/2023]
Abstract
Regeneration is a common phenomenon in various organisms by which tissues restore the damaged or naturally detached parts. In insects, appendage regeneration takes place during the embryonic, larval and pupal stages for individual survival. The wing disc of black cutworm Agrotis ypsilon has the capacity of regeneration after ablation, but understanding of molecular mechanisms in wing disc regeneration is still limited. After ablation of partial or whole wing discs before the fifth instar larval stage, the adult wings appeared to be normal. In the last two larval stages, ablation of the left wing disc led to smaller corresponding adult wing. Cell proliferation was reduced in the ablated wing disc but was gradually recovered two days post ablation. Transcriptome analysis found that genes in the mitogen-activated protein kinase (MAPK) pathway were upregulated. Repression of gene expression in this pathway, including Ras oncogene at 64B (Ras64B), Downstream of raf1 (Dsor1), and cAMP-dependent protein kinase catalytic subunit 3 (Pka-C3) by RNA interference after ablation, led to diminishment of both adult wings, suggesting that the MAPK signaling is essential for wing growth. Additionally, cell proliferation was still decelerated by injecting Ras64B, Dsor, or Pka-C3 dsRNA two days after ablation, indicating that the MAPK signaling-regulated cell proliferation is essential for growth. These results provide molecular clues to the regulation of cell proliferation during regeneration in lepidopteran insects.
Collapse
Affiliation(s)
- Yuanyuan Xu
- Department of Entomology and MOA Lab for Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, 100193, China
| | - Wei Wei
- Department of Entomology and MOA Lab for Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, 100193, China
| | - Guangze Lin
- Department of Entomology and MOA Lab for Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, 100193, China
| | - Shuo Yan
- Department of Entomology and MOA Lab for Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, 100193, China
| | - Junzheng Zhang
- Department of Entomology and MOA Lab for Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, 100193, China
| | - Jie Shen
- Department of Entomology and MOA Lab for Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, 100193, China.
| | - Dan Wang
- Department of Entomology and MOA Lab for Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
27
|
Mase A, Augsburger J, Brückner K. Macrophages and Their Organ Locations Shape Each Other in Development and Homeostasis - A Drosophila Perspective. Front Cell Dev Biol 2021; 9:630272. [PMID: 33777939 PMCID: PMC7991785 DOI: 10.3389/fcell.2021.630272] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 01/25/2021] [Indexed: 12/15/2022] Open
Abstract
Across the animal kingdom, macrophages are known for their functions in innate immunity, but they also play key roles in development and homeostasis. Recent insights from single cell profiling and other approaches in the invertebrate model organism Drosophila melanogaster reveal substantial diversity among Drosophila macrophages (plasmatocytes). Together with vertebrate studies that show genuine expression signatures of macrophages based on their organ microenvironments, it is expected that Drosophila macrophage functional diversity is shaped by their anatomical locations and systemic conditions. In vivo evidence for diverse macrophage functions has already been well established by Drosophila genetics: Drosophila macrophages play key roles in various aspects of development and organogenesis, including embryogenesis and development of the nervous, digestive, and reproductive systems. Macrophages further maintain homeostasis in various organ systems and promote regeneration following organ damage and injury. The interdependence and interplay of tissues and their local macrophage populations in Drosophila have implications for understanding principles of organ development and homeostasis in a wide range of species.
Collapse
Affiliation(s)
- Anjeli Mase
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, United States
| | - Jordan Augsburger
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, United States
| | - Katja Brückner
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, United States
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, United States
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
28
|
Control of Drosophila wing size by morphogen range and hormonal gating. Proc Natl Acad Sci U S A 2020; 117:31935-31944. [PMID: 33257577 DOI: 10.1073/pnas.2018196117] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The stereotyped dimensions of animal bodies and their component parts result from tight constraints on growth. Yet, the mechanisms that stop growth when organs reach the right size are unknown. Growth of the Drosophila wing-a classic paradigm-is governed by two morphogens, Decapentaplegic (Dpp, a BMP) and Wingless (Wg, a Wnt). Wing growth during larval life ceases when the primordium attains full size, concomitant with the larval-to-pupal molt orchestrated by the steroid hormone ecdysone. Here, we block the molt by genetically dampening ecdysone production, creating an experimental paradigm in which the wing stops growing at the correct size while the larva continues to feed and gain body mass. Under these conditions, we show that wing growth is limited by the ranges of Dpp and Wg, and by ecdysone, which regulates the cellular response to their signaling activities. Further, we present evidence that growth terminates because of the loss of two distinct modes of morphogen action: 1) maintenance of growth within the wing proper and 2) induced growth of surrounding "pre-wing" cells and their recruitment into the wing. Our results provide a precedent for the control of organ size by morphogen range and the hormonal gating of morphogen action.
