1
|
Stevens B, Popp R, Valera H, Krueger K, Petersen CP. Injury-induced Neuregulin-ErbB signaling from muscle mobilizes stem cells for whole-body regeneration in Acoels. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.23.630141. [PMID: 39764063 PMCID: PMC11703163 DOI: 10.1101/2024.12.23.630141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2025]
Abstract
The activation of progenitor cells near wound sites is a common feature of regeneration across species, but the conserved signaling mechanisms responsible for this step in whole-body regeneration are still incompletely understood. The acoel Hofstenia miamia undergoes whole-body regeneration using Piwi+ pluripotent adult stem cells (neoblasts) that accumulate at amputation sites early in the regeneration process. The EGFR signaling pathway has broad roles in controlling proliferation, migration, differentiation, and cell survival across metazoans. Using a candidate RNAi screening approach, we identify the Hofstenia EGFR erbB4-2 and Neuregulin nrg-1 genes as essential for blastema formation. Structure prediction of NRG-1 and ERBB4-2 proteins supports the likelihood of these factors interacting directly. After amputation injuries, nrg-1 expression is induced in body-wall muscle cells at the wound site by 6 hours and localizes to the tip of the outgrowing blastema over the next several days, while erbB4-2 is broadly expressed, including in muscle and neoblasts. Under nrg-1(RNAi) and erbB4-2(RNAi) conditions that impair blastema formation, animals still undergo the earliest responses to injury to activate expression of the Early Growth Response transcription factor egr, indicating a crucial role for EGFR signaling downstream of initial wound activation. nrg-1(RNAi) and erbB4-2(RNAi) animals possess Piwi+ and H3P+ mitotic neoblasts which hyperproliferate normally after amputation, but these cells fail to accumulate at the wound site. Therefore, muscle provides a source for Neuregulin-ErbB signaling necessary for the mobilization of proliferative progenitors to enable blastema outgrowth for whole-body regeneration in Hofstenia. These results indicate a shared functional requirement for muscle signaling to enable regeneration between planarians and acoels across 550 million years of evolution.
Collapse
Affiliation(s)
- Brian Stevens
- Department of Molecular Biosciences, Northwestern University; Evanston IL 60208
| | - Riley Popp
- Department of Molecular Biosciences, Northwestern University; Evanston IL 60208
| | - Heather Valera
- Department of Molecular Biosciences, Northwestern University; Evanston IL 60208
| | - Kyle Krueger
- Department of Molecular Biosciences, Northwestern University; Evanston IL 60208
| | - Christian P. Petersen
- Department of Molecular Biosciences, Northwestern University; Evanston IL 60208
- Robert Lurie Comprehensive Cancer Center, Northwestern University; Evanston IL 60208
| |
Collapse
|
2
|
Chavez Ramirez C, Khoo M, Lopez G M, Ferguson S, Walker S, Echeverri K. Comparison of Blastema Formation after Injury in Two Cephalopod Species. MICROPUBLICATION BIOLOGY 2023; 2023:10.17912/micropub.biology.000946. [PMID: 37799205 PMCID: PMC10550383 DOI: 10.17912/micropub.biology.000946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/04/2023] [Accepted: 09/19/2023] [Indexed: 10/07/2023]
Abstract
Regeneration is the ability to functionally replace significant amounts of lost tissue or whole appendages like arms, limbs or tentacles. The amount of tissue that can be regenerated varies among species, but regeneration is found in both invertebrate and vertebrate animals. Cephalopods have been broadly reported in the literature to regenerate their arms. There are over 800 species of Cephalopod; however, regeneration has only been documented in the literature in a few species (1). Here we compare arm regeneration in two species of cephalopod, the Octopus bimaculoides and the hummingbird bobtail squid Euprymna berryi.
Collapse
Affiliation(s)
| | - Miya Khoo
- University of Chicago, Chicago, Illinois, United States
| | - Marco Lopez G
- University of Chicago, Chicago, Illinois, United States
| | - Sophie Ferguson
- Eugene Bell Center for Regenerative Biology and Tissue Engineering, Marine Biological Laboratory, Woods Hole, Massachusetts, United States
| | - Sarah Walker
- Eugene Bell Center for Regenerative Biology and Tissue Engineering, Marine Biological Laboratory, Woods Hole, Massachusetts, United States
| | - Karen Echeverri
- Eugene Bell Center for Regenerative Biology and Tissue Engineering, Marine Biological Laboratory, Woods Hole, Massachusetts, United States
| |
Collapse
|
3
|
Sapède D, Bahraoui S, Abou Nassif L, Barthelaix A, Mathieu M, Jorgensen C, Djouad F. Cartilage regeneration in zebrafish depends on Nrg1/ErbB signaling pathway. Front Cell Dev Biol 2023; 11:1123299. [PMID: 37215080 PMCID: PMC10192884 DOI: 10.3389/fcell.2023.1123299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 04/19/2023] [Indexed: 05/24/2023] Open
Abstract
Objective: Cartilage, as the majority of adult mammalian tissues, has limited regeneration capacity. Cartilage degradation consecutive to joint injury or aging then leads to irreversible joint damage and diseases. In contrast, several vertebrate species such as the zebrafish have the remarkable capacity to spontaneously regenerate skeletal structures after severe injuries. The objective of our study was to test the regenerative capacity of Meckel's cartilage (MC) upon mechanical injury in zebrafish and to identify the mechanisms underlying this process. Methods and Results: Cartilage regenerative capacity in zebrafish larvae was investigated after mechanical injuries of the lower jaw MC in TgBAC(col2a1a:mCherry), to visualize the loss and recovery of cartilage. Confocal analysis revealed the formation of new chondrocytes and complete regeneration of MC at 14 days post-injury (dpi) via chondrocyte cell cycle re-entry and proliferation of pre-existing MC chondrocytes near the wound. Through expression analyses, we showed an increase of nrg1 expression in the regenerating lower jaw, which also expresses Nrg1 receptors, ErbB3 and ErbB2. Pharmacological inhibition of the ErbB pathway and specific knockdown of Nrg1 affected MC regeneration indicating the pivotal role of this pathway for cartilage regeneration. Finally, addition of exogenous NRG1 in an in vitro model of osteoarthritic (OA)-like chondrocytes induced by IL1β suggests that Nrg1/ErbB pathway is functional in mammalian chondrocytes and alleviates the increased expression of catabolic markers characteristic of OA-like chondrocytes. Conclusion: Our results show that the Nrg1/ErbB pathway is required for spontaneous cartilage regeneration in zebrafish and is of interest to design new therapeutic approaches to promote cartilage regeneration in mammals.
Collapse
Affiliation(s)
- Dora Sapède
- IRMB, University Montpellier, INSERM, Montpellier, France
| | - Sarah Bahraoui
- IRMB, University Montpellier, INSERM, Montpellier, France
| | | | | | - Marc Mathieu
- IRMB, University Montpellier, INSERM, Montpellier, France
| | - Christian Jorgensen
- IRMB, University Montpellier, INSERM, Montpellier, France
- CHU Montpellier, Montpellier, France
| | - Farida Djouad
- IRMB, University Montpellier, INSERM, Montpellier, France
| |
Collapse
|
4
|
Zhou S, Liu Z, Kawakami A. A PI3Kγ signal regulates macrophage recruitment to injured tissue for regenerative cell survival. Dev Growth Differ 2022; 64:433-445. [PMID: 36101496 PMCID: PMC9826243 DOI: 10.1111/dgd.12809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/20/2022] [Accepted: 08/03/2022] [Indexed: 01/11/2023]
Abstract
The interaction between immune cells and injured tissues is crucial for regeneration. Previous studies have shown that macrophages attenuate inflammation caused by injuries to support the survival of primed regenerative cells. Macrophage loss in zebrafish mutants like cloche (clo) causes extensive apoptosis in the regenerative cells of the amputated larval fin fold. However, the mechanism of interaction between macrophage and injured tissue is poorly understood. Here, we show that a phosphoinositide 3-kinase gamma (PI3Kγ)-mediated signal is essential for recruiting macrophages to the injured tissue. PI3Kγ inhibition by the PI3Kγ-specific inhibitor, 5-quinoxalin-6-ylmethylene-thiazolidine-2,4-dione (AS605240 or AS), displayed a similar apoptosis phenotype with that observed in clo mutants. We further show that PI3Kγ function during the early regenerative stage is necessary for macrophage recruitment to the injured site. Additionally, protein kinase B (Akt) overexpression in the AS-treated larvae suggested that Akt is not the direct downstream mediator of PI3Kγ for macrophage recruitment, while it independently plays a role for the survival of regenerative cells. Together, our study reveals that PI3Kγ plays a role for recruiting macrophages in response to regeneration.
Collapse
Affiliation(s)
- Siyu Zhou
- School of Life Science and TechnologyTokyo Institute of TechnologyYokohamaJapan
| | - Zhengcheng Liu
- School of Life Science and TechnologyTokyo Institute of TechnologyYokohamaJapan
| | - Atsushi Kawakami
- School of Life Science and TechnologyTokyo Institute of TechnologyYokohamaJapan
| |
Collapse
|
5
|
Lin MJ, Lee CM, Hsu WL, Chen BC, Lee SJ. Macrophages Break Interneuromast Cell Quiescence by Intervening in the Inhibition of Schwann Cells in the Zebrafish Lateral Line. Front Cell Dev Biol 2022; 10:907863. [PMID: 35846366 PMCID: PMC9285731 DOI: 10.3389/fcell.2022.907863] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Abstract
In the zebrafish lateral line system, interneuromast cells (INCs) between neuromasts are kept quiescent by underlying Schwann cells (SWCs). Upon severe injuries that cause the complete loss of an entire neuromast, INCs can occasionally differentiate into neuromasts but how they escape from the inhibition by SWCs is still unclear. Using a genetic/chemical method to ablate a neuromast precisely, we found that a small portion of larvae can regenerate a new neuromast. However, the residual regeneration capacity was hindered by inhibiting macrophages. Using in toto imaging, we further discovered heterogeneities in macrophage behavior and distribution along the lateral line. We witnessed the crawling of macrophages between the injured lateral line and SWCs during regeneration and between the second primordium and the first mature lateral line during development. It implies that macrophages may physically alleviate the nerve inhibition to break the dormancy of INCs during regeneration and development in the zebrafish lateral line.
Collapse
Affiliation(s)
- Meng-Ju Lin
- Department of Life Science, National Taiwan University, Taipei, Taiwan, R.O.C.
| | - Chia-Ming Lee
- Research Center for Applied Sciences, Academia Sinica, Taipei, Taiwan, R.O.C.
| | - Wei-Lin Hsu
- Department of Life Science, National Taiwan University, Taipei, Taiwan, R.O.C.
| | - Bi-Chang Chen
- Research Center for Applied Sciences, Academia Sinica, Taipei, Taiwan, R.O.C.
| | - Shyh-Jye Lee
- Department of Life Science, National Taiwan University, Taipei, Taiwan, R.O.C.
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan, R.O.C.
- Center for Biotechnology, National Taiwan University, Taipei, Taiwan, R.O.C.
- *Correspondence: Shyh-Jye Lee,
| |
Collapse
|
6
|
Wiltbank AT, Steinson ER, Criswell SJ, Piller M, Kucenas S. Cd59 and inflammation regulate Schwann cell development. eLife 2022; 11:e76640. [PMID: 35748863 PMCID: PMC9232220 DOI: 10.7554/elife.76640] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 06/01/2022] [Indexed: 11/13/2022] Open
Abstract
Efficient neurotransmission is essential for organism survival and is enhanced by myelination. However, the genes that regulate myelin and myelinating glial cell development have not been fully characterized. Data from our lab and others demonstrates that cd59, which encodes for a small GPI-anchored glycoprotein, is highly expressed in developing zebrafish, rodent, and human oligodendrocytes (OLs) and Schwann cells (SCs), and that patients with CD59 dysfunction develop neurological dysfunction during early childhood. Yet, the function of Cd59 in the developing nervous system is currently undefined. In this study, we demonstrate that cd59 is expressed in a subset of developing SCs. Using cd59 mutant zebrafish, we show that developing SCs proliferate excessively and nerves may have reduced myelin volume, altered myelin ultrastructure, and perturbed node of Ranvier assembly. Finally, we demonstrate that complement activity is elevated in cd59 mutants and that inhibiting inflammation restores SC proliferation, myelin volume, and nodes of Ranvier to wildtype levels. Together, this work identifies Cd59 and developmental inflammation as key players in myelinating glial cell development, highlighting the collaboration between glia and the innate immune system to ensure normal neural development.
