1
|
Adell T, Cebrià F, Abril JF, Araújo SJ, Corominas M, Morey M, Serras F, González-Estévez C. Cell death in regeneration and cell turnover: Lessons from planarians and Drosophila. Semin Cell Dev Biol 2025; 169:103605. [PMID: 40139139 DOI: 10.1016/j.semcdb.2025.103605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 02/27/2025] [Accepted: 03/09/2025] [Indexed: 03/29/2025]
Abstract
Programmed cell death plays a crucial role during tissue turnover in all animal species, and it is also essential during regeneration, serving as a key signalling mechanism to promote tissue repair and regrowth. In freshwater planarians, remarkable regenerative abilities are supported by neoblasts, a population of adult stem cells, which enable high somatic cell turnover. Cell death in planarians occurs continuously during regeneration and adult homeostasis, underscoring its critical role in tissue remodeling and repair. However, the exact mechanisms regulating cell death in these organisms remain elusive. In contrast, Drosophila melanogaster serves as a powerful model for studying programmed cell death in development, metamorphosis, and adult tissue maintenance, leveraging advanced genetic tools and visualization techniques. In Drosophila, cell death sculpts tissues, eliminates larval structures during metamorphosis, and supports homeostasis in adulthood. Despite limited regenerative capacity compared to planarians, Drosophila provides unique insights into cell death's regulatory mechanisms. Comparative analysis of these two systems highlights both conserved and divergent roles of programmed cell death in tissue renewal and regeneration. This review synthesizes the latest knowledge of programmed cell death in planarians and Drosophila, aiming to illuminate shared principles and system-specific adaptations, with relevance to tissue repair across biological systems.
Collapse
Affiliation(s)
- Teresa Adell
- Department of Genetics, Microbiology and Statistics, School of Biology and Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Av. Diagonal 643, Edifici Prevosti 1st floor, Barcelona 08028, Spain.
| | - Francesc Cebrià
- Department of Genetics, Microbiology and Statistics, School of Biology and Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Av. Diagonal 643, Edifici Prevosti 1st floor, Barcelona 08028, Spain
| | - Josep F Abril
- Department of Genetics, Microbiology and Statistics, School of Biology and Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Av. Diagonal 643, Edifici Prevosti 1st floor, Barcelona 08028, Spain
| | - Sofia J Araújo
- Department of Genetics, Microbiology and Statistics, School of Biology and Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Av. Diagonal 643, Edifici Prevosti 1st floor, Barcelona 08028, Spain
| | - Montserrat Corominas
- Department of Genetics, Microbiology and Statistics, School of Biology and Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Av. Diagonal 643, Edifici Prevosti 1st floor, Barcelona 08028, Spain
| | - Marta Morey
- Department of Genetics, Microbiology and Statistics, School of Biology and Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Av. Diagonal 643, Edifici Prevosti 1st floor, Barcelona 08028, Spain
| | - Florenci Serras
- Department of Genetics, Microbiology and Statistics, School of Biology and Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Av. Diagonal 643, Edifici Prevosti 1st floor, Barcelona 08028, Spain
| | - Cristina González-Estévez
- Department of Genetics, Microbiology and Statistics, School of Biology and Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Av. Diagonal 643, Edifici Prevosti 1st floor, Barcelona 08028, Spain.
| |
Collapse
|
2
|
Booth CLT, Stevens BC, Stubbert CA, Kallgren NT, Deihl EW, Davies EL. Developmental onset of planarian whole-body regeneration depends on axis reset. Curr Biol 2025:S0960-9822(25)00381-1. [PMID: 40239657 DOI: 10.1016/j.cub.2025.03.065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 02/11/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025]
Abstract
Regenerative abilities vary across species and developmental stages of animal life cycles. Determining mechanisms that promote or limit regeneration in certain life cycle stages may pinpoint the most critical factors for successful regeneration and suggest strategies for reverse-engineering regenerative responses in therapeutic settings. In contrast to many mammalian systems, which typically show a loss of regenerative abilities with age, planarian flatworms remain highly regenerative throughout adulthood. The robust reproductive and regenerative capabilities of the planarian Schmidtea polychroa (S. polychroa) make them an ideal model to determine when and how regeneration competence is established during development. We report that S. polychroa gradually acquires whole-body regenerative abilities during late embryonic and early juvenile stages. Anterior fragments are capable of regenerating missing trunk and tail tissues from stage 6.5 onward. By contrast, the ability of posterior fragments to make new head tissue depends on the developmental stage, tissue composition of the amputated fragment, and axial position of the cut plane. Irradiation-sensitive cells are required, but not sufficient, for the onset of head regeneration ability. We propose that regulation of the main body axis reset, specifically the ability to remake an anterior organizing center, determines when whole-body regeneration competence arises during development. Supporting this hypothesis, knockdown of the canonical Wnt pathway effector Spol-β-catenin-1, a posterior determinant, induces precocious head regeneration under conditions that are normally head regeneration-incompetent. Our results suggest that regeneration competence emerges through interactions between irradiation-sensitive cells, the cellular source of new tissue, and developing adult tissue(s) harboring axial patterning information.
Collapse
Affiliation(s)
- Clare L T Booth
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21704, USA; Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - Brian C Stevens
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21704, USA; Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
| | - Clover A Stubbert
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21704, USA; Molecular Biology Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Neil T Kallgren
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21704, USA
| | - Ennis W Deihl
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21704, USA
| | - Erin L Davies
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21704, USA.
| |
Collapse
|
3
|
Tang SKY, Marshall WF. Physical Forces in Regeneration of Cells and Tissues. Cold Spring Harb Perspect Biol 2025; 17:a041527. [PMID: 38806241 PMCID: PMC11602525 DOI: 10.1101/cshperspect.a041527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
The ability to regenerate after the loss of a part is a hallmark of living systems and occurs at both the tissue and organ scales, but also within individual cells. Regeneration entails many processes that are physical and mechanical in nature, including the closure of wounds, the repositioning of material from one place to another, and the restoration of symmetry following perturbations. However, we currently know far more about the genetics and molecular signaling pathways involved in regeneration, and there is a need to investigate the role of physical forces in the process. Here, we will provide an overview of how physical forces may play a role in wound healing and regeneration, in which we compare and contrast regenerative processes at the tissue and cell scales.