Collapse
|
29
|
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
|
30
|
Wnt/β-catenin Signaling in Tissue Self-Organization. Genes (Basel) 2020; 11:genes11080939. [PMID: 32823838 PMCID: PMC7464740 DOI: 10.3390/genes11080939] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/10/2020] [Accepted: 08/11/2020] [Indexed: 12/13/2022] Open
Abstract
Across metazoans, animal body structures and tissues exist in robust patterns that arise seemingly out of stochasticity of a few early cells in the embryo. These patterns ensure proper tissue form and function during early embryogenesis, development, homeostasis, and regeneration. Fundamental questions are how these patterns are generated and maintained during tissue homeostasis and regeneration. Though fascinating scientists for generations, these ideas remain poorly understood. Today, it is apparent that the Wnt/β-catenin pathway plays a central role in tissue patterning. Wnt proteins are small diffusible morphogens which are essential for cell type specification and patterning of tissues. In this review, we highlight several mechanisms described where the spatial properties of Wnt/β-catenin signaling are controlled, allowing them to work in combination with other diffusible molecules to control tissue patterning. We discuss examples of this self-patterning behavior during development and adult tissues' maintenance. The combination of new physiological culture systems, mathematical approaches, and synthetic biology will continue to fuel discoveries about how tissues are patterned. These insights are critical for understanding the intricate interplay of core patterning signals and how they become disrupted in disease.
Collapse
|
31
|
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
|
32
|
tpHusion: An efficient tool for clonal pH determination in Drosophila. PLoS One 2020; 15:e0228995. [PMID: 32059043 PMCID: PMC7021318 DOI: 10.1371/journal.pone.0228995] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 01/27/2020] [Indexed: 02/04/2023] Open
Abstract
Genetically encoded pH indicators (GEpHI) have emerged as important tools for investigating intracellular pH (pHi) dynamics in Drosophila. However, most of the indicators are based on the Gal4/UAS binary expression system. Here, we report the generation of a ubiquitously-expressed GEpHI. The fusion protein of super ecliptic pHluorin and FusionRed was cloned under the tubulin promoter (tpHusion) to drive it independently of the Gal4/UAS system. The function of tpHusion was validated in various tissues from different developmental stages of Drosophila. Differences in pHi were also indicated correctly in fixed tissues. Finally, we describe the use of tpHusion for comparative analysis of pHi in manipulated clones and the surrounding cells in epithelial tissues. Our findings establish tpHusion as a robust tool for studying pHi in Drosophila.
Collapse
|
33
|
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]
|
34
|
JNK-mediated Slit-Robo signaling facilitates epithelial wound repair by extruding dying cells. Sci Rep 2019; 9:19549. [PMID: 31863086 PMCID: PMC6925126 DOI: 10.1038/s41598-019-56137-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 12/03/2019] [Indexed: 01/09/2023] Open
Abstract
Multicellular organisms repair injured epithelium by evolutionarily conserved biological processes including activation of c-Jun N-terminal kinase (JNK) signaling. Here, we show in Drosophila imaginal epithelium that physical injury leads to the emergence of dying cells, which are extruded from the wounded tissue by JNK-induced Slit-Roundabout2 (Robo2) repulsive signaling. Reducing Slit-Robo2 signaling in the wounded tissue suppresses extrusion of dying cells and generates aberrant cells with highly upregulated growth factors Wingless (Wg) and Decapentaplegic (Dpp). The inappropriately elevated Wg and Dpp impairs wound repair, as halving one of these growth factor genes cancelled wound healing defects caused by Slit-Robo2 downregulation. Our data suggest that JNK-mediated Slit-Robo2 signaling contributes to epithelial wound repair by promoting extrusion of dying cells from the wounded tissue, which facilitates transient and appropriate induction of growth factors for proper wound healing.