Collapse
Affiliation(s)
- Ashtyn T Wiltbank
- Neuroscience Graduate Program, University of VirginiaCharlottesvilleUnited States
- Program in Fundamental Neuroscience, University of VirginiaCharlottesvilleUnited States
| | - Emma R Steinson
- Department of Biology, University of VirginiaCharlottesvilleUnited States
| | - Stacey J Criswell
- Department of Cell Biology, University of VirginiaCharlottesvilleUnited States
| | - Melanie Piller
- Department of Biology, University of VirginiaCharlottesvilleUnited States
| | - Sarah Kucenas
- Neuroscience Graduate Program, University of VirginiaCharlottesvilleUnited States
- Program in Fundamental Neuroscience, University of VirginiaCharlottesvilleUnited States
- Department of Biology, University of VirginiaCharlottesvilleUnited States
| |
Collapse
|
7
|
Brown RI, Kawakami K, Kucenas S. A novel gene trap line for visualization and manipulation of erbb3b + neural crest and glial cells in zebrafish. Dev Biol 2022; 482:114-123. [PMID: 34932993 DOI: 10.1016/j.ydbio.2021.12.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/08/2021] [Accepted: 12/16/2021] [Indexed: 12/22/2022]
Abstract
Glia are a diverse and essential cell type in the vertebrate nervous system. Transgenic tools and fluorescent reporter lines are critical resources to investigate how glial subtypes develop and function. However, despite the many lines available in zebrafish, the community still lacks the ability to label all unique stages of glial development and specific subpopulations of cells. To address this issue, we screened zebrafish gene and enhancer trap lines to find a novel reporter for peripheral glial subtypes. From these, we generated the gSAIzGFFD37A transgenic line that expresses GFP in neural crest cells and central and peripheral glia. We found that the gene trap construct is located within an intron of erbb3b, a gene essential for glial development. Additionally, we confirmed that GFP+ cells express erbb3b along with sox10, a known glial marker. From our screen, we have identified the gSAIzGFFD37A line as a novel and powerful tool for studying glia in the developing zebrafish, as well as a new resource to manipulate erbb3b+ cells.
Collapse
Affiliation(s)
- Robin Isadora Brown
- Department of Biology, University of Virginia, Charlottesville, VA, 22904, USA; Program in Fundamental Neuroscience, University of Virginia, Charlottesville, VA, 22904, USA
| | - Koichi Kawakami
- Laboratory of Molecular and Developmental Biology, National Institute of Genetics, and Department of Genetics, SOKENDAI The Graduate University for Advanced Studies, Mishima, Shizuoka, 444-8540, Japan
| | - Sarah Kucenas
- Department of Biology, University of Virginia, Charlottesville, VA, 22904, USA; Program in Fundamental Neuroscience, University of Virginia, Charlottesville, VA, 22904, USA.
| |
Collapse
|
8
|
NRG1/ErbB signalling controls the dialogue between macrophages and neural crest-derived cells during zebrafish fin regeneration. Nat Commun 2021; 12:6336. [PMID: 34732706 PMCID: PMC8566576 DOI: 10.1038/s41467-021-26422-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 09/07/2021] [Indexed: 11/12/2022] Open
Abstract
Fish species, such as zebrafish (Danio rerio), can regenerate their appendages after amputation through the formation of a heterogeneous cellular structure named blastema. Here, by combining live imaging of triple transgenic zebrafish embryos and single-cell RNA sequencing we established a detailed cell atlas of the regenerating caudal fin in zebrafish larvae. We confirmed the presence of macrophage subsets that govern zebrafish fin regeneration, and identified a foxd3-positive cell population within the regenerating fin. Genetic depletion of these foxd3-positive neural crest-derived cells (NCdC) showed that they are involved in blastema formation and caudal fin regeneration. Finally, chemical inhibition and transcriptomic analysis demonstrated that these foxd3-positive cells regulate macrophage recruitment and polarization through the NRG1/ErbB pathway. Here, we show the diversity of the cells required for blastema formation, identify a discrete foxd3-positive NCdC population, and reveal the critical function of the NRG1/ErbB pathway in controlling the dialogue between macrophages and NCdC. Some fish can regenerate appendages by formation of a structure called the blastema. Here, the authors use single-cell RNA sequencing to characterize the cells required for blastema formation and fin regeneration and identified neural crest cells that orchestrate regeneration via the NRG1/ErbB axis
Collapse
|
9
|
Chiang KY, Li YW, Li YH, Huang SJ, Wu CL, Gong HY, Wu JL. Progranulin A Promotes Compensatory Hepatocyte Proliferation via HGF/c-Met Signaling after Partial Hepatectomy in Zebrafish. Int J Mol Sci 2021; 22:ijms222011217. [PMID: 34681875 PMCID: PMC8538350 DOI: 10.3390/ijms222011217] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/14/2021] [Accepted: 10/16/2021] [Indexed: 01/11/2023] Open
Abstract
Compensatory hepatocyte proliferation and other liver regenerative processes are activated to sustain normal physiological function after liver injury. A major mitogen for liver regeneration is hepatocyte growth factor (HGF), and a previous study indicated that progranulin could modulate c-met, the receptor for HGF, to initiate hepatic outgrowth from hepatoblasts during embryonic development. However, a role for progranulin in compensatory hepatocyte proliferation has not been shown previously. Therefore, this study was undertaken to clarify whether progranulin plays a regulatory role during liver regeneration. To this end, we established a partial hepatectomy regeneration model in adult zebrafish that express a liver-specific fluorescent reporter. Using this model, we found that loss of progranulin A (GrnA) function by intraperitoneal-injection of a Vivo-Morpholino impaired and delayed liver regeneration after partial hepatectomy. Furthermore, transcriptome analysis and confirmatory quantitative real-time PCR suggested that cell cycle progression and cell proliferation was not as active in the morphants as controls, which may have been the result of comparative downregulation of the HGF/c-met axis by 36 h after partial hepatectomy. Finally, liver-specific overexpression of GrnA in transgenic zebrafish caused more abundant cell proliferation after partial hepatectomy compared to wild types. Thus, we conclude that GrnA positively regulates HGF/c-met signaling to promote hepatocyte proliferation during liver regeneration.
Collapse
Affiliation(s)
- Keng-Yu Chiang
- Department of Life Science, National Taiwan University, Taipei 10617, Taiwan;
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan; (Y.-W.L.); (Y.-H.L.); (S.-J.H.)
| | - Ya-Wen Li
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan; (Y.-W.L.); (Y.-H.L.); (S.-J.H.)
| | - Yen-Hsing Li
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan; (Y.-W.L.); (Y.-H.L.); (S.-J.H.)
| | - Shin-Jie Huang
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan; (Y.-W.L.); (Y.-H.L.); (S.-J.H.)
| | - Chih-Lu Wu
- Department of Chemistry and Biochemistry, National Chung Cheng University, Chiayi 62145, Taiwan;
| | - Hong-Yi Gong
- Department of Aquaculture, National Taiwan Ocean University, Keelung 20224, Taiwan;
- Center of Excellence for the Oceans, National Taiwan Ocean University, Keelung 20224, Taiwan
| | - Jen-Leih Wu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan; (Y.-W.L.); (Y.-H.L.); (S.-J.H.)
- College of Life Sciences, National Taiwan Ocean University, Keelung 20224, Taiwan
- Correspondence: ; Tel.: +886-2-27899568
| |
Collapse
|
10
|
Sinclair JW, Hoying DR, Bresciani E, Nogare DD, Needle CD, Berger A, Wu W, Bishop K, Elkahloun AG, Chitnis A, Liu P, Burgess SM. The Warburg effect is necessary to promote glycosylation in the blastema during zebrafish tail regeneration. NPJ Regen Med 2021; 6:55. [PMID: 34518542 PMCID: PMC8437957 DOI: 10.1038/s41536-021-00163-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 08/11/2021] [Indexed: 12/21/2022] Open
Abstract
Throughout their lifetime, fish maintain a high capacity for regenerating complex tissues after injury. We utilized a larval tail regeneration assay in the zebrafish Danio rerio, which serves as an ideal model of appendage regeneration due to its easy manipulation, relatively simple mixture of cell types, and superior imaging properties. Regeneration of the embryonic zebrafish tail requires development of a blastema, a mass of dedifferentiated cells capable of replacing lost tissue, a crucial step in all known examples of appendage regeneration. Using this model, we show that tail amputation triggers an obligate metabolic shift to promote glucose metabolism during early regeneration similar to the Warburg effect observed in tumor forming cells. Inhibition of glucose metabolism did not affect the overall health of the embryo but completely blocked the tail from regenerating after amputation due to the failure to form a functional blastema. We performed a time series of single-cell RNA sequencing on regenerating tails with and without inhibition of glucose metabolism. We demonstrated that metabolic reprogramming is required for sustained TGF-β signaling and blocking glucose metabolism largely mimicked inhibition of TGF-β receptors, both resulting in an aberrant blastema. Finally, we showed using genetic ablation of three possible metabolic pathways for glucose, that metabolic reprogramming is required to provide glucose specifically to the hexosamine biosynthetic pathway while neither glycolysis nor the pentose phosphate pathway were necessary for regeneration.
Collapse
Affiliation(s)
- Jason W Sinclair
- Translational and Functional Genomics Branch, National Human Genome Research Institute, Bethesda, MD, USA
| | - David R Hoying
- Translational and Functional Genomics Branch, National Human Genome Research Institute, Bethesda, MD, USA
| | - Erica Bresciani
- Translational and Functional Genomics Branch, National Human Genome Research Institute, Bethesda, MD, USA
| | - Damian Dalle Nogare
- Aquatic Models of Human Development Affinity Group, National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Carli D Needle
- Translational and Functional Genomics Branch, National Human Genome Research Institute, Bethesda, MD, USA
| | - Alexandra Berger
- Translational and Functional Genomics Branch, National Human Genome Research Institute, Bethesda, MD, USA
| | - Weiwei Wu
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, Bethesda, MD, USA
| | - Kevin Bishop
- Translational and Functional Genomics Branch, National Human Genome Research Institute, Bethesda, MD, USA
| | - Abdel G Elkahloun
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, Bethesda, MD, USA
| | - Ajay Chitnis
- Aquatic Models of Human Development Affinity Group, National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Paul Liu
- Translational and Functional Genomics Branch, National Human Genome Research Institute, Bethesda, MD, USA
| | - Shawn M Burgess
- Translational and Functional Genomics Branch, National Human Genome Research Institute, Bethesda, MD, USA.
| |
Collapse
|
11
|
Srivastava M. Beyond Casual Resemblances: Rigorous Frameworks for Comparing Regeneration Across Species. Annu Rev Cell Dev Biol 2021; 37:415-440. [PMID: 34288710 DOI: 10.1146/annurev-cellbio-120319-114716] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The majority of animal phyla have species that can regenerate. Comparing regeneration across animals can reconstruct the molecular and cellular evolutionary history of this process. Recent studies have revealed some similarity in regeneration mechanisms, but rigorous comparative methods are needed to assess whether these resemblances are ancestral pathways (homology) or are the result of convergent evolution (homoplasy). This review aims to provide a framework for comparing regeneration across animals, focusing on gene regulatory networks (GRNs), which are substrates for assessing process homology. The homology of the wound-induced activation of Wnt signaling and of adult stem cells are discussed as examples of ongoing studies of regeneration that enable comparisons in a GRN framework. Expanding the study of regeneration GRNs in currently studied species and broadening taxonomic sampling for these approaches will identify processes that are unifying principles of regeneration biology across animals. These insights are important both for evolutionary studies of regeneration and for human regenerative medicine. Expected final online publication date for the Annual Review of Cell and Developmental Biology, Volume 37 is October 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Mansi Srivastava
- Department of Organismic and Evolutionary Biology and Museum of Comparative Zoology, Harvard University, Cambridge, Massachusetts 02138, USA;
| |
Collapse
|
12
|
Golenberg N, Squirrell JM, Bennin DA, Rindy J, Pistono PE, Eliceiri KW, Shelef MA, Kang J, Huttenlocher A. Citrullination regulates wound responses and tissue regeneration in zebrafish. J Cell Biol 2020; 219:133858. [PMID: 32328635 PMCID: PMC7147109 DOI: 10.1083/jcb.201908164] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 12/17/2019] [Accepted: 01/31/2020] [Indexed: 01/05/2023] Open
Abstract
Calcium is an important early signal in wound healing, yet how these early signals promote regeneration remains unclear. Peptidylarginine deiminases (PADs), a family of calcium-dependent enzymes, catalyze citrullination, a post-translational modification that alters protein function and has been implicated in autoimmune diseases. We generated a mutation in the single zebrafish ancestral pad gene, padi2, that results in a loss of detectable calcium-dependent citrullination. The mutants exhibit impaired resolution of inflammation and regeneration after caudal fin transection. We identified a new subpopulation of cells displaying citrullinated histones within the notochord bead following tissue injury. Citrullination of histones in this region was absent, and wound-induced proliferation was perturbed in Padi2-deficient larvae. Taken together, our results show that Padi2 is required for the citrullination of histones within a group of cells in the notochord bead and for promoting wound-induced proliferation required for efficient regeneration. These findings identify Padi2 as a potential intermediary between early calcium signaling and subsequent tissue regeneration.