Collapse
Affiliation(s)
- Sindy K Y Tang
- Department of Mechanical Engineering, Stanford University, Stanford, California 94305-3030, USA
| | - Wallace F Marshall
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, California 94158-2517, USA
| |
Collapse
|
4
|
Cheung S, Bredikhin D, Gerber T, Steenbergen PJ, Basu S, Bailleul R, Hansen P, Paix A, Benton MA, Korswagen HC, Arendt D, Stegle O, Ikmi A. Systemic coordination of whole-body tissue remodeling during local regeneration in sea anemones. Dev Cell 2025; 60:780-793.e7. [PMID: 39615481 DOI: 10.1016/j.devcel.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 08/06/2024] [Accepted: 11/01/2024] [Indexed: 03/14/2025]
Abstract
The complexity of regeneration extends beyond local wound responses, eliciting systemic processes across the entire organism. However, the functional relevance and coordination of distant molecular processes remain unclear. In the cnidarian Nematostella vectensis, we show that local regeneration triggers a systemic homeostatic response, leading to coordinated whole-body remodeling. Leveraging spatial transcriptomics, endogenous protein tagging, and live imaging, we comprehensively dissect this systemic response at the organismal scale. We identify proteolysis as a critical process driven by both local and systemic upregulation of metalloproteases. We show that metalloproteinase expression levels and activity scale with the extent of tissue loss. This proportional response drives long-range tissue and extracellular matrix movement. Our findings demonstrate the adaptive nature of the systematic response in regeneration, enabling the organism to maintain shape homeostasis while coping with a wide range of injuries.
Collapse
Affiliation(s)
- Stephanie Cheung
- European Molecular Biology Laboratory (EMBL), Developmental Biology Unit, Heidelberg 69117, Germany
| | - Danila Bredikhin
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg 69117, Germany; Division of Computational Genomics and Systems Genetics, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Tobias Gerber
- European Molecular Biology Laboratory (EMBL), Developmental Biology Unit, Heidelberg 69117, Germany; European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg 69117, Germany
| | - Petrus J Steenbergen
- European Molecular Biology Laboratory (EMBL), Developmental Biology Unit, Heidelberg 69117, Germany
| | - Soham Basu
- European Molecular Biology Laboratory (EMBL), Developmental Biology Unit, Heidelberg 69117, Germany
| | - Richard Bailleul
- European Molecular Biology Laboratory (EMBL), Developmental Biology Unit, Heidelberg 69117, Germany
| | - Pauline Hansen
- European Molecular Biology Laboratory (EMBL), Developmental Biology Unit, Heidelberg 69117, Germany
| | - Alexandre Paix
- European Molecular Biology Laboratory (EMBL), Developmental Biology Unit, Heidelberg 69117, Germany
| | - Matthew A Benton
- European Molecular Biology Laboratory (EMBL), Developmental Biology Unit, Heidelberg 69117, Germany
| | - Hendrik C Korswagen
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | - Detlev Arendt
- European Molecular Biology Laboratory (EMBL), Developmental Biology Unit, Heidelberg 69117, Germany
| | - Oliver Stegle
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg 69117, Germany; Division of Computational Genomics and Systems Genetics, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany; Faculty of Biosciences, Heidelberg University, Heidelberg, Germany; Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton CB10 1SA, Cambridgeshire, UK.
| | - Aissam Ikmi
- European Molecular Biology Laboratory (EMBL), Developmental Biology Unit, Heidelberg 69117, Germany.
| |
Collapse
|
5
|
Canales BII, King HO, Reddien PW. map3k1 is required for spatial restriction of progenitor differentiation in planarians. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.04.641450. [PMID: 40093160 PMCID: PMC11908231 DOI: 10.1101/2025.03.04.641450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Planarian regeneration and tissue turnover involve fate specification in pluripotent stem cells called neoblasts. Neoblasts select fates through the expression of fate-specific transcription factors (FSTFs), generating specialized neoblasts. Specialized neoblasts are spatially intermingled and can be dispersed broadly, frequently being far from their target tissue. The post-mitotic progeny of neoblasts, serving as progenitors, migrate and differentiate into mature cell types. Pattern formation is thus strongly influenced by the migratory assortment and differentiation of fate-specified progenitors in precise locations, which we refer to as progenitor targeting. This central step of pattern maintenance and formation, however, is poorly understood. Here, we describe a requirement for the conserved map3k1 gene in targeting, restricting post-mitotic progenitor differentiation to precise locations. RNAi of map3k1 causes ectopic differentiation of eye progenitors along their migratory path, resulting in dispersed ectopic eyes and eye cells. Other neural tissues similarly display ectopic posterior differentiation and pharynx cells emerge dispersed laterally and anteriorly in map3k1 RNAi animals. Ectopic differentiated cells are also found within the incorrect organs after map3k1 RNAi, and ultimately teratomas form. These findings implicate map3k1 signaling in controlling the positional regulation of progenitor behavior - restricting progenitor differentiation to targeted locations in response to external cues in the local tissue environment.