Collapse
|
35
|
Held LI, Sessions SK. Reflections on Bateson's rule: Solving an old riddle about why extra legs are mirror‐symmetric. JOURNAL OF EXPERIMENTAL ZOOLOGY PART B-MOLECULAR AND DEVELOPMENTAL EVOLUTION 2019; 332:219-237. [DOI: 10.1002/jez.b.22910] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 08/18/2019] [Accepted: 09/26/2019] [Indexed: 12/17/2022]
Affiliation(s)
- Lewis I. Held
- Department of Biological SciencesTexas Tech University Lubbock Texas
| | | |
Collapse
|
36
|
Rotelli MD, Bolling AM, Killion AW, Weinberg AJ, Dixon MJ, Calvi BR. An RNAi Screen for Genes Required for Growth of Drosophila Wing Tissue. G3 (BETHESDA, MD.) 2019; 9:3087-3100. [PMID: 31387856 PMCID: PMC6778782 DOI: 10.1534/g3.119.400581] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 07/31/2019] [Indexed: 12/23/2022]
Abstract
Cell division and tissue growth must be coordinated with development. Defects in these processes are the basis for a number of diseases, including developmental malformations and cancer. We have conducted an unbiased RNAi screen for genes that are required for growth in the Drosophila wing, using GAL4-inducible short hairpin RNA (shRNA) fly strains made by the Drosophila RNAi Screening Center. shRNA expression down the center of the larval wing disc using dpp-GAL4, and the central region of the adult wing was then scored for tissue growth and wing hair morphology. Out of 4,753 shRNA crosses that survived to adulthood, 18 had impaired wing growth. FlyBase and the new Alliance of Genome Resources knowledgebases were used to determine the known or predicted functions of these genes and the association of their human orthologs with disease. The function of eight of the genes identified has not been previously defined in Drosophila The genes identified included those with known or predicted functions in cell cycle, chromosome segregation, morphogenesis, metabolism, steroid processing, transcription, and translation. All but one of the genes are similar to those in humans, and many are associated with disease. Knockdown of lin-52, a subunit of the Myb-MuvB transcription factor, or βNACtes6, a gene involved in protein folding and trafficking, resulted in a switch from cell proliferation to an endoreplication growth program through which wing tissue grew by an increase in cell size (hypertrophy). It is anticipated that further analysis of the genes that we have identified will reveal new mechanisms that regulate tissue growth during development.
Collapse
Affiliation(s)
- Michael D Rotelli
- Department of Biology, Indiana University, Bloomington, IN 47405 and
| | - Anna M Bolling
- Department of Biology, Indiana University, Bloomington, IN 47405 and
| | - Andrew W Killion
- Department of Biology, Indiana University, Bloomington, IN 47405 and
| | | | - Michael J Dixon
- Department of Biology, Indiana University, Bloomington, IN 47405 and
| | - Brian R Calvi
- Department of Biology, Indiana University, Bloomington, IN 47405 and
- Melvin and Bren Simon Cancer Center, Indiana University, Indianapolis, IN 46202
| |
Collapse
|
37
|
Tsai CR, Galko MJ. Casein kinase 1α decreases β-catenin levels at adherens junctions to facilitate wound closure in Drosophila larvae. Development 2019; 146:dev175133. [PMID: 31511254 PMCID: PMC6826034 DOI: 10.1242/dev.175133] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 09/04/2019] [Indexed: 12/25/2022]
Abstract
Skin wound repair is essential to restore barrier function and prevent infection after tissue damage. Wound-edge epidermal cells migrate as a sheet to close the wound. However, it is still unclear how cell-cell junctions are regulated during wound closure (WC). To study this, we examined adherens junctions during WC in Drosophila larvae. β-Catenin is reduced at the lateral cell-cell junctions of wound-edge epidermal cells in the early healing stages. Destruction complex components, including Ck1α, GSK3β and β-TrCP, suppress β-catenin levels in the larval epidermis. Tissue-specific RNAi targeting these genes also caused severe WC defects. The Ck1αRNAi -induced WC defect is related to adherens junctions because loss of either β-catenin or E-cadherin significantly rescued this WC defect. In contrast, TCFRNAi does not rescue the Ck1αRNAi -induced WC defect, suggesting that Wnt signaling is not related to this defect. Direct overexpression of β-catenin recapitulates most of the features of Ck1α reduction during wounding. Finally, loss of Ck1α also blocked junctional E-cadherin reduction around the wound. Our results suggest that Ck1α and the destruction complex locally regulate cell adhesion to facilitate efficient wound repair.