Collapse
Affiliation(s)
- Netta Golenberg
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI.,Cell and Molecular Biology Doctoral Training Program, University of Wisconsin-Madison, Madison, WI
| | - Jayne M Squirrell
- Laboratory for Optical and Computational Instrumentation, University of Wisconsin-Madison, Madison, WI
| | - David A Bennin
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI.,Department of Pediatrics, University of Wisconsin-Madison, Madison, WI
| | - Julie Rindy
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI.,Department of Pediatrics, University of Wisconsin-Madison, Madison, WI
| | - Paige E Pistono
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI.,Department of Pediatrics, University of Wisconsin-Madison, Madison, WI
| | - Kevin W Eliceiri
- Laboratory for Optical and Computational Instrumentation, University of Wisconsin-Madison, Madison, WI
| | - Miriam A Shelef
- Department of Medicine, University of Wisconsin-Madison, Madison, WI.,William S. Middleton Memorial Veterans Hospital, Madison, WI
| | - Junsu Kang
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI
| | - Anna Huttenlocher
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI.,Department of Pediatrics, University of Wisconsin-Madison, Madison, WI
| |
Collapse
|
13
|
Thompson JD, Ou J, Lee N, Shin K, Cigliola V, Song L, Crawford GE, Kang J, Poss KD. Identification and requirements of enhancers that direct gene expression during zebrafish fin regeneration. Development 2020; 147:dev.191262. [PMID: 32665240 DOI: 10.1242/dev.191262] [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] [Received: 04/06/2020] [Accepted: 07/01/2020] [Indexed: 12/13/2022]
Abstract
To identify candidate tissue regeneration enhancer elements (TREEs) important for zebrafish fin regeneration, we performed ATAC-seq from bulk tissue or purified fibroblasts of uninjured and regenerating caudal fins. We identified tens of thousands of DNA regions from each sample type with dynamic accessibility during regeneration, and assigned these regions to proximal genes with corresponding expression changes by RNA-seq. To determine whether these profiles reveal bona fide TREEs, we tested the sufficiency and requirements of several sequences in stable transgenic lines and mutant lines with homozygous deletions. These experiments validated new non-coding regulatory sequences near induced and/or essential genes during fin regeneration, including fgf20a, mdka and cx43, identifying distinct domains of directed expression for each confirmed TREE. Whereas deletion of the previously identified LEN enhancer abolished detectable induction of the nearby leptin b gene during regeneration, deletions of enhancers linked to fgf20a, mdka and cx43 had no effect or partially reduced gene expression. Our study generates a new resource for dissecting the regulatory mechanisms of appendage generation and reveals a range of requirements for individual TREEs in control of regeneration programs.
Collapse
Affiliation(s)
- John D Thompson
- Regeneration Next, Duke University, Durham, NC 27710, USA.,Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Jianhong Ou
- Regeneration Next, Duke University, Durham, NC 27710, USA
| | - Nutishia Lee
- Regeneration Next, Duke University, Durham, NC 27710, USA.,Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Kwangdeok Shin
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Valentina Cigliola
- Regeneration Next, Duke University, Durham, NC 27710, USA.,Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Lingyun Song
- Department of Pediatrics, Division of Medical Genetics, Duke University Medical Center; Center for Genomic and Computational Biology; Center for Advanced Genomic Technologies, Durham, NC 27710, USA
| | - Gregory E Crawford
- Department of Pediatrics, Division of Medical Genetics, Duke University Medical Center; Center for Genomic and Computational Biology; Center for Advanced Genomic Technologies, Durham, NC 27710, USA
| | - Junsu Kang
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Kenneth D Poss
- Regeneration Next, Duke University, Durham, NC 27710, USA .,Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
14
|
Pei W, Xu L, Chen Z, Slevin CC, Pettie KP, Wincovitch S, Burgess SM. A subset of SMN complex members have a specific role in tissue regeneration via ERBB pathway-mediated proliferation. NPJ Regen Med 2020; 5:6. [PMID: 32218991 PMCID: PMC7096462 DOI: 10.1038/s41536-020-0089-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 01/27/2020] [Indexed: 12/24/2022] Open
Abstract
Spinal muscular atrophy (SMA) is the most common genetic disease in children. SMA is generally caused by mutations in the gene SMN1. The survival of motor neurons (SMN) complex consists of SMN1, Gemins (2-8), and Strap/Unrip. We previously demonstrated smn1 and gemin5 inhibited tissue regeneration in zebrafish. Here we investigated each individual SMN complex member and identified gemin3 as another regeneration-essential gene. These three genes are likely pan-regenerative, since they affect the regeneration of hair cells, liver, and caudal fin. RNA-Seq analysis reveals that smn1, gemin3, and gemin5 are linked to a common set of genetic pathways, including the tp53 and ErbB pathways. Additional studies indicated all three genes facilitate regeneration by inhibiting the ErbB pathway, thereby allowing cell proliferation in the injured neuromasts. This study provides a new understanding of the SMN complex and a potential etiology for SMA and potentially other rare unidentified genetic diseases with similar symptoms.
Collapse
Affiliation(s)
- Wuhong Pei
- Translational and Functional Genomics Branch, National Human Genome Research Institute, Bethesda, MD 20892 USA
| | - Lisha Xu
- Translational and Functional Genomics Branch, National Human Genome Research Institute, Bethesda, MD 20892 USA
| | - Zelin Chen
- Translational and Functional Genomics Branch, National Human Genome Research Institute, Bethesda, MD 20892 USA
| | - Claire C. Slevin
- Translational and Functional Genomics Branch, National Human Genome Research Institute, Bethesda, MD 20892 USA
| | - Kade P. Pettie
- Translational and Functional Genomics Branch, National Human Genome Research Institute, Bethesda, MD 20892 USA
| | - Stephen Wincovitch
- Cytogenetics and Microscopy Core, National Human Genome Research Institute, Bethesda, MD 20892 USA
| | - Shawn M. Burgess
- Translational and Functional Genomics Branch, National Human Genome Research Institute, Bethesda, MD 20892 USA
| |
Collapse
|
15
|
Grigorian Shamagian L, Madonna R, Taylor D, Climent AM, Prosper F, Bras-Rosario L, Bayes-Genis A, Ferdinandy P, Fernández-Avilés F, Izpisua Belmonte JC, Fuster V, Bolli R. Perspectives on Directions and Priorities for Future Preclinical Studies in Regenerative Medicine. Circ Res 2019; 124:938-951. [PMID: 30870121 DOI: 10.1161/circresaha.118.313795] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The myocardium consists of numerous cell types embedded in organized layers of ECM (extracellular matrix) and requires an intricate network of blood and lymphatic vessels and nerves to provide nutrients and electrical coupling to the cells. Although much of the focus has been on cardiomyocytes, these cells make up <40% of cells within a healthy adult heart. Therefore, repairing or regenerating cardiac tissue by merely reconstituting cardiomyocytes is a simplistic and ineffective approach. In fact, when an injury occurs, cardiac tissue organization is disrupted at the level of the cells, the tissue architecture, and the coordinated interaction among the cells. Thus, reconstitution of a functional tissue must reestablish electrical and mechanical communication between cardiomyocytes and restore their surrounding environment. It is also essential to restore distinctive myocardial features, such as vascular patency and pump function. In this article, we review the current status, challenges, and future priorities in cardiac regenerative or reparative medicine. In the first part, we provide an overview of our current understanding of heart repair and comment on the main contributors and mechanisms involved in innate regeneration. A brief section is dedicated to the novel concept of rejuvenation or regeneration, which we think may impact future development in the field. The last section describes regenerative therapies, where the most advanced and disruptive strategies used for myocardial repair are discussed. Our recommendations for priority areas in studies of cardiac regeneration or repair are summarized in Tables 1 and 2 .
Collapse
Affiliation(s)
- Lilian Grigorian Shamagian
- From the Hospital Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain (L.G.S., A.M.C., F.F.-A.).,CIBERCV, ISCIII, Madrid, Spain (L.G.S., A.M.C., A.B.-G., F.F.-A., V.F.)
| | - Rosalinda Madonna
- Center of Aging Sciences and Translational Medicine (CESI-MeT), Institute of Cardiology, G. d'Annunzio University, Chieti, Italy (R.M.).,Department of Internal Medicine, the University of Texas Health Science Center at Houston (R.M., )
| | | | - Andreu M Climent
- From the Hospital Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain (L.G.S., A.M.C., F.F.-A.).,CIBERCV, ISCIII, Madrid, Spain (L.G.S., A.M.C., A.B.-G., F.F.-A., V.F.)
| | | | - Luis Bras-Rosario
- Cardiology Department, Santa Maria University Hospital (CHLN), Lisbon Academic Medical Centre and Cardiovascular Centre of the University of Lisbon, Faculty of Medicine, Portugal (L.B.-R.)
| | - Antoni Bayes-Genis
- CIBERCV, ISCIII, Madrid, Spain (L.G.S., A.M.C., A.B.-G., F.F.-A., V.F.).,Hospital Germans Trias i Pujol, Badalona, Spain (A.B.-G.)
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.).,Pharmahungary Group, Szeged, Hungary (P.F.)
| | - Francisco Fernández-Avilés
- From the Hospital Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain (L.G.S., A.M.C., F.F.-A.).,CIBERCV, ISCIII, Madrid, Spain (L.G.S., A.M.C., A.B.-G., F.F.-A., V.F.)
| | | | - Valentin Fuster
- CIBERCV, ISCIII, Madrid, Spain (L.G.S., A.M.C., A.B.-G., F.F.-A., V.F.).,The Mount Sinai Hospital, New York, NY (V.F.).,Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (V.F.)
| | | |
Collapse
|
16
|
Nishiya N, Murai M, Hosoda A, Yonezawa H, Omori N. Bucillamine Prevents Afatinib-Mediated Inhibition of Epidermal Growth Factor Receptor Signaling. Pharmaceuticals (Basel) 2019; 12:ph12040165. [PMID: 31703435 PMCID: PMC6958386 DOI: 10.3390/ph12040165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/29/2019] [Accepted: 11/05/2019] [Indexed: 11/16/2022] Open
Abstract
Molecular targeting therapies often cause characteristic adverse effects, such as skin rash during anti-epidermal growth factor receptor (EGFR) therapies, making treatment continuation difficult. In contrast, skin symptoms induced by EGFR inhibition are strongly correlated with the overall survival of the therapies. Therefore, controlling adverse effects not only facilitates treatment continuation but also increases clinical benefits. In this study, we proposed a novel strategy for reducing EGFR–tyrosine kinase inhibitor (TKI)-induced adverse effects in nontumorous organs by repositioning approved medicines using a zebrafish model. We developed a model system for evaluating chemical quenchers of afatinib, a clinically available irreversible EGFR-TKI, by scoring the inhibition of afatinib-induced hyperformation of lateral line neuromasts in zebrafish larvae. Bucillamine, an antirheumatic drug, was identified as an afatinib quencher in the zebrafish system and inhibited TKI activity in vitro. In addition, bucillamine restored EGFR autophosphorylation and downstream signaling in afatinib-treated A431 cells. Thus, topical bucillamine is a potential reliever of irreversible EGFR-TKI-induced skin rash. The zebrafish model can be applied to a screening for quenchers of other anti-EGFR-targeting therapies, including reversible TKIs and biologics.