Collapse
Affiliation(s)
- Bryanna Isela-Inez Canales
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Hunter O King
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Peter W Reddien
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
6
|
Guo W, Liu X, Pang L, Kong Z, Lin Z, Ren J, Dong Z, Chen G, Liu D. DjsoxP-1 and Djsox5 are essential for tissue homeostasis and regeneration in Dugesia japonica. Cell Tissue Res 2025; 399:337-350. [PMID: 39762587 DOI: 10.1007/s00441-024-03939-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 11/15/2024] [Indexed: 03/01/2025]
Abstract
Sox genes encode a family of transcription factors that regulate multiple biological processes during metazoan development, including embryogenesis, tissue homeostasis, nervous system specification, and stem cell maintenance. The planarian Dugesia japonica contains a reservoir of stem cells that grow and divide continuously to support cellular turnover. However, whether SOX proteins retain these conserved functions in planarians remains to be determined. In this study, three sox gene homologs, DjsoxP-1, DjsoxP-5, and Djsox5, were identified in the planarian transcriptome, and their roles were investigated. The results showed that the amino acids deduced from the three sox genes all contained high-mobility group (HMG) domain sequences, which are highly conserved in sox family members. Whole-mount in situ hybridization (WISH) and real-time quantitative PCR (RT-qPCR) results indicated that the three sox genes were mainly expressed in parenchymal tissues and regenerative blastema. Additionally, X-ray irradiation assay and dFISH suggested that the three Djsox genes were expressed in neoblasts and other cell types. Head regression in intact planarian and smaller blastemas in both head or tail fragments of regenerating planarians were exhibited with DjsoxP-1 and Djsox5 RNA interference (RNAi) compared to the control animals, suggesting that DjsoxP-1 and Djsox5 have essential roles during cellular turnover and regeneration in planarians; conversely, there was no obvious phenotypic abnormalities or regeneration defect in DjsoxP-5 RNAi animals. Knockdown of DjsoxP-1 or Djsox5 decreased neoblast proliferation and promoted cell apoptosis. In conclusion, our findings demonstrate that DjsoxP-1 and Djsox5 are involved in cellular turnover and regeneration in planarians by modulating coordination between cell proliferation and apoptosis.
Collapse
Affiliation(s)
- Weiyun Guo
- College of Life Science, Henan Normal University, No. 46, Jianshe Road, Xinxiang 453007, Henan, China
- School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang 453002, Henan, China
| | - Xiao Liu
- College of Life Science, Henan Normal University, No. 46, Jianshe Road, Xinxiang 453007, Henan, China
| | - Lina Pang
- College of Life Science, Henan Normal University, No. 46, Jianshe Road, Xinxiang 453007, Henan, China
| | - Zhihong Kong
- College of Life Science, Henan Normal University, No. 46, Jianshe Road, Xinxiang 453007, Henan, China
| | - Ziyi Lin
- College of Life Science, Henan Normal University, No. 46, Jianshe Road, Xinxiang 453007, Henan, China
| | - Jing Ren
- College of Life Science, Henan Normal University, No. 46, Jianshe Road, Xinxiang 453007, Henan, China
| | - Zimei Dong
- College of Life Science, Henan Normal University, No. 46, Jianshe Road, Xinxiang 453007, Henan, China.
| | - Guangwen Chen
- College of Life Science, Henan Normal University, No. 46, Jianshe Road, Xinxiang 453007, Henan, China.
| | - Dezeng Liu
- College of Life Science, Henan Normal University, No. 46, Jianshe Road, Xinxiang 453007, Henan, China
| |
Collapse
|
7
|
Moreno-Blas D, Adell T, González-Estévez C. Autophagy in Tissue Repair and Regeneration. Cells 2025; 14:282. [PMID: 39996754 PMCID: PMC11853389 DOI: 10.3390/cells14040282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 02/01/2025] [Accepted: 02/13/2025] [Indexed: 02/26/2025] Open
Abstract
Autophagy is a cellular recycling system that, through the sequestration and degradation of intracellular components regulates multiple cellular functions to maintain cellular homeostasis and survival. Dysregulation of autophagy is closely associated with the development of physiological alterations and human diseases, including the loss of regenerative capacity. Tissue regeneration is a highly complex process that relies on the coordinated interplay of several cellular processes, such as injury sensing, defense responses, cell proliferation, differentiation, migration, and cellular senescence. These processes act synergistically to repair or replace damaged tissues and restore their morphology and function. In this review, we examine the evidence supporting the involvement of the autophagy pathway in the different cellular mechanisms comprising the processes of regeneration and repair across different regenerative contexts. Additionally, we explore how modulating autophagy can enhance or accelerate regeneration and repair, highlighting autophagy as a promising therapeutic target in regenerative medicine for the development of autophagy-based treatments for human diseases.
Collapse
Affiliation(s)
| | | | - Cristina González-Estévez
- Department of Genetics, Microbiology and Statistics, School of Biology and Institute of Biomedicine (IBUB), University of Barcelona, Av. Diagonal, 643, 08028 Barcelona, Spain; (D.M.-B.); (T.A.)