Collapse
Affiliation(s)
- Chang-Ru Tsai
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Michael J Galko
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Genetics & Epigenetics Graduate Program, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
38
|
Mehta AS, Singh A. Insights into regeneration tool box: An animal model approach. Dev Biol 2019; 453:111-129. [PMID: 30986388 PMCID: PMC6684456 DOI: 10.1016/j.ydbio.2019.04.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 04/04/2019] [Accepted: 04/09/2019] [Indexed: 12/20/2022]
Abstract
For ages, regeneration has intrigued countless biologists, clinicians, and biomedical engineers. In recent years, significant progress made in identification and characterization of a regeneration tool kit has helped the scientific community to understand the mechanism(s) involved in regeneration across animal kingdom. These mechanistic insights revealed that evolutionarily conserved pathways like Wnt, Notch, Hedgehog, BMP, and JAK/STAT are involved in regeneration. Furthermore, advancement in high throughput screening approaches like transcriptomic analysis followed by proteomic validations have discovered many novel genes, and regeneration specific enhancers that are specific to highly regenerative species like Hydra, Planaria, Newts, and Zebrafish. Since genetic machinery is highly conserved across the animal kingdom, it is possible to engineer these genes and regeneration specific enhancers in species with limited regeneration properties like Drosophila, and mammals. Since these models are highly versatile and genetically tractable, cross-species comparative studies can generate mechanistic insights in regeneration for animals with long gestation periods e.g. Newts. In addition, it will allow extrapolation of regenerative capabilities from highly regenerative species to animals with low regeneration potential, e.g. mammals. In future, these studies, along with advancement in tissue engineering applications, can have strong implications in the field of regenerative medicine and stem cell biology.
Collapse
Affiliation(s)
- Abijeet S Mehta
- Department of Biology, University of Dayton, Dayton, OH, 45469, USA
| | - Amit Singh
- Department of Biology, University of Dayton, Dayton, OH, 45469, USA; Premedical Program, University of Dayton, Dayton, OH, 45469, USA; Center for Tissue Regeneration and Engineering at Dayton (TREND), University of Dayton, Dayton, OH, 45469, USA; The Integrative Science and Engineering Center, University of Dayton, Dayton, OH, 45469, USA; Center for Genomic Advocacy (TCGA), Indiana State University, Terre Haute, IN, USA.
| |
Collapse
|
39
|
Houtz P, Bonfini A, Bing X, Buchon N. Recruitment of Adult Precursor Cells Underlies Limited Repair of the Infected Larval Midgut in Drosophila. Cell Host Microbe 2019; 26:412-425.e5. [PMID: 31492656 DOI: 10.1016/j.chom.2019.08.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 06/11/2019] [Accepted: 08/06/2019] [Indexed: 12/19/2022]
Abstract
Surviving infection requires immune and repair mechanisms. Developing organisms face the additional challenge of integrating these mechanisms with tightly controlled developmental processes. The larval Drosophila midgut lacks dedicated intestinal stem cells. We show that, upon infection, larvae perform limited repair using adult midgut precursors (AMPs). AMPs differentiate in response to damage to generate new enterocytes, transiently depleting their pool. Developmental delay allows for AMP reconstitution, ensuring the completion of metamorphosis. Notch signaling is required for the differentiation of AMPs into the encasing, niche-like peripheral cells (PCs), but not to differentiate PCs into enterocytes. Dpp (TGF-β) signaling is sufficient, but not necessary, to induce PC differentiation into enterocytes. Infection-induced JAK-STAT pathway is both required and sufficient for differentiation of AMPs and PCs into new enterocytes. Altogether, this work highlights the constraints imposed by development on an organism's response to infection and demonstrates the transient use of adult precursors for tissue repair.