Collapse
Affiliation(s)
- Naoyuki Nishiya
- Division of Integrated Information for Pharmaceutical Sciences, Department of Clinical Pharmacy, Iwate Medical University School of Pharmacy, 1-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate 028-3694, Japan; (M.M.); (A.H.); (H.Y.); (N.O.)
- Correspondence:
| | - Moeka Murai
- Division of Integrated Information for Pharmaceutical Sciences, Department of Clinical Pharmacy, Iwate Medical University School of Pharmacy, 1-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate 028-3694, Japan; (M.M.); (A.H.); (H.Y.); (N.O.)
| | - Ayumi Hosoda
- Division of Integrated Information for Pharmaceutical Sciences, Department of Clinical Pharmacy, Iwate Medical University School of Pharmacy, 1-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate 028-3694, Japan; (M.M.); (A.H.); (H.Y.); (N.O.)
| | - Honami Yonezawa
- Division of Integrated Information for Pharmaceutical Sciences, Department of Clinical Pharmacy, Iwate Medical University School of Pharmacy, 1-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate 028-3694, Japan; (M.M.); (A.H.); (H.Y.); (N.O.)
| | - Norikazu Omori
- Division of Integrated Information for Pharmaceutical Sciences, Department of Clinical Pharmacy, Iwate Medical University School of Pharmacy, 1-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate 028-3694, Japan; (M.M.); (A.H.); (H.Y.); (N.O.)
- Department of Pharmacy, Iwate Medical University Hospital, 2-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate 028-3695, Japan
| |
Collapse
|
17
|
Franco JJ, Atieh Y, Bryan CD, Kwan KM, Eisenhoffer GT. Cellular crowding influences extrusion and proliferation to facilitate epithelial tissue repair. Mol Biol Cell 2019; 30:1890-1899. [PMID: 30785842 PMCID: PMC6727764 DOI: 10.1091/mbc.e18-05-0295] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Epithelial wound healing requires a complex orchestration of cellular rearrangements and movements to restore tissue architecture and function after injury. While it is well known that mechanical forces can affect tissue morphogenesis and patterning, how the biophysical cues generated after injury influence cellular behaviors during tissue repair is not well understood. Using time-lapse confocal imaging of epithelial tissues in living zebrafish larvae, we provide evidence that localized increases in cellular crowding during wound closure promote the extrusion of nonapoptotic cells via mechanically regulated stretch-activated ion channels (SACs). Directed cell migration toward the injury site promoted rapid changes in cell number and generated shifts in tension at cellular interfaces over long spatial distances. Perturbation of SAC activity resulted in failed extrusion and increased proliferation in crowded areas of the tissue. Together, we conclude that localized cell number plays a key role in dictating cellular behaviors that facilitate wound closure and tissue repair.
Collapse
Affiliation(s)
- Jovany J Franco
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030.,Department of BioSciences, Rice University, Houston, TX 77251
| | - Youmna Atieh
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Chase D Bryan
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112
| | - Kristen M Kwan
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112
| | - George T Eisenhoffer
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030.,Genetics and Epigenetics Graduate Program, Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| |
Collapse
|
18
|
Kakebeen AD, Wills AE. More Than Just a Bandage: Closing the Gap Between Injury and Appendage Regeneration. Front Physiol 2019; 10:81. [PMID: 30800076 PMCID: PMC6376490 DOI: 10.3389/fphys.2019.00081] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 01/24/2019] [Indexed: 01/19/2023] Open
Abstract
The remarkable regenerative capabilities of amphibians have captured the attention of biologists for centuries. The frogs Xenopus laevis and Xenopus tropicalis undergo temporally restricted regenerative healing of appendage amputations and spinal cord truncations, injuries that are both devastating and relatively common in human patients. Rapidly expanding technological innovations have led to a resurgence of interest in defining the factors that enable regenerative healing, and in coupling these factors to human therapeutic interventions. It is well-established that early embryonic signaling pathways are critical for growth and patterning of new tissue during regeneration. A growing body of research now indicates that early physiological injury responses are also required to initiate a regenerative program, and that these differ in regenerative and non-regenerative contexts. Here we review recent insights into the biophysical, biochemical, and epigenetic processes that underlie regenerative healing in amphibians, focusing particularly on tail and limb regeneration in Xenopus. We also discuss the more elusive potential mechanisms that link wounding to tissue growth and patterning.
Collapse
Affiliation(s)
- Anneke D Kakebeen
- Department of Biochemistry, University of Washington School of Medicine, Seattle, WA, United States
| | - Andrea E Wills
- Department of Biochemistry, University of Washington School of Medicine, Seattle, WA, United States
| |
Collapse
|
19
|
Romero MMG, McCathie G, Jankun P, Roehl HH. Damage-induced reactive oxygen species enable zebrafish tail regeneration by repositioning of Hedgehog expressing cells. Nat Commun 2018; 9:4010. [PMID: 30275454 PMCID: PMC6167316 DOI: 10.1038/s41467-018-06460-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 08/30/2018] [Indexed: 12/20/2022] Open
Abstract
Many aquatic vertebrates have a remarkable ability to regenerate limbs and tails after amputation. Previous studies indicate that reactive oxygen species (ROS) signalling initiates regeneration, but the mechanism by which this takes place is poorly understood. Developmental signalling pathways have been shown to have proregenerative roles in many systems. However, whether these are playing roles that are specific to regeneration, or are simply recapitulating their developmental functions is unclear. Here, we analyse zebrafish larval tail regeneration and find evidence that ROS released upon wounding cause repositioning of notochord cells to the damage site. These cells secrete Hedgehog ligands that are required for regeneration. Hedgehog signalling is not required for normal tail development suggesting that it has a regeneration-specific role. Our results provide a model for how ROS initiate tail regeneration, and indicate that developmental signalling pathways can play regenerative functions that are not directly related to their developmental roles. Reactive oxygen species (ROS) are required to initiate regeneration but the mechanisms regulating its production are unclear. Here, the authors show in zebrafish larval tail regeneration that ROS is released by mobilised notochord cells enables their repositioning in the damage site, assisted by secreted Hh.
Collapse
Affiliation(s)
- Maria Montserrat Garcia Romero
- Bateson Centre, Department of Biomedical Sciences, The University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK
| | - Gareth McCathie
- Bateson Centre, Department of Biomedical Sciences, The University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK
| | - Philip Jankun
- Bateson Centre, Department of Biomedical Sciences, The University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK
| | - Henry Hamilton Roehl
- Bateson Centre, Department of Biomedical Sciences, The University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK.
| |
Collapse
|
20
|
The role of the EGFR signaling pathway in stem cell differentiation during planarian regeneration and homeostasis. Semin Cell Dev Biol 2018; 87:45-57. [PMID: 29775660 DOI: 10.1016/j.semcdb.2018.05.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 05/07/2018] [Accepted: 05/14/2018] [Indexed: 12/25/2022]
Abstract
Cell signaling is essential for cells to adequately respond to their environment. One of the most evolutionarily conserved signaling pathways is that of the epidermal growth factor receptor (EGFR). Transmembrane receptors with intracellular tyrosine kinase activity are activated by the binding of their corresponding ligands. This in turn activates a wide variety of intracellular cascades and induces the up- or downregulation of target genes, leading to a specific cellular response. Freshwater planarians are an excellent model in which to study the role of cell signaling in the context of stem-cell based regeneration. Owing to the presence of a population of pluripotent stem cells called neoblasts, these animals can regenerate the entire organism from a tiny piece of the body. Here, we review the current state of knowledge of the planarian EGFR pathway. We describe the main components of the pathway and their functions in other animals, and focus in particular on receptors and ligands identified in the planarian Schmidtea mediterranea. Moreover, we summarize current data on the function of some of these components during planarian regeneration and homeostasis. We hypothesize that the EGFR pathway may act as a key regulator of the terminal differentiation of distinct populations of lineage-committed progenitors.
Collapse
|
21
|
Ricci L, Srivastava M. Wound-induced cell proliferation during animal regeneration. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2018; 7:e321. [PMID: 29719123 DOI: 10.1002/wdev.321] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 03/20/2018] [Accepted: 03/20/2018] [Indexed: 12/19/2022]
Abstract
Many animal species are capable of replacing missing tissues that are lost upon injury or amputation through the process of regeneration. Although the extent of regeneration is variable across animals, that is, some animals can regenerate any missing cell type whereas some can only regenerate certain organs or tissues, regulated cell proliferation underlies the formation of new tissues in most systems. Notably, many species display an increase in proliferation within hours or days upon wounding. While different cell types proliferate in response to wounding in various animal taxa, comparative molecular data are beginning to point to shared wound-induced mechanisms that regulate cell division during regeneration. Here, we synthesize current insights about early molecular pathways of regeneration from diverse model and emerging systems by considering these species in their evolutionary contexts. Despite the great diversity of mechanisms underlying injury-induced cell proliferation across animals, and sometimes even in the same species, similar pathways for proliferation have been implicated in distantly related species (e.g., small diffusible molecules, signaling from apoptotic cells, growth factor signaling, mTOR and Hippo signaling, and Wnt and Bmp pathways). Studies that explicitly interrogate molecular and cellular regenerative mechanisms in understudied animal phyla will reveal the extent to which early pathways in the process of regeneration are conserved or independently evolved. This article is categorized under: Comparative Development and Evolution > Body Plan Evolution Adult Stem Cells, Tissue Renewal, and Regeneration > Regeneration Comparative Development and Evolution > Model Systems.
Collapse
Affiliation(s)
- Lorenzo Ricci
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, Massachusetts
| | - Mansi Srivastava
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, Massachusetts
| |
Collapse
|
22
|
Hale AJ, den Hertog J. Shp2-Mitogen-Activated Protein Kinase Signaling Drives Proliferation during Zebrafish Embryo Caudal Fin Fold Regeneration. Mol Cell Biol 2018; 38:e00515-17. [PMID: 29203641 PMCID: PMC5789028 DOI: 10.1128/mcb.00515-17] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 10/18/2017] [Accepted: 11/23/2017] [Indexed: 11/25/2022] Open
Abstract
Regeneration of the zebrafish caudal fin following amputation occurs through wound healing, followed by formation of a blastema, which produces cells to replace the lost tissue in the final phase of regenerative outgrowth. We show that ptpn11a-/- ptpn11b-/- zebrafish embryos, lacking functional Shp2, fail to regenerate their caudal fin folds. Rescue experiments indicated that Shp2a has a functional signaling role, requiring its catalytic activity and SH2 domains but not the two C-terminal tyrosine phosphorylation sites. Surprisingly, expression of Shp2a variants with increased and reduced catalytic activity, respectively, rescued caudal fin fold regeneration to similar extents. Expression of mmp9 and junbb, indicative of formation of the wound epidermis and distal blastema, respectively, suggested that these processes occurred in ptpn11a-/- ptpn11b-/- zebrafish embryos. However, cell proliferation and MAPK phosphorylation were reduced. Pharmacological inhibition of MEK1 in wild-type zebrafish embryos phenocopied loss of Shp2. Our results suggest an essential role for Shp2a-mitogen-activated protein kinase (MAPK) signaling in promoting cell proliferation during zebrafish embryo caudal fin fold regeneration.