| |
Collapse
|
8
|
Rivadeneira EO, Allen R, Adam M, Seifert AW. Specific cell states underlie complex tissue regeneration in spiny mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.10.637521. [PMID: 39990382 PMCID: PMC11844359 DOI: 10.1101/2025.02.10.637521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Cell proliferation is an elemental feature of epimorphic regeneration in vertebrate taxa. We previously reported that in contrast to fibrotic repair observed in laboratory mouse ( Mus ) strains, highly regenerative spiny mice ( Acomys spp.) exhibit cell cycle progression and cell proliferation to faithfully replace missing tissue. However, little is known about proliferation dynamics, and specific cell types and states that may contribute to complex tissue regeneration in mammals. Using temporal pulse-chase experiments, we show that stromal cells in Acomys dimidiatus rapidly re-enter the cell cycle in response to injury and maintain tight spatiotemporal control of cell cycle progression to restrict the proliferative population to a distal area relative to the injury. Conversely, Mus stromal cells incorporate thymidine analogs without cell division supporting an S-phase arrest after D10. Deploying immunostaining and scRNA-seq, we identify several key cell types (CRABP1+, αSMA+) differentially associated with regenerating versus scar tissue. Importantly, our single cell data revealed distinct gene expression profiles for cross-species stromal cell types, identifying cell states specific for regenerative or fibrotic healing. While CRABP1+ fibroblasts are enriched in Acomys ears before and after injury, similar fibroblasts enriched in young, postnatal Mus ears remain unable to promote regeneration. Our data underscore the finely regulated dynamics of proliferating cells during regeneration and emphasize that regeneration depends on multiple factors including the presence of specific cell types and the ability of cells to acquire specific states. Key Conclusions Differentiated cells in Acomys , Mus and Danio re-enter the cell cycle in response to injury, while homeostatic cycling cells contribute to blastema formation in Ambystoma Pulse-chase thymidine analog labeling shows tight spatiotemporal control of proliferating stromal cells during regeneration in Acomys . Following injury, CRABP1 and αSMA are expressed in distinct stromal cell populations in Acomys but are co-expressed in Mus stromal cell populations. Species-specific cell states underlie regenerative and fibrotic repair CRABP1+ cells are lost during embryonic development in Mus ear pinna but are retained in Acomys to adulthood. Young neonatal Mus with abundant CRABP1+ cells still fail to execute regenerative healing. SUMMARY STATEMENT Comparing regenerative vs. fibrotic healing, we identify injury-induced cell states associated with persistent cell cycle progression and complex tissue regeneration in mammals.
Collapse
|
9
|
Saad LO, Cooke TF, Atabay KD, Reddien PW, Brown FD. Reduced adult stem cell fate specification led to eye reduction in cave planarians. Nat Commun 2025; 16:304. [PMID: 39746937 PMCID: PMC11696554 DOI: 10.1038/s41467-024-54478-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 11/12/2024] [Indexed: 01/04/2025] Open
Abstract
Eye loss occurs convergently in numerous animal phyla as an adaptation to dark environments. We investigate the cave planarian Girardia multidiverticulata (Gm), a representative species of the Spiralian clade, to study mechanisms of eye loss. We found that Gm, which was previously described as an eyeless species, retains rudimentary and functional eyes. Eyes are maintained in homeostasis and regenerated in adult planarians by stem cells, called neoblasts, through their fate specification to eye progenitors. The reduced number of eye cells in cave planarians is associated with a decreased rate of stem cell fate specification to eye progenitors during homeostasis and regeneration. Conversely, the homeostatic formation of new cells from stem cell-derived progenitors for other tissues, including for neurons, pharynx, and epidermis, is comparable between cave and surface species. These findings reveal a mode of evolutionary trait loss, with change in rate of fate specification in adult stem cells leading to tissue size reduction.
Collapse
Affiliation(s)
- Luiza O Saad
- Departamento de Zoologia, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology, MIT, Cambridge, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Thomas F Cooke
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology, MIT, Cambridge, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Kutay D Atabay
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology, MIT, Cambridge, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Peter W Reddien
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA.
- Department of Biology, MIT, Cambridge, MA, USA.
- Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| | - Federico D Brown
- Departamento de Zoologia, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil.
| |
Collapse
|
10
|
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
|
11
|
Pan X, Zhao Y, Li Y, Chen J, Zhang W, Yang L, Xiong YZ, Ying Y, Xu H, Zhang Y, Gao C, Sun Y, Li N, Chen L, Chen Z, Lei K. Mitochondrial dynamics govern whole-body regeneration through stem cell pluripotency and mitonuclear balance. Nat Commun 2024; 15:10681. [PMID: 39672898 PMCID: PMC11645412 DOI: 10.1038/s41467-024-54720-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 11/19/2024] [Indexed: 12/15/2024] Open
Abstract
Tissue regeneration is a complex process involving large changes in cell proliferation, fate determination, and differentiation. Mitochondrial dynamics and metabolism play a crucial role in development and wound repair, but their function in large-scale regeneration remains poorly understood. Planarians offer an excellent model to investigate this process due to their remarkable regenerative abilities. In this study, we examine mitochondrial dynamics during planarian regeneration. We find that knockdown of the mitochondrial fusion gene, opa1, impairs both tissue regeneration and stem cell pluripotency. Interestingly, the regeneration defects caused by opa1 knockdown are rescued by simultaneous knockdown of the mitochondrial fission gene, drp1, which partially restores mitochondrial dynamics. Furthermore, we discover that Mitolow stem cells exhibit an enrichment of pluripotency due to their fate choices at earlier stages. Transcriptomic analysis reveals the delicate mitonuclear balance in metabolism and mitochondrial proteins in regeneration, controlled by mitochondrial dynamics. These findings highlight the importance of maintaining mitochondrial dynamics in large-scale tissue regeneration and suggest the potential for manipulating these dynamics to enhance stem cell functionality and regenerative processes.
Collapse
Affiliation(s)
- Xue Pan
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Yun Zhao
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
- Fudan University, Shanghai, China
| | - Yucong Li
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
- Fudan University, Shanghai, China
| | - Jiajia Chen
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Wenya Zhang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
- Fudan University, Shanghai, China
| | - Ling Yang
- HPC Center, Westlake University, Hangzhou, Zhejiang, China
| | - Yuanyi Zhou Xiong
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
- Fudan University, Shanghai, China
| | - Yuqing Ying
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Hao Xu
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Yuhong Zhang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Chong Gao
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Yuhan Sun
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Nan Li
- HPC Center, Westlake University, Hangzhou, Zhejiang, China
| | - Liangyi Chen
- College of Future Technology, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, National Biomedical Imaging Center, Peking University, Beijing, China.
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China.
- State Key Laboratory of Membrane Biology, Peking University, Beijing, China.