Collapse
Affiliation(s)
- Philip Houtz
- Cornell Institute of Host-Microbe Interactions and Disease, Department of Entomology, Cornell University, 129 Garden Ave., Ithaca, NY 14853, USA
| | - Alessandro Bonfini
- Cornell Institute of Host-Microbe Interactions and Disease, Department of Entomology, Cornell University, 129 Garden Ave., Ithaca, NY 14853, USA
| | - Xiaoli Bing
- Cornell Institute of Host-Microbe Interactions and Disease, Department of Entomology, Cornell University, 129 Garden Ave., Ithaca, NY 14853, USA
| | - Nicolas Buchon
- Cornell Institute of Host-Microbe Interactions and Disease, Department of Entomology, Cornell University, 129 Garden Ave., Ithaca, NY 14853, USA.
| |
Collapse
|
40
|
Mehta AS, Luz-Madrigal A, Li JL, Tsonis PA, Singh A. Comparative transcriptomic analysis and structure prediction of novel Newt proteins. PLoS One 2019; 14:e0220416. [PMID: 31419228 PMCID: PMC6697330 DOI: 10.1371/journal.pone.0220416] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 07/15/2019] [Indexed: 01/25/2023] Open
Abstract
Notophthalmus viridescens (Red-spotted Newt) possess amazing capabilities to regenerate their organs and other tissues. Previously, using a de novo assembly of the newt transcriptome combined with proteomic validation, our group identified a novel family of five protein members expressed in adult tissues during regeneration in Notophthalmus viridescens. The presence of a putative signal peptide suggests that all these proteins are secretory in nature. Here we employed iterative threading assembly refinement (I-TASSER) server to generate three-dimensional structure of these novel Newt proteins and predicted their function. Our data suggests that these proteins could act as ion transporters, and be involved in redox reaction(s). Due to absence of transgenic approaches in N. viridescens, and conservation of genetic machinery across species, we generated transgenic Drosophila melanogaster to misexpress these genes. Expression of 2775 transcripts were compared between these five newly identified Newt genes. We found that genes involved in the developmental process, cell cycle, apoptosis, and immune response are among those that are highly enriched. To validate the RNA Seq. data, expression of six highly regulated genes were verified using real time Quantitative Polymerase Chain Reaction (RT-qPCR). These graded gene expression patterns provide insight into the function of novel protein family identified in Newt, and layout a map for future studies in the field.
Collapse
Affiliation(s)
- Abijeet Singh Mehta
- Department of Biology, University of Dayton, Dayton, Ohio, United States of America
| | - Agustin Luz-Madrigal
- Department of Biology, University of Dayton, Dayton, Ohio, United States of America
| | - Jian-Liang Li
- Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, Florida, United States of America
| | - Panagiotis A Tsonis
- Department of Biology, University of Dayton, Dayton, Ohio, United States of America
| | - Amit Singh
- Department of Biology, University of Dayton, Dayton, Ohio, United States of America
- Premedical Program, University of Dayton, Dayton, Ohio, United States of America
- Center for Tissue Regeneration and Engineering at Dayton (TREND), University of Dayton, Dayton, Ohio, United States of America
- The Integrative Science and Engineering Center, University of Dayton, Dayton, Ohio, United States of America
- Center for Genomic Advocacy (TCGA), Indiana State University, Terre Haute, Indiana, United States of America
| |
Collapse
|
41
|
Akmammedov A, Geigges M, Paro R. Bivalency in Drosophila embryos is associated with strong inducibility of Polycomb target genes. Fly (Austin) 2019; 13:42-50. [PMID: 31094269 DOI: 10.1080/19336934.2019.1619438] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Polycomb group (PcG) and Trithorax group (TrxG) proteins orchestrate development of a multicellular organism by faithfully maintaining cell fate decisions made early in embryogenesis. An important chromatin mark connected to PcG/TrxG regulation is bivalent domains, the simultaneous presence of H3K27me3 and H3K4me3 on a given locus, originally identified in mammalian embryonic stem cells but considered to be absent in invertebrates. Here, we provide evidence for the existence of bivalency in fly embryos. Using a recently described PcG reporter fly line, we observed a strong reporter inducibility in the embryo and its sharp decrease in larval and adult stages. Analysis of the chromatin landscape of the reporter revealed a strong signal for the repressive PcG mark, H3K27me3, in all three developmental stages and, surprisingly, a strong signal for a transcriptionally activating H3K4me3 mark in the embryo. Using re-chromatin immunoprecipitation experiments, bivalent domains were also uncovered at endogenous PcG targets like the Hox genes.