Collapse
Affiliation(s)
- Alexander James Hale
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, the Netherlands
- Institute Biology Leiden, Leiden University, Leiden, the Netherlands
| | - Jeroen den Hertog
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, the Netherlands
- Institute Biology Leiden, Leiden University, Leiden, the Netherlands
| |
Collapse
|
23
|
Brown D, Samsa LA, Ito C, Ma H, Batres K, Arnaout R, Qian L, Liu J. Neuregulin-1 is essential for nerve plexus formation during cardiac maturation. J Cell Mol Med 2017; 22:2007-2017. [PMID: 29265764 PMCID: PMC5824398 DOI: 10.1111/jcmm.13408] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 08/30/2017] [Indexed: 01/08/2023] Open
Abstract
The Neuregulin‐1 (Nrg1)/ErbB pathway plays multiple, critical roles in early cardiac and nervous system development and has been implicated in both heart and nerve repair processes. However, the early embryonic lethality of mouse Nrg1 mutants precludes an analysis of Nrg1's function in later cardiac development and homeostasis. In this study, we generated a novel nrg1 null allele targeting all known isoforms of nrg1 in zebrafish and examined cardiac structural and functional parameters throughout development. We found that zebrafish nrg1 mutants instead survived until young adult stages when they exhibited reduced survivorship. This coincided with structural and functional defects in the developing juvenile and young adult hearts, as demonstrated by reduced intracardiac myocardial density, cardiomyocyte cell number, swimming performance and dysregulated heartbeat. Interestingly, nrg1 mutant hearts were missing long axons on the ventricle surface by standard length (SL) 5 mm, which preceded juvenile and adult cardiac defects. Given that the autonomic nervous system normally exerts fine control of cardiac output through this nerve plexus, these data suggest that Nrg1 may play a critical role in establishing the cardiac nerve plexus such that inadequate innervation leads to deficits in cardiac maturation, function and survival.
Collapse
Affiliation(s)
- Daniel Brown
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Leigh Ann Samsa
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA
| | - Cade Ito
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Hong Ma
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Karla Batres
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Rima Arnaout
- Department of Medicine, Division of Cardiology, Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Li Qian
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Jiandong Liu
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
24
|
García-Caballero M, Quesada AR, Medina MA, Marí-Beffa M. Fishing anti(lymph)angiogenic drugs with zebrafish. Drug Discov Today 2017; 23:366-374. [PMID: 29081356 DOI: 10.1016/j.drudis.2017.10.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Revised: 10/13/2017] [Accepted: 10/19/2017] [Indexed: 10/18/2022]
Abstract
Zebrafish, an amenable small teleost fish with a complex mammal-like circulatory system, is being increasingly used for drug screening and toxicity studies. It combines the biological complexity of in vivo models with a higher-throughput screening capability compared with other available animal models. Externally growing, transparent embryos, displaying well-defined blood and lymphatic vessels, allow the inexpensive, rapid, and automatable evaluation of drug candidates that are able to inhibit neovascularisation. Here, we briefly review zebrafish as a model for the screening of anti(lymph)angiogenic drugs, with emphasis on the advantages and limitations of the different zebrafish-based in vivo assays.
Collapse
Affiliation(s)
- Melissa García-Caballero
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, and IBIMA (Biomedical Research Institute of Málaga), University of Málaga, Andalucía Tech, Málaga, Spain; Unit 741 of CIBER de Enfermedades Raras, Málaga, Spain
| | - Ana R Quesada
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, and IBIMA (Biomedical Research Institute of Málaga), University of Málaga, Andalucía Tech, Málaga, Spain; Unit 741 of CIBER de Enfermedades Raras, Málaga, Spain
| | - Miguel A Medina
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, and IBIMA (Biomedical Research Institute of Málaga), University of Málaga, Andalucía Tech, Málaga, Spain; Unit 741 of CIBER de Enfermedades Raras, Málaga, Spain.
| | - Manuel Marí-Beffa
- Department of Cellular Biology, Genetics and Physiology, Faculty of Sciences, University of Málaga, Málaga, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina, Málaga, Spain.
| |
Collapse
|
25
|
Hale AJ, Kiai A, Sikkens J, den Hertog J. Impaired caudal fin-fold regeneration in zebrafish deficient for the tumor suppressor Pten. REGENERATION (OXFORD, ENGLAND) 2017; 4:217-226. [PMID: 29299324 PMCID: PMC5743786 DOI: 10.1002/reg2.88] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 09/16/2017] [Accepted: 09/19/2017] [Indexed: 01/01/2023]
Abstract
Zebrafish are able to completely regrow their caudal fin-folds after amputation. Following injury, wound healing occurs, followed by the formation of a blastema, which produces cells to replace the lost tissue in the final phase of regenerative outgrowth. Here we show that, surprisingly, the phosphatase and tumor suppressor Pten, an antagonist of phosphoinositide-3-kinase (PI3K) signaling, is required for zebrafish caudal fin-fold regeneration. We found that homozygous knock-out mutant (ptena-/-ptenb-/- ) zebrafish embryos, lacking functional Pten, did not regenerate their caudal fin-folds. AKT phosphorylation was enhanced, which is consistent with the function of Pten. Reexpression of Pten, but not catalytically inactive mutant Pten-C124S, rescued regeneration, as did pharmacological inhibition of PI3K. Blastema formation, determined by in situ hybridization for the blastema marker junbb, appeared normal upon caudal fin-fold amputation of ptena-/-ptenb-/- zebrafish embryos. Whole-mount immunohistochemistry using specific markers indicated that proliferation was arrested in embryos lacking functional Pten, and that apoptosis was enhanced. Together, these results suggest a critical role for Pten by limiting PI3K signaling during the regenerative outgrowth phase of zebrafish caudal fin-fold regeneration.
Collapse
Affiliation(s)
- Alexander James Hale
- Hubrecht Institute – KNAW and University Medical Center UtrechtUtrechtThe Netherlands
- Institute Biology LeidenLeiden UniversityLeidenThe Netherlands
| | - Ali Kiai
- Hubrecht Institute – KNAW and University Medical Center UtrechtUtrechtThe Netherlands
| | - Jelte Sikkens
- Hubrecht Institute – KNAW and University Medical Center UtrechtUtrechtThe Netherlands
| | - Jeroen den Hertog
- Hubrecht Institute – KNAW and University Medical Center UtrechtUtrechtThe Netherlands
- Institute Biology LeidenLeiden UniversityLeidenThe Netherlands
| |
Collapse
|
26
|
Chen Z, Xie J, Hao H, Lin H, Wang L, Zhang Y, Chen L, Cao S, Huang X, Liao W, Bin J, Liao Y. Ablation of periostin inhibits post-infarction myocardial regeneration in neonatal mice mediated by the phosphatidylinositol 3 kinase/glycogen synthase kinase 3β/cyclin D1 signalling pathway. Cardiovasc Res 2017; 113:620-632. [PMID: 28453729 PMCID: PMC5412017 DOI: 10.1093/cvr/cvx001] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 09/27/2016] [Accepted: 01/06/2017] [Indexed: 02/06/2023] Open
Abstract
AIMS To resolve the controversy as to whether periostin plays a role in myocardial regeneration after myocardial infarction (MI), we created a neonatal mouse model of MI to investigate the influence of periostin ablation on myocardial regeneration and clarify the underlying mechanisms. METHODS AND RESULTS Neonatal periostin-knockout mice and their wildtype littermates were subjected to MI or sham surgery. In the wildtype mice after MI, fibrosis was detectable at 3 days and fibrotic tissue was completely replaced by regenerated myocardium at 21 days. In contrast, in the knockout mice, significant fibrosis in the infarcted area was present at even 3 weeks after MI. Levels of phosphorylated-histone 3 and aurora B in the myocardium, detected by immunofluorescence and western blotting, were significantly lower in knockout than in wildtype mice at 7 days after MI. Similarly, angiogenesis was decreased in the knockout mice after MI. Expression of both the endothelial marker CD-31 and α-smooth muscle actin was markedly lower in the knockout than in wildtype mice at 7 days after MI. The knockout MI group had elevated levels of glycogen synthase kinase (GSK) 3β and decreased phosphatidylinositol 3-kinase (PI3K), phosphorylated serine/threonine protein kinase B (p-Akt), and cyclin D1, compared with the wildtype MI group. Similar effects were observed in experiments using cultured cardiomyocytes from neonatal wildtype or periostin knockout mice. Administration of SB216763, a GSK3β inhibitor, to knockout neonatal mice decreased myocardial fibrosis and increased angiogenesis in the infarcted area after MI. CONCLUSION Ablation of periostin suppresses post-infarction myocardial regeneration by inhibiting the PI3K/GSK3β/cyclin D1 signalling pathway, indicating that periostin is essential for myocardial regeneration.
Collapse
Affiliation(s)
- Zhenhuan Chen
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838, Guangzhou Avenue North, Guangzhou 510515, China
| | - Jiahe Xie
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838, Guangzhou Avenue North, Guangzhou 510515, China
| | - Huixin Hao
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838, Guangzhou Avenue North, Guangzhou 510515, China
| | - Hairuo Lin
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838, Guangzhou Avenue North, Guangzhou 510515, China
| | - Long Wang
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838, Guangzhou Avenue North, Guangzhou 510515, China
| | - Yingxue Zhang
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838, Guangzhou Avenue North, Guangzhou 510515, China
| | - Lin Chen
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838, Guangzhou Avenue North, Guangzhou 510515, China
| | - Shiping Cao
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838, Guangzhou Avenue North, Guangzhou 510515, China
| | - Xiaobo Huang
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838, Guangzhou Avenue North, Guangzhou 510515, China
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jianping Bin
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838, Guangzhou Avenue North, Guangzhou 510515, China
| | - Yulin Liao
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838, Guangzhou Avenue North, Guangzhou 510515, China
| |
Collapse
|
27
|
Hasegawa T, Hall CJ, Crosier PS, Abe G, Kawakami K, Kudo A, Kawakami A. Transient inflammatory response mediated by interleukin-1β is required for proper regeneration in zebrafish fin fold. eLife 2017; 6:22716. [PMID: 28229859 PMCID: PMC5360449 DOI: 10.7554/elife.22716] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 02/13/2017] [Indexed: 12/19/2022] Open
Abstract
Cellular responses to injury are crucial for complete tissue regeneration, but their underlying processes remain incompletely elucidated. We have previously reported that myeloid-defective zebrafish mutants display apoptosis of regenerative cells during fin fold regeneration. Here, we found that the apoptosis phenotype is induced by prolonged expression of interleukin 1 beta (il1b). Myeloid cells are considered to be the principal source of Il1b, but we show that epithelial cells express il1b in response to tissue injury and initiate the inflammatory response, and that its resolution by macrophages is necessary for survival of regenerative cells. We further show that Il1b plays an essential role in normal fin fold regeneration by regulating expression of regeneration-induced genes. Our study reveals that proper levels of Il1b signaling and tissue inflammation, which are tuned by macrophages, play a crucial role in tissue regeneration. DOI:http://dx.doi.org/10.7554/eLife.22716.001 Animals and other multicellular organisms all have at least some ability to regenerate lost or wounded tissues. Zebrafish are particularly good at this to the extent that they can replace damaged or lost body parts with exact replicas of the originals. In 2015, a team of researchers found that some mutant zebrafish that lack blood cells including immune cells are unable to regenerate lost tissues. This is because the cells that are primed to regenerate die instead, but it was not clear why this happens. Many immune cells have roles in fighting infection and in responding to tissue damage.When a tissue is damaged, the area often becomes inflamed as white blood cells called macrophages flock to the damaged area to protect it from infection and remove damaged cells. Hasegawa et al. – who include several researchers involved in the 2015 study – used genetic approaches to investigate the role of inflammation in tissue regeneration in zebrafish. The experiments show that several genes involved in inflammation – including one called interleukin 1b – were active over longer periods of time in the mutant fish compared with normal zebrafish. The gene produces a signal protein and this prolonged activity causes the primed regenerative cells to die. However, the cells can survive if interleukin 1b activity is quickly suppressed by macrophages. The experiments also show that, in order for tissues to regenerate properly, interleukin 1b needs to be active for only a short period of time. The findings reveal that some inflammation is needed for tissues to regenerate, but that a more severe inflammatory response can block the process. A future challenge will be to identify the signals that macrophages produce to suppress inflammation to allow tissues to regenerate. These anti-inflammatory signals may have the potential to be used as drugs to cure chronic inflammatory diseases and boost tissue regeneration potential in humans. DOI:http://dx.doi.org/10.7554/eLife.22716.002
Collapse
Affiliation(s)
- Tomoya Hasegawa
- School of Bioscience and Biotechnology, Tokyo Institute of Technology, Midori-ku, Japan
| | - Christopher J Hall
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Philip S Crosier
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Gembu Abe
- Division of Molecular and Developmental Biology, National Institute of Genetics, and Department of Genetics, SOKENDAI (The Graduate University for Advanced Studies), Mishima, Japan
| | - Koichi Kawakami
- Division of Molecular and Developmental Biology, National Institute of Genetics, and Department of Genetics, SOKENDAI (The Graduate University for Advanced Studies), Mishima, Japan
| | - Akira Kudo
- School of Bioscience and Biotechnology, Tokyo Institute of Technology, Midori-ku, Japan
| | - Atsushi Kawakami
- School of Bioscience and Biotechnology, Tokyo Institute of Technology, Midori-ku, Japan
| |
Collapse
|
28
|
Ouyang X, Panetta NJ, Talbott MD, Payumo AY, Halluin C, Longaker MT, Chen JK. Hyaluronic acid synthesis is required for zebrafish tail fin regeneration. PLoS One 2017; 12:e0171898. [PMID: 28207787 PMCID: PMC5313160 DOI: 10.1371/journal.pone.0171898] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Accepted: 01/27/2017] [Indexed: 01/20/2023] Open
Abstract
Using genome-wide transcriptional profiling and whole-mount expression analyses of zebrafish larvae, we have identified hyaluronan synthase 3 (has3) as an upregulated gene during caudal fin regeneration. has3 expression is induced in the wound epithelium within hours after tail amputation, and its onset and maintenance requires fibroblast growth factor, phosphoinositide 3-kinase, and transforming growth factor-ß signaling. Inhibition of hyaluronic acid (HA) synthesis by the small molecule 4-methylumbelliferone (4-MU) impairs tail regeneration in zebrafish larvae by preventing injury-induced cell proliferation. In addition, 4-MU reduces the expression of genes associated with wound epithelium and blastema function. Treatment with glycogen synthase kinase 3 inhibitors rescues 4-MU-induced defects in cell proliferation and tail regeneration, while restoring a subset of wound epithelium and blastema markers. Our findings demonstrate a role for HA biosynthesis in zebrafish tail regeneration and delineate its epistatic relationships with other regenerative processes.