- PKU-Nanjing Institute of Translational Medicine, Nanjing, China.
| | - Zhixing Chen
- College of Future Technology, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, National Biomedical Imaging Center, Peking University, Beijing, China.
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China.
| | - Kai Lei
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China.
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China.
| |
Collapse
|
12
|
Mansor NI, Balqis TN, Lani MN, Lye KL, Nor Muhammad NA, Ismail WIW, Abidin SZ. Nature's Secret Neuro-Regeneration Pathway in Axolotls, Polychaetes and Planarians for Human Therapeutic Target Pathways. Int J Mol Sci 2024; 25:11904. [PMID: 39595973 PMCID: PMC11593954 DOI: 10.3390/ijms252211904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/03/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
Despite significant improvements in the comprehension of neuro-regeneration, restoring nerve injury in humans continues to pose a substantial therapeutic difficulty. In the peripheral nervous system (PNS), the nerve regeneration process after injury relies on Schwann cells. These cells play a crucial role in regulating and releasing different extracellular matrix proteins, including laminin and fibronectin, which are essential for facilitating nerve regeneration. However, during regeneration, the nerve is required to regenerate for a long distance and, subsequently, loses its capacity to facilitate regeneration during this progression. Meanwhile, it has been noted that nerve regeneration has limited capabilities in the central nervous system (CNS) compared to in the PNS. The CNS contains factors that impede the regeneration of axons following injury to the axons. The presence of glial scar formation results from this unfavourable condition, where glial cells accumulate at the injury site, generating a physical and chemical barrier that hinders the regeneration of neurons. In contrast to humans, several species, such as axolotls, polychaetes, and planarians, possess the ability to regenerate their neural systems following amputation. This ability is based on the vast amount of pluripotent stem cells that have the remarkable capacity to differentiate and develop into any cell within their body. Although humans also possess these cells, their numbers are extremely limited. Examining the molecular pathways exhibited by these organisms has the potential to offer a foundational understanding of the human regeneration process. This review provides a concise overview of the molecular pathways involved in axolotl, polychaete, and planarian neuro-regeneration. It has the potential to offer a new perspective on therapeutic approaches for neuro-regeneration in humans.
Collapse
Affiliation(s)
- Nur Izzati Mansor
- Department of Nursing, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras 56000, Kuala Lumpur, Malaysia;
| | - Tengku Nabilatul Balqis
- Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, Kuala Nerus 21030, Terengganu, Malaysia; (T.N.B.); (W.I.W.I.)
| | - Mohd Nizam Lani
- Faculty of Fisheries and Food Science, Universiti Malaysia Terengganu, Kuala Nerus 21030, Terengganu, Malaysia;
| | - Kwan Liang Lye
- ME Scientifique Sdn Bhd, Taman Universiti Indah, Seri Kembangan 43300, Selangor, Malaysia;
| | - Nor Azlan Nor Muhammad
- Institute of Systems Biology (INBIOSIS), Universiti Kebangsaan Malaysia, Bangi 43600, Selangor, Malaysia;
| | - Wan Iryani Wan Ismail
- Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, Kuala Nerus 21030, Terengganu, Malaysia; (T.N.B.); (W.I.W.I.)
- Research Interest Group Biological Security and Sustainability (BIOSES), Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, Kuala Nerus 21030, Terengganu, Malaysia
| | - Shahidee Zainal Abidin
- Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, Kuala Nerus 21030, Terengganu, Malaysia; (T.N.B.); (W.I.W.I.)
- Research Interest Group Biological Security and Sustainability (BIOSES), Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, Kuala Nerus 21030, Terengganu, Malaysia
| |
Collapse
|
13
|
Lozano-Flores C, Trujillo-Barrientos J, Brito-Domínguez DA, Téllez-Chávez E, Cortés-Encarnación R, Medina-Durazno LD, Cornelio-Martínez S, de León-Cuevas A, Ávalos-Fernández A, Gutiérrez-Sarmiento W, Torres-Barrera A, Soto-Barragán FJ, Herrera-Oropeza G, Martínez-Olvera R, Martínez-Acevedo D, Cruz-Blake LC, Rangel-García V, Martínez-Cabrera G, Larriva-Sahd J, Pimentel-Domínguez R, Ávila R, Varela-Echavarría A. SlugAtlas, a histological and 3D online resource of the land slugs Deroceras laeve and Ambigolimax valentianus. PLoS One 2024; 19:e0312407. [PMID: 39436899 PMCID: PMC11495586 DOI: 10.1371/journal.pone.0312407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 10/04/2024] [Indexed: 10/25/2024] Open
Abstract
Due to their distinctive anatomical characteristics, land slugs are appealing research subjects from a variety of angles, including stem cell biology, regeneration, embryonic development, allometry, and neurophysiology. Here we present SlugAtlas, an anatomical and histological online resource of the land slugs Deroceras laeve and Ambigolimax valentianus. The atlas is composed of series of histological sections on the horizontal, sagittal, and transversal planes for both species with 3D viewing tools of their major organs. The atlas was used in this work for a comparative analysis of the major organs and tissues of these two species. We provide a comprehensive histological description of them and an explanation of novel findings of unique features of their anatomy. For D. laeve, we additionally studied its ability for degrowth and regrowth, a feature that characterizes animals with high regeneration potential and obtained initial results on the study of the regeneration of its tail. SlugAtlas is a resource that is also built to accommodate future growth and, along with the experimental techniques that we have developed, will provide the foundation for research projects in a variety of biological domains.