Collapse
Affiliation(s)
- Arslan Akmammedov
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Marco Geigges
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Renato Paro
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland.,Faculty of Science, University of Basel, Basel, Switzerland
| |
Collapse
|
42
|
Almudi I, Martín-Blanco CA, García-Fernandez IM, López-Catalina A, Davie K, Aerts S, Casares F. Establishment of the mayfly Cloeon dipterum as a new model system to investigate insect evolution. EvoDevo 2019; 10:6. [PMID: 30984364 PMCID: PMC6446309 DOI: 10.1186/s13227-019-0120-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 03/21/2019] [Indexed: 02/07/2023] Open
Abstract
The great capability of insects to adapt to new environments promoted their extraordinary diversification, resulting in the group of Metazoa with the largest number of species distributed worldwide. To understand this enormous diversity, it is essential to investigate lineages that would allow the reconstruction of the early events in the evolution of insects. However, research on insect ecology, physiology, development and evolution has mostly focused on few well-established model species. The key phylogenetic position of mayflies within Paleoptera as the sister group of the rest of winged insects and life history traits of mayflies make them an essential order to understand insect evolution. Here, we describe the establishment of a continuous culture system of the mayfly Cloeon dipterum and a series of experimental protocols and omics resources that allow the study of its development and its great regenerative capability. Thus, the establishment of Cloeon as an experimental platform paves the way to understand genomic and morphogenetic events that occurred at the origin of winged insects.
Collapse
Affiliation(s)
- Isabel Almudi
- 1GEM-DMC2 Unit, The CABD (CSIC-UPO-JA), Ctra. de Utrera km 1, 41013 Seville, Spain
| | | | | | | | - Kristofer Davie
- Laboratory of Computational Biology, VIB Center for Brain & Disease Research, Herestraat 49, 3000 Louvain, Belgium.,3Department of Human Genetics, KU Leuven, Oude Markt 13, 3000 Louvain, Belgium
| | - Stein Aerts
- Laboratory of Computational Biology, VIB Center for Brain & Disease Research, Herestraat 49, 3000 Louvain, Belgium.,3Department of Human Genetics, KU Leuven, Oude Markt 13, 3000 Louvain, Belgium
| | - Fernando Casares
- 1GEM-DMC2 Unit, The CABD (CSIC-UPO-JA), Ctra. de Utrera km 1, 41013 Seville, Spain
| |
Collapse
|
43
|
Herrera SC, Bach EA. JAK/STAT signaling in stem cells and regeneration: from Drosophila to vertebrates. Development 2019; 146:dev167643. [PMID: 30696713 PMCID: PMC6361132 DOI: 10.1242/dev.167643] [Citation(s) in RCA: 131] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 12/03/2018] [Indexed: 12/19/2022]
Abstract
The JAK/STAT pathway is a conserved metazoan signaling system that transduces cues from extracellular cytokines into transcriptional changes in the nucleus. JAK/STAT signaling is best known for its roles in immunity. However, recent work has demonstrated that it also regulates critical homeostatic processes in germline and somatic stem cells, as well as regenerative processes in several tissues, including the gonad, intestine and appendages. Here, we provide an overview of JAK/STAT signaling in stem cells and regeneration, focusing on Drosophila and highlighting JAK/STAT pathway functions in proliferation, survival and cell competition that are conserved between Drosophila and vertebrates.