Collapse
Affiliation(s)
- Xiaohu Ouyang
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Nicholas J. Panetta
- Department of Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Maya D. Talbott
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Alexander Y. Payumo
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Caroline Halluin
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Michael T. Longaker
- Department of Surgery, Stanford University School of Medicine, Stanford, California, United States of America
- Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, United States of America
| | - James K. Chen
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, California, United States of America
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California, United States of America
| |
Collapse
|
29
|
Tanaka EM. The Molecular and Cellular Choreography of Appendage Regeneration. Cell 2017; 165:1598-1608. [PMID: 27315477 DOI: 10.1016/j.cell.2016.05.038] [Citation(s) in RCA: 156] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 05/02/2016] [Accepted: 05/09/2016] [Indexed: 10/21/2022]
Abstract
Recent advances in limb regeneration are revealing the molecular events that integrate growth control, cell fate programming, and positional information to yield the exquisite replacement of the amputated limb. Parallel progress in several invertebrate and vertebrate models has provided a broader context for understanding the mechanisms and the evolution of regeneration. Together, these discoveries provide a foundation for describing the principles underlying regeneration of complex, multi-tissue structures. As such these findings should provide a wealth of ideas for engineers seeking to reconstitute regeneration from constituent parts or to elicit full regeneration from partial regeneration events.
Collapse
Affiliation(s)
- Elly M Tanaka
- DFG Research Center for Regenerative Therapies, Technische Universität Dresden Fetscherstrasse 105, 01307 Dresden, GERMANY.
| |
Collapse
|
30
|
Fraguas S, Umesono Y, Agata K, Cebrià F. Analyzing pERK Activation During Planarian Regeneration. Methods Mol Biol 2017; 1487:303-315. [PMID: 27924577 DOI: 10.1007/978-1-4939-6424-6_23] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Planarians are an ideal model in which to study stem cell-based regeneration. After amputation, planarian pluripotent stem cells surrounding the wound proliferate to produce the regenerative blastema, in which they differentiate into the missing tissues and structures. Recent independent studies in planarians have shown that Smed-egfr-3, a gene encoding a homologue of epidermal growth factor (EGF) receptors, and DjerkA, which encodes an extracellular signal-regulated kinase (ERK), may control cell differentiation and blastema growth. However, because these studies were carried in two different planarian species, the relationship between these two genes remains unclear. We have optimized anti-pERK immunostaining in Schmidtea mediterranea using the original protocol developed in Dugesia japonica. Both protocols are reported here as most laboratories worldwide work with one of these two species. Using this protocol we have determined that Smed-egfr-3 appears to be necessary for pERK activation during planarian regeneration.
Collapse
Affiliation(s)
- Susanna Fraguas
- Departament de Genètica i Institut de Biomedicina de la Universitat de Barcelona (IBUB), Facultat de Biologia, Universitat de Barcelona, Av. Diagonal 643, Edifici Prevosti, Planta 1, 08028, Barcelona, Catalunya, Spain
| | - Yoshihiko Umesono
- Graduate School of Life Science, University of Hyogo, Ako-gun, Hyogo, Japan
| | - Kiyokazu Agata
- Department of Biophysics, Graduate School of Science, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Francesc Cebrià
- Departament de Genètica i Institut de Biomedicina de la Universitat de Barcelona (IBUB), Facultat de Biologia, Universitat de Barcelona, Av. Diagonal 643, Edifici Prevosti, Planta 1, 08028, Barcelona, Catalunya, Spain.
| |
Collapse
|
31
|
Cardiomyocyte proliferation in zebrafish and mammals: lessons for human disease. Cell Mol Life Sci 2016; 74:1367-1378. [PMID: 27812722 PMCID: PMC5357290 DOI: 10.1007/s00018-016-2404-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Revised: 10/14/2016] [Accepted: 10/28/2016] [Indexed: 01/08/2023]
Abstract
Cardiomyocytes proliferate profusely during early development and for a brief period after birth in mammals. Within a month after birth, this proliferative capability is dramatically reduced in mammals unlike lower vertebrates where it persists into adult life. The zebrafish, for example, retains the ability to regenerate the apex of the heart following resection by a mechanism predominantly driven by cardiomyocyte proliferation. Differences in proliferative capacity of cardiomyocytes in adulthood between mammals and lower vertebrates are closely liked to ontogenetic or phylogenetic factors. Elucidation of these factors has the potential to provide enormous benefits if they lead to the development of therapeutic strategies that facilitate cardiomyocyte proliferation. In this review, we highlight the differences between Mammalian and Zebrafish cardiomyocytes, which could explain at least in part the different proliferative capacities in these two species. We discuss the advantages of the zebrafish as a model of cardiomyocyte proliferation, particularly at the embryonic stage. We also identify a number of key molecular pathways with potential to reveal key steps in switching cardiomyocytes from a quiescent to a proliferative phenotype.
Collapse
|
32
|
Grillo M, Konstantinides N, Averof M. Old questions, new models: unraveling complex organ regeneration with new experimental approaches. Curr Opin Genet Dev 2016; 40:23-31. [DOI: 10.1016/j.gde.2016.05.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Revised: 05/12/2016] [Accepted: 05/13/2016] [Indexed: 10/21/2022]
|
33
|
Uygur A, Lee RT. Mechanisms of Cardiac Regeneration. Dev Cell 2016; 36:362-74. [PMID: 26906733 DOI: 10.1016/j.devcel.2016.01.018] [Citation(s) in RCA: 204] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 01/13/2016] [Accepted: 01/25/2016] [Indexed: 02/07/2023]
Abstract
Adult humans fail to regenerate their hearts following injury, and this failure to regenerate myocardium is a leading cause of heart failure and death worldwide. Although all adult mammals appear to lack significant cardiac regeneration potential, some vertebrates can regenerate myocardium throughout life. In addition, new studies indicate that mammals have cardiac regeneration potential during development and very soon after birth. The mechanisms of heart regeneration among model organisms, including neonatal mice, appear remarkably similar. Orchestrated waves of inflammation, matrix deposition and remodeling, and cardiomyocyte proliferation are commonly seen in heart regeneration models. Understanding why adult mammals develop extensive scarring instead of regeneration is a crucial goal for regenerative biology.
Collapse
Affiliation(s)
- Aysu Uygur
- Department of Stem Cell and Regenerative Biology, Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Harvard Medical School, Harvard University, Cambridge, MA 02139, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Richard T Lee
- Department of Stem Cell and Regenerative Biology, Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Harvard Medical School, Harvard University, Cambridge, MA 02139, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA.
| |
Collapse
|
34
|
King BL, Yin VP. A Conserved MicroRNA Regulatory Circuit Is Differentially Controlled during Limb/Appendage Regeneration. PLoS One 2016; 11:e0157106. [PMID: 27355827 PMCID: PMC4927183 DOI: 10.1371/journal.pone.0157106] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 05/24/2016] [Indexed: 01/25/2023] Open
Abstract
Background Although regenerative capacity is evident throughout the animal kingdom, it is not equally distributed throughout evolution. For instance, complex limb/appendage regeneration is muted in mammals but enhanced in amphibians and teleosts. The defining characteristic of limb/appendage regenerative systems is the formation of a dedifferentiated tissue, termed blastema, which serves as the progenitor reservoir for regenerating tissues. In order to identify a genetic signature that accompanies blastema formation, we employ next-generation sequencing to identify shared, differentially regulated mRNAs and noncoding RNAs in three different, highly regenerative animal systems: zebrafish caudal fins, bichir pectoral fins and axolotl forelimbs. Results These studies identified a core group of 5 microRNAs (miRNAs) that were commonly upregulated and 5 miRNAs that were commonly downregulated, as well as 4 novel tRNAs fragments with sequences conserved with humans. To understand the potential function of these miRNAs, we built a network of 1,550 commonly differentially expressed mRNAs that had functional relationships to 11 orthologous blastema-associated genes. As miR-21 was the most highly upregulated and most highly expressed miRNA in all three models, we validated the expression of known target genes, including the tumor suppressor, pdcd4, and TGFβ receptor subunit, tgfbr2 and novel putative target genes such as the anti-apoptotic factor, bcl2l13, Choline kinase alpha, chka and the regulator of G-protein signaling, rgs5. Conclusions Our extensive analysis of RNA-seq transcriptome profiling studies in three regenerative animal models, that diverged in evolution ~420 million years ago, reveals a common miRNA-regulated genetic network of blastema genes. These comparative studies extend our current understanding of limb/appendage regeneration by identifying previously unassociated blastema genes and the extensive regulation by miRNAs, which could serve as a foundation for future functional studies to examine the process of natural cellular reprogramming in an injury context.