Collapse
Affiliation(s)
- Carlos Lozano-Flores
- Department of Developmental Neurobiology and Neurophysiology, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, México
| | - Jessica Trujillo-Barrientos
- Department of Developmental Neurobiology and Neurophysiology, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, México
| | - Diego A. Brito-Domínguez
- Department of Developmental Neurobiology and Neurophysiology, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, México
| | - Elisa Téllez-Chávez
- Department of Developmental Neurobiology and Neurophysiology, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, México
| | - Rocío Cortés-Encarnación
- Department of Developmental Neurobiology and Neurophysiology, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, México
| | - Lizbeth D. Medina-Durazno
- Department of Developmental Neurobiology and Neurophysiology, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, México
| | - Sergio Cornelio-Martínez
- Department of Developmental Neurobiology and Neurophysiology, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, México
| | | | | | - Wilbert Gutiérrez-Sarmiento
- Department of Developmental Neurobiology and Neurophysiology, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, México
| | - Aldo Torres-Barrera
- Department of Developmental Neurobiology and Neurophysiology, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, México
| | - Fernando Javier Soto-Barragán
- Department of Developmental Neurobiology and Neurophysiology, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, México
| | - Gabriel Herrera-Oropeza
- Department of Developmental Neurobiology and Neurophysiology, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, México
| | - Ramón Martínez-Olvera
- Department of Developmental Neurobiology and Neurophysiology, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, México
| | - David Martínez-Acevedo
- Department of Developmental Neurobiology and Neurophysiology, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, México
| | - Luis C. Cruz-Blake
- Department of Developmental Neurobiology and Neurophysiology, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, México
| | - Vanessa Rangel-García
- Department of Developmental Neurobiology and Neurophysiology, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, México
| | - Gema Martínez-Cabrera
- Department of Developmental Neurobiology and Neurophysiology, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, México
| | - Jorge Larriva-Sahd
- Department of Developmental Neurobiology and Neurophysiology, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, México
| | | | - Remy Ávila
- Centro de Física Aplicada y Tecnología Avanzada (UNAM), Querétaro, México
| | - Alfredo Varela-Echavarría
- Department of Developmental Neurobiology and Neurophysiology, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, México
- Laboratorio Nacional de Visualización Científica Avanzada (LAVIS), Querétaro, México
| |
Collapse
|
14
|
Bideau L, Velasquillo-Ramirez Z, Baduel L, Basso M, Gilardi-Hebenstreit P, Ribes V, Vervoort M, Gazave E. Variations in cell plasticity and proliferation underlie distinct modes of regeneration along the antero-posterior axis in the annelid Platynereis. Development 2024; 151:dev202452. [PMID: 38950937 DOI: 10.1242/dev.202452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 05/22/2024] [Indexed: 07/03/2024]
Abstract
The capacity to regenerate lost tissues varies significantly among animals. Some phyla, such as the annelids, display substantial regenerating abilities, although little is known about the cellular mechanisms underlying the process. To precisely determine the origin, plasticity and fate of the cells participating in blastema formation and posterior end regeneration after amputation in the annelid Platynereis dumerilii, we developed specific tools to track different cell populations. Using these tools, we find that regeneration is partly promoted by a population of proliferative gut cells whose regenerative potential varies as a function of their position along the antero-posterior axis of the worm. Gut progenitors from anterior differentiated tissues are lineage restricted, whereas gut progenitors from the less differentiated and more proliferative posterior tissues are much more plastic. However, they are unable to regenerate the stem cells responsible for the growth of the worms. Those stem cells are of local origin, deriving from the cells present in the segment abutting the amputation plane, as are most of the blastema cells. Our results favour a hybrid and flexible cellular model for posterior regeneration in Platynereis relying on different degrees of cell plasticity.
Collapse
Affiliation(s)
- Loïc Bideau
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013 Paris, France
| | | | - Loeiza Baduel
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013 Paris, France
| | - Marianne Basso
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013 Paris, France
| | | | - Vanessa Ribes
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013 Paris, France
| | - Michel Vervoort
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013 Paris, France
| | - Eve Gazave
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013 Paris, France
| |
Collapse
|
15
|
Xing N, Gao L, Xie W, Deng H, Yang F, Liu D, Li A, Pang Q. Mining of potentially stem cell-related miRNAs in planarians. Mol Biol Rep 2024; 51:1045. [PMID: 39377855 DOI: 10.1007/s11033-024-09977-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 09/27/2024] [Indexed: 10/09/2024]
Abstract
Stem cells and regenerative medicine have recently become important research topics. However, the complex stem cell regulatory networks involved in various microRNA (miRNA)-mediated mechanisms have not yet been fully elucidated. Planarians are ideal animal models for studying stem cells owing to their rich stem cell populations (neoblasts) and extremely strong regeneration capacity. The roles of planarian miRNAs in stem cells and regeneration have long attracted attention. However, previous studies have generally provided simple datasets lacking integrative analysis. Here, we have summarized the miRNA family reported in planarians and highlighted conservation in both sequence and function. Furthermore, we summarized miRNA data related to planarian stem cells and regeneration and screened potential involved candidates. Nevertheless, the roles of these miRNAs in planarian regeneration and stem cells remain unclear. The identification of potential stem cell-related miRNAs offers more precise suggestions and references for future investigations of miRNAs in planarians. Furthermore, it provides potential research avenues for understanding the mechanisms of stem cell regulatory networks. Finally, we compiled a summary of the experimental methods employed for studying planarian miRNAs, with the aim of highlighting special considerations in certain procedures and providing more convenient technical support for future research endeavors.