Collapse
Affiliation(s)
- Salvador C Herrera
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY 10016, USA
| | - Erika A Bach
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY 10016, USA
- Helen L. and Martin S. Kimmel Center for Stem Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| |
Collapse
|
44
|
Suzuki Y, Chou J, Garvey SL, Wang VR, Yanes KO. Evolution and Regulation of Limb Regeneration in Arthropods. Results Probl Cell Differ 2019; 68:419-454. [PMID: 31598866 DOI: 10.1007/978-3-030-23459-1_17] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Regeneration has fascinated both scientists and non-scientists for centuries. Many organisms can regenerate, and arthropod limbs are no exception although their ability to regenerate is a product shaped by natural and sexual selection. Recent studies have begun to uncover cellular and molecular processes underlying limb regeneration in several arthropod species. Here we argue that an evo-devo approach to the study of arthropod limb regeneration is needed to understand aspects of limb regeneration that are conserved and divergent. In particular, we argue that limbs of different species are comprised of cells at distinct stages of differentiation at the time of limb loss and therefore provide insights into regeneration involving both stem cell-like cells/precursor cells and differentiated cells. In addition, we review recent studies that demonstrate how limb regeneration impacts the development of the whole organism and argue that studies on the link between local tissue damage and the rest of the body should provide insights into the integrative nature of development. Molecular studies on limb regeneration are only beginning to take off, but comparative studies on the mechanisms of limb regeneration across various taxa should not only yield interesting insights into development but also answer how this remarkable ability evolved across arthropods and beyond.
Collapse
Affiliation(s)
- Yuichiro Suzuki
- Department of Biological Sciences, Wellesley College, Wellesley, MA, USA.
| | - Jacquelyn Chou
- Department of Biological Sciences, Wellesley College, Wellesley, MA, USA
| | - Sarah L Garvey
- Department of Biological Sciences, Wellesley College, Wellesley, MA, USA
| | - Victoria R Wang
- Department of Biological Sciences, Wellesley College, Wellesley, MA, USA
| | - Katherine O Yanes
- Department of Biological Sciences, Wellesley College, Wellesley, MA, USA
| |
Collapse
|
45
|
Santabárbara-Ruiz P, Esteban-Collado J, Pérez L, Viola G, Abril JF, Milán M, Corominas M, Serras F. Ask1 and Akt act synergistically to promote ROS-dependent regeneration in Drosophila. PLoS Genet 2019; 15:e1007926. [PMID: 30677014 PMCID: PMC6363233 DOI: 10.1371/journal.pgen.1007926] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 02/05/2019] [Accepted: 01/01/2019] [Indexed: 12/30/2022] Open
Abstract
How cells communicate to initiate a regenerative response after damage has captivated scientists during the last few decades. It is known that one of the main signals emanating from injured cells is the Reactive Oxygen Species (ROS), which propagate to the surrounding tissue to trigger the replacement of the missing cells. However, the link between ROS production and the activation of regenerative signaling pathways is not yet fully understood. We describe here the non-autonomous ROS sensing mechanism by which living cells launch their regenerative program. To this aim, we used Drosophila imaginal discs as a model system due to its well-characterized regenerative ability after injury or cell death. We genetically-induced cell death and found that the Apoptosis signal-regulating kinase 1 (Ask1) is essential for regenerative growth. Ask1 senses ROS both in dying and living cells, but its activation is selectively attenuated in living cells by Akt1, the core kinase component of the insulin/insulin-like growth factor pathway. Akt1 phosphorylates Ask1 in a secondary site outside the kinase domain, which attenuates its activity. This modulation of Ask1 activity results in moderate levels of JNK signaling in the living tissue, as well as in activation of p38 signaling, both pathways required to turn on the regenerative response. Our findings demonstrate a non-autonomous activation of a ROS sensing mechanism by Ask1 and Akt1 to replace the missing tissue after damage. Collectively, these results provide the basis for understanding the molecular mechanism of communication between dying and living cells that triggers regeneration.