Collapse
Affiliation(s)
- Benjamin L. King
- Kathryn W. Davis Center for Regenerative Biology and Medicine, Mount Desert Island, Biological Laboratory, Salisbury Cove, Maine, United States of America
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, Maine, United States of America
| | - Viravuth P. Yin
- Kathryn W. Davis Center for Regenerative Biology and Medicine, Mount Desert Island, Biological Laboratory, Salisbury Cove, Maine, United States of America
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, Maine, United States of America
- * E-mail:
| |
Collapse
|
35
|
Sánchez M, Ceci ML, Gutiérrez D, Anguita-Salinas C, Allende ML. Mechanosensory organ regeneration in zebrafish depends on a population of multipotent progenitor cells kept latent by Schwann cells. BMC Biol 2016; 14:27. [PMID: 27055439 PMCID: PMC4823859 DOI: 10.1186/s12915-016-0249-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 03/22/2016] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Regenerating damaged tissue is a complex process, requiring progenitor cells that must be stimulated to undergo proliferation, differentiation and, often, migratory behaviors and morphological changes. Multiple cell types, both resident within the damaged tissue and recruited to the lesion site, have been shown to participate. However, the cellular and molecular mechanisms involved in the activation of progenitor cell proliferation and differentiation after injury, and their regulation by different cells types, are not fully understood. The zebrafish lateral line is a suitable system to study regeneration because most of its components are fully restored after damage. The posterior lateral line (PLL) is a mechanosensory system that develops embryonically and is initially composed of seven to eight neuromasts distributed along the trunk and tail, connected by a continuous stripe of interneuromastic cells (INCs). The INCs remain in a quiescent state owing to the presence of underlying Schwann cells. They become activated during development to form intercalary neuromasts. However, no studies have described if INCs can participate in a regenerative event, for example, after the total loss of a neuromast. RESULTS We used electroablation in transgenic larvae expressing fluorescent proteins in PLL components to completely ablate single neuromasts in larvae and adult fish. This injury results in discontinuity of the INCs, Schwann cells, and the PLL nerve. In vivo imaging showed that the INCs fill the gap left after the injury and can regenerate a new neuromast in the injury zone. Further, a single INC is able to divide and form all cell types in a regenerated neuromast and, during this process, it transiently expresses the sox2 gene, a neural progenitor cell marker. We demonstrate a critical role for Schwann cells as negative regulators of INC proliferation and neuromast regeneration, and that this inhibitory property is completely dependent on active ErbB signaling. CONCLUSIONS The potential to regenerate a neuromast after damage requires that progenitor cells (INCs) be temporarily released from an inhibitory signal produced by nearby Schwann cells. This simple yet highly effective two-component niche offers the animal robust mechanisms for organ growth and regeneration, which can be sustained throughout life.
Collapse
Affiliation(s)
- Mario Sánchez
- FONDAP Center for Genome Regulation, Facultad de Ciencias, Universidad de Chile, Casilla 653, Las Palmeras 3425, Santiago, Chile
| | - Maria Laura Ceci
- FONDAP Center for Genome Regulation, Facultad de Ciencias, Universidad de Chile, Casilla 653, Las Palmeras 3425, Santiago, Chile
| | - Daniela Gutiérrez
- FONDAP Center for Genome Regulation, Facultad de Ciencias, Universidad de Chile, Casilla 653, Las Palmeras 3425, Santiago, Chile
| | - Consuelo Anguita-Salinas
- FONDAP Center for Genome Regulation, Facultad de Ciencias, Universidad de Chile, Casilla 653, Las Palmeras 3425, Santiago, Chile
| | - Miguel L Allende
- FONDAP Center for Genome Regulation, Facultad de Ciencias, Universidad de Chile, Casilla 653, Las Palmeras 3425, Santiago, Chile.
| |
Collapse
|
36
|
Abstract
Regeneration involves interactions between multiple signaling pathways acting in a spatially and temporally complex manner. As signaling pathways are highly conserved, understanding how regeneration is controlled in animal models exhibiting robust regenerative capacities should aid efforts to stimulate repair in humans. One way to discover molecular regulators of regeneration is to alter gene/protein function and quantify effect(s) on the regenerative process: dedifferentiation/reprograming, stem/progenitor proliferation, migration/remodeling, progenitor cell differentiation and resolution. A powerful approach for applying this strategy to regenerative biology is chemical genetics, the use of small-molecule modulators of specific targets or signaling pathways. Here, we review advances that have been made using chemical genetics for hypothesis-focused and discovery-driven studies aimed at furthering understanding of how regeneration is controlled.
Collapse
|
37
|
Westcot SE, Hatzold J, Urban MD, Richetti SK, Skuster KJ, Harm RM, Lopez Cervera R, Umemoto N, McNulty MS, Clark KJ, Hammerschmidt M, Ekker SC. Protein-Trap Insertional Mutagenesis Uncovers New Genes Involved in Zebrafish Skin Development, Including a Neuregulin 2a-Based ErbB Signaling Pathway Required during Median Fin Fold Morphogenesis. PLoS One 2015; 10:e0130688. [PMID: 26110643 PMCID: PMC4482254 DOI: 10.1371/journal.pone.0130688] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 05/24/2015] [Indexed: 01/13/2023] Open
Abstract
Skin disorders are widespread, but available treatments are limited. A more comprehensive understanding of skin development mechanisms will drive identification of new treatment targets and modalities. Here we report the Zebrafish Integument Project (ZIP), an expression-driven platform for identifying new skin genes and phenotypes in the vertebrate model Danio rerio (zebrafish). In vivo selection for skin-specific expression of gene-break transposon (GBT) mutant lines identified eleven new, revertible GBT alleles of genes involved in skin development. Eight genes—fras1, grip1, hmcn1, msxc, col4a4, ahnak, capn12, and nrg2a—had been described in an integumentary context to varying degrees, while arhgef25b, fkbp10b, and megf6a emerged as novel skin genes. Embryos homozygous for a GBT insertion within neuregulin 2a (nrg2a) revealed a novel requirement for a Neuregulin 2a (Nrg2a) – ErbB2/3 – AKT signaling pathway governing the apicobasal organization of a subset of epidermal cells during median fin fold (MFF) morphogenesis. In nrg2a mutant larvae, the basal keratinocytes within the apical MFF, known as ridge cells, displayed reduced pAKT levels as well as reduced apical domains and exaggerated basolateral domains. Those defects compromised proper ridge cell elongation into a flattened epithelial morphology, resulting in thickened MFF edges. Pharmacological inhibition verified that Nrg2a signals through the ErbB receptor tyrosine kinase network. Moreover, knockdown of the epithelial polarity regulator and tumor suppressor lgl2 ameliorated the nrg2a mutant phenotype. Identifying Lgl2 as an antagonist of Nrg2a – ErbB signaling revealed a significantly earlier role for Lgl2 during epidermal morphogenesis than has been described to date. Furthermore, our findings demonstrated that successive, coordinated ridge cell shape changes drive apical MFF development, making MFF ridge cells a valuable model for investigating how the coordinated regulation of cell polarity and cell shape changes serves as a crucial mechanism of epithelial morphogenesis.
Collapse
Affiliation(s)
- Stephanie E. Westcot
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota, United States of America
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Julia Hatzold
- Institute for Developmental Biology, University of Cologne, Biocenter, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Mark D. Urban
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Stefânia K. Richetti
- Institute for Developmental Biology, University of Cologne, Biocenter, Cologne, Germany
| | - Kimberly J. Skuster
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Rhianna M. Harm
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Roberto Lopez Cervera
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Noriko Umemoto
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Melissa S. McNulty
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Karl J. Clark
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Matthias Hammerschmidt
- Institute for Developmental Biology, University of Cologne, Biocenter, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Cologne Cluster of Excellence in Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Stephen C. Ekker
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota, United States of America
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
38
|
Lush ME, Piotrowski T. ErbB expressing Schwann cells control lateral line progenitor cells via non-cell-autonomous regulation of Wnt/β-catenin. eLife 2014; 3:e01832. [PMID: 24642408 PMCID: PMC3957165 DOI: 10.7554/elife.01832] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 02/13/2014] [Indexed: 12/31/2022] Open
Abstract
Proper orchestration of quiescence and activation of progenitor cells is crucial during embryonic development and adult homeostasis. We took advantage of the zebrafish sensory lateral line to define niche-progenitor interactions to understand how integration of diverse signaling pathways spatially and temporally regulates the coordination of these processes. Our previous studies demonstrated that Schwann cells play a crucial role in negatively regulating lateral line progenitor proliferation. Here we demonstrate that ErbB/Neuregulin signaling is not only required for Schwann cell migration but that it plays a continued role in postmigratory Schwann cells. ErbB expressing Schwann cells inhibit lateral line progenitor proliferation and differentiation through non-cell-autonomous inhibition of Wnt/β-catenin signaling. Subsequent activation of Fgf signaling controls sensory organ differentiation, but not progenitor proliferation. In addition to the lateral line, these findings have important implications for understanding how niche-progenitor cells segregate interactions during development, and how they may go wrong in disease states. DOI: http://dx.doi.org/10.7554/eLife.01832.001.
Collapse
Affiliation(s)
- Mark E Lush
- Stowers Institute for Medical Research, Kansas City, United States
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, United States
| | - Tatjana Piotrowski
- Stowers Institute for Medical Research, Kansas City, United States
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, United States
| |
Collapse
|
39
|
Moore AC, Mark TE, Hogan AK, Topczewski J, LeClair EE. Peripheral axons of the adult zebrafish maxillary barbel extensively remyelinate during sensory appendage regeneration. J Comp Neurol 2013; 520:4184-203. [PMID: 22592645 DOI: 10.1002/cne.23147] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Myelination is a cellular adaptation allowing rapid conduction along axons. We have investigated peripheral axons of the zebrafish maxillary barbel (ZMB), an optically clear sensory appendage. Each barbel carries taste buds, solitary chemosensory cells, and epithelial nerve endings, all of which regenerate after amputation (LeClair and Topczewski [2010] PLoS One 5:e8737). The ZMB contains axons from the facial nerve; however, myelination within the barbel itself has not been established. Transcripts of myelin basic protein (mbp) are expressed in normal and regenerating adult barbels, indicating activity in both maintenance and repair. Myelin was confirmed in situ by using toluidine blue, an anti-MBP antibody, and transmission electron microscopy (TEM). The adult ZMB contains ∼180 small-diameter axons (<2 μm), approximately 60% of which are myelinated. Developmental myelination was observed via whole-mount immunohistochemistry 4-6 weeks postfertilization, showing myelin sheaths lagging behind growing axons. Early-regenerating axons (10 days postsurgery), having no or few myelin layers, were disorganized within a fibroblast-rich collagenous scar. Twenty-eight days postsurgery, barbel axons had grown out several millimeters and were organized with compact myelin sheaths. Fiber types and axon areas were similar between normal and regenerated tissue; within 4 weeks, regenerating axons restored ∼85% of normal myelin thickness. Regenerating barbels express multiple promyelinating transcription factors (sox10, oct6 = pou3f1; krox20a/b = egr2a/b) typical of Schwann cells. These observations extend our understanding of the zebrafish peripheral nervous system within a little-studied sensory appendage. The accessible ZMB provides a novel context for studying axon regeneration, Schwann cell migration, and remyelination in a model vertebrate.
Collapse
Affiliation(s)
- Alex C Moore
- Department of Biological Sciences, DePaul University, Chicago, Illinois 60614, USA
| | | | | | | | | |
Collapse
|
40
|
Dooley CM, Mongera A, Walderich B, Nüsslein-Volhard C. On the embryonic origin of adult melanophores: the role of ErbB and Kit signalling in establishing melanophore stem cells in zebrafish. Development 2013; 140:1003-13. [PMID: 23364329 DOI: 10.1242/dev.087007] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Pigment cells in vertebrates are derived from the neural crest (NC), a pluripotent and migratory embryonic cell population. In fishes, larval melanophores develop during embryogenesis directly from NC cells migrating along dorsolateral and ventromedial paths. The embryonic origin of the melanophores that emerge during juvenile development in the skin to contribute to the striking colour patterns of adult fishes remains elusive. We have identified a small set of melanophore progenitor cells (MPs) in the zebrafish (Danio rerio, Cyprinidae) that is established within the first 2 days of embryonic development in close association with the segmentally reiterated dorsal root ganglia (DRGs). Lineage analysis and 4D in vivo imaging indicate that progeny of these embryonic MPs spread segmentally, giving rise to the melanophores that create the adult melanophore stripes. Upon depletion of larval melanophores by morpholino knockdown of Mitfa, the embryonic MPs are prematurely activated; their progeny migrate along the spinal nerves restoring the larval pattern and giving rise to postembryonic MPs associated with the spinal nerves. Mutational or chemical inhibition of ErbB receptors blocks all early NC migration along the ventromedial path, causing a loss of DRGs and embryonic MPs. We show that the sparse like (slk) mutant lacks larval and metamorphic melanophores and identify kit ligand a (kitlga) as the underlying gene. Our data suggest that kitlga is required for the establishment or survival of embryonic MPs. We propose a model in which DRGs provide a niche for the stem cells of adult melanophores.