Collapse
Affiliation(s)
- Nianhong Xing
- Anti-aging & Regenerative Medicine Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo, 255049, China
| | - Lili Gao
- Anti-aging & Regenerative Medicine Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo, 255049, China.
| | - Wenshuo Xie
- Anti-aging & Regenerative Medicine Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo, 255049, China
| | - Hongkuan Deng
- Anti-aging & Regenerative Medicine Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo, 255049, China
| | - Fengtang Yang
- Anti-aging & Regenerative Medicine Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo, 255049, China
| | - Dongwu Liu
- Anti-aging & Regenerative Medicine Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo, 255049, China
| | - Ao Li
- Anti-aging & Regenerative Medicine Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo, 255049, China
| | - Qiuxiang Pang
- Anti-aging & Regenerative Medicine Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo, 255049, China.
| |
Collapse
|
16
|
Kim RT, Whited JL. Putative epithelial-mesenchymal transitions during salamander limb regeneration: Current perspectives and future investigations. Ann N Y Acad Sci 2024; 1540:89-103. [PMID: 39269330 PMCID: PMC11471381 DOI: 10.1111/nyas.15210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
Previous studies have implicated epithelial-mesenchymal transition (EMT) in salamander limb regeneration. In this review, we describe putative roles for EMT during each stage of limb regeneration in axolotls and other salamanders. We hypothesize that EMT and EMT-like gene expression programs may regulate three main cellular processes during limb regeneration: (1) keratinocyte migration during wound closure; (2) transient invasion of the stump by epithelial cells undergoing EMT; and (3) use of EMT-like programs by non-epithelial blastemal progenitor cells to escape the confines of their niches. Finally, we propose nontraditional roles for EMT during limb regeneration that warrant further investigation, including alternative EMT regulators, stem cell activation, and fibrosis induced by aberrant EMT.
Collapse
Affiliation(s)
- Ryan T Kim
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA
| | - Jessica L Whited
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| |
Collapse
|
17
|
Mukundan N, Hariharan N, Sasidharan V, Lakshmanan V, Palakodeti D, Jamora C. Poly (A) binding protein 2 is critical for stem cell differentiation during regeneration in the planarian Schmidtea mediterranea. Front Cell Dev Biol 2024; 12:1433142. [PMID: 39376632 PMCID: PMC11456742 DOI: 10.3389/fcell.2024.1433142] [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: 05/15/2024] [Accepted: 08/21/2024] [Indexed: 10/09/2024] Open
Abstract
Post-transcriptional regulation has emerged as a key mechanism for regulating stem cell renewal and differentiation, which is essential for understanding tissue regeneration and homeostasis. Poly(A)-binding proteins are a family of RNA-binding proteins that play a vital role in post-transcriptional regulation by controlling mRNA stability and protein synthesis. The involvement of poly(A) binding proteins in a wide range of cellular functions is increasingly being investigated. In this study, we used the regenerative model planarian organism Schmidtea mediterranea to demonstrate the critical role of poly(A)-binding protein 2 (PABP2) in regulating neoblast maintenance and differentiation. A deficit in PABP2 blocks the transition of neoblasts toward immediate early progenitors, leading to an enhanced pool of non-committed neoblasts and a decreased progenitor population. This is reflected in variations in the transcriptome profile, providing evidence of downregulation in multiple lineages. Thus, an insufficiency of PABP2 resulted in defective formation and organization of tissue, leading to abnormal regeneration. Our study reveals the essential role of PABP2 in regulating genes that mediate stem cell commitment to early progenitors during tissue regeneration.
Collapse
Affiliation(s)
- Namita Mukundan
- Integrative Chemical Biology (ICB), Institute for Stem Cell Science and Regenerative Medicine, Bangalore, India
- Manipal Academy of Higher Education, Manipal, India
| | - Nivedita Hariharan
- Integrative Chemical Biology (ICB), Institute for Stem Cell Science and Regenerative Medicine, Bangalore, India
| | | | - Vairavan Lakshmanan
- Integrative Chemical Biology (ICB), Institute for Stem Cell Science and Regenerative Medicine, Bangalore, India
| | - Dasaradhi Palakodeti
- Integrative Chemical Biology (ICB), Institute for Stem Cell Science and Regenerative Medicine, Bangalore, India
- Manipal Academy of Higher Education, Manipal, India
| | - Colin Jamora
- Integrative Chemical Biology (ICB), Institute for Stem Cell Science and Regenerative Medicine, Bangalore, India
- Department of Life Science, Shiv Nadar Institution of Eminence, Greater Noida, Uttar Pradesh, India
| |
Collapse
|
18
|
Poss KD, Tanaka EM. Hallmarks of regeneration. Cell Stem Cell 2024; 31:1244-1261. [PMID: 39163854 PMCID: PMC11410156 DOI: 10.1016/j.stem.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/12/2024] [Accepted: 07/24/2024] [Indexed: 08/22/2024]
Abstract
Regeneration is a heroic biological process that restores tissue architecture and function in the face of day-to-day cell loss or the aftershock of injury. Capacities and mechanisms for regeneration can vary widely among species, organs, and injury contexts. Here, we describe "hallmarks" of regeneration found in diverse settings of the animal kingdom, including activation of a cell source, initiation of regenerative programs in the source, interplay with supporting cell types, and control of tissue size and function. We discuss these hallmarks with an eye toward major challenges and applications of regenerative biology.
Collapse
Affiliation(s)
- Kenneth D Poss
- Duke Regeneration Center and Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA.
| | - Elly M Tanaka
- Institute of Molecular Biotechnology (IMBA), Austrian Academy of Sciences, Vienna Biocenter (VBC), 1030 Vienna, Austria.
| |
Collapse
|
19
|
Dooling KE, Kim RT, Kim EM, Chen E, Abouelela A, Tajer BJ, Lopez NJ, Paoli JC, Powell CJ, Luong AG, Wu SC, Thornton KN, Singer HD, Savage AM, Bateman J, DiTommaso T, Payzin-Dogru D, Whited JL. Amputation Triggers Long-Range Epidermal Permeability Changes in Evolutionarily Distant Regenerative Organisms. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.29.610385. [PMID: 39257748 PMCID: PMC11383696 DOI: 10.1101/2024.08.29.610385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Previous studies have reported that amputation invokes body-wide responses in regenerative organisms, but most have not examined the implications of these changes beyond the region of tissue regrowth. Specifically, long-range epidermal responses to amputation are largely uncharacterized, with research on amputation-induced epidermal responses in regenerative organisms traditionally being restricted to the wound site. Here, we investigate the effect of amputation on long-range epidermal permeability in two evolutionarily distant, regenerative organisms: axolotls and planarians. We find that amputation triggers a long-range increase in epidermal permeability in axolotls, accompanied by a long-range epidermal downregulation in MAPK signaling. Additionally, we provide functional evidence that pharmacologically inhibiting MAPK signaling in regenerating planarians increases long-range epidermal permeability. These findings advance our knowledge of body-wide changes due to amputation in regenerative organisms and warrant further study on whether epidermal permeability dysregulation in the context of amputation may lead to pathology in both regenerative and non-regenerative organisms.