Collapse
Affiliation(s)
- Paula Santabárbara-Ruiz
- Department of Genetics, Microbiology and Statistics, School of Biology and Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, Spain
| | - José Esteban-Collado
- Department of Genetics, Microbiology and Statistics, School of Biology and Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, Spain
| | - Lidia Pérez
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, Barcelona, Spain
| | - Giacomo Viola
- Department of Genetics, Microbiology and Statistics, School of Biology and Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, Spain
| | - Josep F. Abril
- Department of Genetics, Microbiology and Statistics, School of Biology and Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, Spain
| | - Marco Milán
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Pg. Lluís Companys, Barcelona, Spain
| | - Montserrat Corominas
- Department of Genetics, Microbiology and Statistics, School of Biology and Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Pg. Lluís Companys, Barcelona, Spain
| | - Florenci Serras
- Department of Genetics, Microbiology and Statistics, School of Biology and Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, Spain
| |
Collapse
|
46
|
Vizcaya-Molina E, Klein CC, Serras F, Mishra RK, Guigó R, Corominas M. Damage-responsive elements in Drosophila regeneration. Genome Res 2018; 28:1852-1866. [PMID: 30459214 PMCID: PMC6280756 DOI: 10.1101/gr.233098.117] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 10/10/2018] [Indexed: 12/21/2022]
Abstract
One of the most important questions in regenerative biology is to unveil how and when genes change expression and trigger regeneration programs. The resetting of gene expression patterns during response to injury is governed by coordinated actions of genomic regions that control the activity of multiple sequence-specific DNA binding proteins. Using genome-wide approaches to interrogate chromatin function, we here identify the elements that regulate tissue recovery in Drosophila imaginal discs, which show a high regenerative capacity after genetically induced cell death. Our findings indicate there is global coregulation of gene expression as well as a regeneration program driven by different types of regulatory elements. Novel enhancers acting exclusively within damaged tissue cooperate with enhancers co-opted from other tissues and other developmental stages, as well as with endogenous enhancers that show increased activity after injury. Together, these enhancers host binding sites for regulatory proteins that include a core set of conserved transcription factors that control regeneration across metazoans.
Collapse
Affiliation(s)
- Elena Vizcaya-Molina
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia and Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona 08028, Catalonia, Spain
| | - Cecilia C Klein
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia and Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona 08028, Catalonia, Spain
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona 08003, Catalonia, Spain
| | - Florenci Serras
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia and Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona 08028, Catalonia, Spain
| | - Rakesh K Mishra
- The Centre for Cellular and Molecular Biology (CCMB), Hyderabad 500007, India
| | - Roderic Guigó
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona 08003, Catalonia, Spain
- Universitat Pompeu Fabra (UPF), Barcelona 08003, Catalonia, Spain
| | - Montserrat Corominas
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia and Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona 08028, Catalonia, Spain
| |
Collapse
|
47
|
Modelling Cooperative Tumorigenesis in Drosophila. BIOMED RESEARCH INTERNATIONAL 2018; 2018:4258387. [PMID: 29693007 PMCID: PMC5859872 DOI: 10.1155/2018/4258387] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 01/21/2018] [Indexed: 12/13/2022]
Abstract
The development of human metastatic cancer is a multistep process, involving the acquisition of several genetic mutations, tumour heterogeneity, and interactions with the surrounding microenvironment. Due to the complexity of cancer development in mammals, simpler model organisms, such as the vinegar fly, Drosophila melanogaster, are being utilized to provide novel insights into the molecular mechanisms involved. In this review, we highlight recent advances in modelling tumorigenesis using the Drosophila model, focusing on the cooperation of oncogenes or tumour suppressors, and the interaction of mutant cells with the surrounding tissue in epithelial tumour initiation and progression.
Collapse
|
48
|
Boone E, Boulan L, Andersen DS, Romero N, Léopold P, Colombani J. Des insulines pour orchestrer la croissance. Med Sci (Paris) 2017; 33:637-641. [DOI: 10.1051/medsci/20173306021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|