Collapse
Affiliation(s)
- Christopher M Dooley
- Max-Planck-Institut für Entwicklungsbiologie, Spemannstr 35, 72076 Tübingen, Germany
| | | | | | | |
Collapse
|
41
|
Challa AK, Chatti K. Conservation and early expression of zebrafish tyrosine kinases support the utility of zebrafish as a model for tyrosine kinase biology. Zebrafish 2012; 10:264-74. [PMID: 23234507 DOI: 10.1089/zeb.2012.0781] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Tyrosine kinases have significant roles in cell growth, apoptosis, development, and disease. To explore the use of zebrafish as a vertebrate model for tyrosine kinase signaling and to better understand their roles, we have identified all of the tyrosine kinases encoded in the zebrafish genome and quantified RNA expression of selected tyrosine kinases during early development. Using profile hidden Markov model analysis, we identified 122 zebrafish tyrosine kinase genes and proposed unambiguous gene names where needed. We found them to be organized into 39 nonreceptor and 83 receptor type, and 30 families consistent with human tyrosine kinase family assignments. We found five human tyrosine kinase genes (epha1, bmx, fgr, srm, and insrr) with no identifiable zebrafish ortholog, and one zebrafish gene (yrk) with no identifiable human ortholog. We also found that receptor tyrosine kinase genes were duplicated more often than nonreceptor tyrosine kinase genes in zebrafish. We profiled expression levels of 30 tyrosine kinases representing all families using direct digital detection at different stages during the first 24 hours of development. The profiling experiments clearly indicate regulated expression of tyrosine kinases in the zebrafish, suggesting their role during early embryonic development. In summary, our study has resulted in the first comprehensive description of the zebrafish tyrosine kinome.
Collapse
Affiliation(s)
- Anil Kumar Challa
- Dr. Reddy's Institute of Life Sciences, University of Hyderabad Campus, Hyderabad, India
| | | |
Collapse
|
42
|
Abstract
Transgenesis promises a powerful means for assessing gene function during amphibian limb regeneration. This approach is complicated, however, by the need for embryonic appendage development to proceed unimpeded despite the genetic alterations one wishes to test later in the context of regeneration. Achieving conditional gene regulation in this amphibian has not proved to be as straightforward as in many other systems. In this report we describe a unique method for obtaining temporal control over exogenous gene expression in the axolotl. Based on technology derived from the Escherichia coli Lac operon, uninduced transgenes are kept in a repressed state by the binding of constitutively expressed Lac repressor protein (LacI) to operator sequences within the expression construct. Addition of a lactose analog, IPTG, to the swimming water of the axolotl is sufficient for the sugar to be taken up by cells, where it binds the LacI protein, thereby inducing expression of the repressed gene. We use this system to demonstrate an in vivo role for thrombospondin-4 in limb regeneration. This inducible system will allow for systematic analysis of phenotypes at defined developmental or regenerative time points. The tight regulation and robustness of gene induction combined with the simplicity of this strategy will prove invaluable for studying many aspects of axolotl biology.
Collapse
|
43
|
Tal T, Sengupta S, Tanguay RL. Caudal Fin Regeneration in Zebrafish. Zebrafish 2011. [DOI: 10.1002/9781118102138.ch22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
44
|
Regenerative phenotype in mice with a point mutation in transforming growth factor beta type I receptor (TGFBR1). Proc Natl Acad Sci U S A 2011; 108:14560-5. [PMID: 21841138 DOI: 10.1073/pnas.1111056108] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Regeneration of peripheral differentiated tissue in mammals is rare, and regulators of this process are largely unknown. We carried out a forward genetic screen in mice using N-ethyl-N-nitrosourea mutagenesis to identify genetic mutations that affect regenerative healing in vivo. More than 400 pedigrees were screened for closure of a through-and-through punch wound in the mouse ear. This led to the identification of a single pedigree with a heritable, fast, and regenerative wound-healing phenotype. Within 5 wk after ear-punch, a threefold decrease in the diameter of the wound was observed in the mutant mice compared with the wild-type mice. At 22 wk, new cartilage, hair follicles, and sebaceous glands were observed in the newly generated tissue. This trait was mapped to a point mutation in a receptor for TGF-β, TGFBR1. Mouse embryonic fibroblasts from the affected mice had increased expression of a subset of TGF-β target genes, suggesting that the mutation caused partial activation of the receptor. Further, bone marrow stromal cells from the mutant mice more readily differentiated to chondrogenic precursors, providing a plausible explanation for the enhanced development of cartilage islands in the regenerated ears. This mutant mouse strain provides a unique model to further explore regeneration in mammals and, in particular, the role of TGFBR1 in chondrogenesis and regenerative wound healing.
Collapse
|
45
|
Yoshinari N, Kawakami A. Mature and juvenile tissue models of regeneration in small fish species. THE BIOLOGICAL BULLETIN 2011; 221:62-78. [PMID: 21876111 DOI: 10.1086/bblv221n1p62] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
The multitude of cells constituting organisms are fragile and easily damaged day by day. Therefore, maintenance of tissue morphology and function is fundamental for multicellular organisms to attain long life. For proper maintenance of tissue integrity, organisms must have mechanisms that detect the loss of tissue mass, activate the de novo production of cells, and organize those cells into functional tissues. However, these processes are only poorly understood. Here we give an overview of adult and juvenile tissue regeneration models in small fish species, such as zebrafish and medaka, and highlight recent advances at the molecular level. From these advances, we have come to realize that the epidermal and mesenchymal parts of the regenerating fish fin-that is, the wound epidermis and blastema, respectively-comprise heterogeneous populations of cells with different molecular identities that can be termed "compartments." These compartments and their mutual interactions are thought to play important roles in promoting the proper progression of tissue regeneration. We further describe the current understanding of these compartments and discuss the possible approaches to affording a better understanding of their roles and interactions during regeneration.
Collapse
Affiliation(s)
- Nozomi Yoshinari
- Department of Biological Information, Tokyo Institute of Technology, Midori-ku, Yokohama, Japan
| | | |
Collapse
|
46
|
Fraguas S, Barberán S, Cebrià F. EGFR signaling regulates cell proliferation, differentiation and morphogenesis during planarian regeneration and homeostasis. Dev Biol 2011; 354:87-101. [PMID: 21458439 DOI: 10.1016/j.ydbio.2011.03.023] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2010] [Revised: 02/14/2011] [Accepted: 03/23/2011] [Indexed: 02/08/2023]
Abstract
Similarly to development, the process of regeneration requires that cells accurately sense and respond to their external environment. Thus, intrinsic cues must be integrated with signals from the surrounding environment to ensure appropriate temporal and spatial regulation of tissue regeneration. Identifying the signaling pathways that control these events will not only provide insights into a fascinating biological phenomenon but may also yield new molecular targets for use in regenerative medicine. Among classical models to study regeneration, freshwater planarians represent an attractive system in which to investigate the signals that regulate cell proliferation and differentiation, as well as the proper patterning of the structures being regenerated. Recent studies in planarians have begun to define the role of conserved signaling pathways during regeneration. Here, we extend these analyses to the epidermal growth factor (EGF) receptor pathway. We report the characterization of three epidermal growth factor (EGF) receptors in the planarian Schmidtea mediterranea. Silencing of these genes by RNA interference (RNAi) yielded multiple defects in intact and regenerating planarians. Smed-egfr-1(RNAi) resulted in decreased differentiation of eye pigment cells, abnormal pharynx regeneration and maintenance, and the development of dorsal outgrowths. In contrast, Smed-egfr-3(RNAi) animals produced smaller blastemas associated with abnormal differentiation of certain cell types. Our results suggest important roles for the EGFR signaling in controlling cell proliferation, differentiation and morphogenesis during planarian regeneration and homeostasis.
Collapse
Affiliation(s)
- Susanna Fraguas
- Department of Genetics, Faculty of Biology, University of Barcelona and Institute of Biomedicine of the University of Barcelona (IBUB), Av. Diagonal 645, Barcelona, Catalunya, Spain
| | | | | |
Collapse
|
47
|
Bando T, Mito T, Nakamura T, Ohuchi H, Noji S. Regulation of leg size and shape: Involvement of the Dachsous-fat signaling pathway. Dev Dyn 2011; 240:1028-41. [DOI: 10.1002/dvdy.22590] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/25/2011] [Indexed: 11/11/2022] Open
|
48
|
Blassberg RA, Garza-Garcia A, Janmohamed A, Gates PB, Brockes JP. Functional convergence of signalling by GPI-anchored and anchorless forms of a salamander protein implicated in limb regeneration. J Cell Sci 2011; 124:47-56. [PMID: 21118959 PMCID: PMC3001407 DOI: 10.1242/jcs.076331] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/06/2010] [Indexed: 01/12/2023] Open
Abstract
The GPI-anchor is an established determinant of molecular localisation and various functional roles have been attributed to it. The newt GPI-anchored three-finger protein (TFP) Prod1 is an important regulator of cell behaviour during limb regeneration, but it is unclear how it signals to the interior of the cell. Prod1 was expressed by transfection in cultured newt limb cells and activated transcription and expression of matrix metalloproteinase 9 (MMP9) by a pathway involving ligand-independent activation of epidermal growth factor receptor (EGFR) signalling and phosphorylation of extracellular regulated kinase 1 and 2 (ERK1/2). This was dependent on the presence of the GPI-anchor and critical residues in the α-helical region of the protein. Interestingly, Prod1 in the axolotl, a salamander species that also regenerates its limbs, was shown to activate ERK1/2 signalling and MMP9 transcription despite being anchorless, and both newt and axolotl Prod1 co-immunoprecipitated with the newt EGFR after transfection. The substitution of the axolotl helical region activated a secreted, anchorless version of the newt molecule. The activity of the newt molecule cannot therefore depend on a unique property conferred by the anchor. Prod1 is a salamander-specific TFP and its interaction with the phylogenetically conserved EGFR has implications for our view of regeneration as an evolutionary variable.
Collapse
Affiliation(s)
- Robert A. Blassberg
- Institute of Structural and Molecular Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Acely Garza-Garcia
- Division of Molecular Structure, MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, UK
| | - Azara Janmohamed
- Institute of Structural and Molecular Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Phillip B. Gates
- Institute of Structural and Molecular Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Jeremy P. Brockes
- Institute of Structural and Molecular Biology, University College London, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
49
|
Paranjpe S, Bowen WC, Tseng GC, Luo JH, Orr A, Michalopoulos GK. RNA interference against hepatic epidermal growth factor receptor has suppressive effects on liver regeneration in rats. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 176:2669-81. [PMID: 20395437 PMCID: PMC2877830 DOI: 10.2353/ajpath.2010.090605] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Accepted: 02/04/2010] [Indexed: 02/06/2023]
Abstract
Liver regeneration after a two-thirds partial hepatectomy (PHx) is a complex process requiring interaction and cooperation of many growth factors and cytokines and cross talk between multiple pathways. Along with hepatocyte growth factor and its receptor MET (HGF-MET), the epidermal growth factor receptor (EGFR) signaling pathway is activated within 60 minutes after PHx. To investigate the role of EGFR in liver regeneration, we used two EGFR-specific short hairpin silencing RNAs to inhibit EGFR expression in regenerating normal rat liver. Suppression of EGFR mRNA and protein was evident in treated rats. There was also a demonstrable decrease but not complete elimination of bromo-deoxyuridine incorporation and mitoses at 24 hours after PHx. In addition, we observed up-regulation of MET and Src as well as activation of the ErbB-3-ErbB-2-PI3K-Akt pathway and down-regulation of STAT 3, cyclin D1, cyclin E1, p21, and C/EBP beta. The decrease in the ratio of C/EBP alpha to C/EBP beta known to occur after PHx was offset in shEGFR-treated rats. Despite suppression of hepatocyte proliferation lasting into day 3 after PHx, liver weight restoration occurred. Interestingly, hepatocytes in shEGFR-treated rats were considerably larger when compared with ScrRNA-treated controls. The data indicate that although the MET and EGFR pathways are similar, the contributions made by MET and EGFR are unique and are not compensated by each other or other cytokines.
Collapse
Affiliation(s)
- Shirish Paranjpe
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | | | | | | |
Collapse
|
50
|
Phosphorylation of Junb family proteins by the Jun N-terminal kinase supports tissue regeneration in zebrafish. Dev Biol 2010; 340:468-79. [DOI: 10.1016/j.ydbio.2010.01.036] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2009] [Revised: 01/28/2010] [Accepted: 01/29/2010] [Indexed: 01/22/2023]
|