Collapse
Affiliation(s)
- Kelly E. Dooling
- Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave., Cambridge, MA, USA 02138
| | - Ryan T. Kim
- Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave., Cambridge, MA, USA 02138
| | - Elane M. Kim
- Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave., Cambridge, MA, USA 02138
| | - Erica Chen
- Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave., Cambridge, MA, USA 02138
| | - Adnan Abouelela
- Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave., Cambridge, MA, USA 02138
| | - Benjamin J. Tajer
- Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave., Cambridge, MA, USA 02138
| | - Noah J. Lopez
- Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave., Cambridge, MA, USA 02138
| | - Julia C. Paoli
- Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave., Cambridge, MA, USA 02138
| | - Connor J. Powell
- Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave., Cambridge, MA, USA 02138
| | - Anna G. Luong
- Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave., Cambridge, MA, USA 02138
| | - S.Y. Celeste Wu
- Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave., Cambridge, MA, USA 02138
| | - Kara N. Thornton
- Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave., Cambridge, MA, USA 02138
| | - Hani D. Singer
- Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave., Cambridge, MA, USA 02138
| | - Aaron M. Savage
- Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave., Cambridge, MA, USA 02138
| | - Joel Bateman
- Brigham Regenerative Medicine Center and Department of Orthopedic Surgery, Brigham & Women’s Hospital, Cambridge, MA, USA 02138
| | - Tia DiTommaso
- Brigham Regenerative Medicine Center and Department of Orthopedic Surgery, Brigham & Women’s Hospital, Cambridge, MA, USA 02138
| | - Duygu Payzin-Dogru
- Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave., Cambridge, MA, USA 02138
| | - Jessica L. Whited
- Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave., Cambridge, MA, USA 02138
- Brigham Regenerative Medicine Center and Department of Orthopedic Surgery, Brigham & Women’s Hospital, Cambridge, MA, USA 02138
- The Broad Institute of Harvard and MIT, Cambridge, MA, USA 02138
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA 02138
| |
Collapse
|
20
|
Hulett RE, Rivera-López C, Gehrke AR, Gompers A, Srivastava M. A wound-induced differentiation trajectory for neurons. Proc Natl Acad Sci U S A 2024; 121:e2322864121. [PMID: 38976727 PMCID: PMC11260127 DOI: 10.1073/pnas.2322864121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 05/03/2024] [Indexed: 07/10/2024] Open
Abstract
Animals capable of whole-body regeneration can replace any missing cell type and regenerate fully functional new organs, including new brains, de novo. The regeneration of a new brain requires the formation of diverse neural cell types and their assembly into an organized structure with correctly wired circuits. Recent work in various regenerative animals has revealed transcriptional programs required for the differentiation of distinct neural subpopulations, however, how these transcriptional programs are initiated in response to injury remains unknown. Here, we focused on the highly regenerative acoel worm, Hofstenia miamia, to study wound-induced transcriptional regulatory events that lead to the production of neurons and subsequently a functional brain. Footprinting analysis using chromatin accessibility data on a chromosome-scale genome assembly revealed that binding sites for the Nuclear Factor Y (NFY) transcription factor complex were significantly bound during regeneration, showing a dynamic increase in binding within one hour upon amputation specifically in tail fragments, which will regenerate a new brain. Strikingly, NFY targets were highly enriched for genes with neuronal function. Single-cell transcriptome analysis combined with functional studies identified soxC+ stem cells as a putative progenitor population for multiple neural subtypes. Further, we found that wound-induced soxC expression is likely under direct transcriptional control by NFY, uncovering a mechanism for the initiation of a neural differentiation pathway by early wound-induced binding of a transcriptional regulator.
Collapse
Affiliation(s)
- Ryan E. Hulett
- Department of Organismic and Evolutionary Biology, Museum of Comparative Zoology, Harvard University, Cambridge, MA02138
| | - Carlos Rivera-López
- Department of Organismic and Evolutionary Biology, Museum of Comparative Zoology, Harvard University, Cambridge, MA02138
- Department of Molecular and Cell Biology, Harvard University, Cambridge, MA02138
| | - Andrew R. Gehrke
- Department of Organismic and Evolutionary Biology, Museum of Comparative Zoology, Harvard University, Cambridge, MA02138
| | - Annika Gompers
- Department of Organismic and Evolutionary Biology, Museum of Comparative Zoology, Harvard University, Cambridge, MA02138
| | - Mansi Srivastava
- Department of Organismic and Evolutionary Biology, Museum of Comparative Zoology, Harvard University, Cambridge, MA02138
| |
Collapse
|
21
|
Avalos PN, Wong LL, Forsthoefel DJ. Extracellular vesicles promote proliferation in an animal model of regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.22.586206. [PMID: 38712279 PMCID: PMC11071309 DOI: 10.1101/2024.03.22.586206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Extracellular vesicles (EVs) are secreted nanoparticles composed of a lipid bilayer that carry lipid, protein, and nucleic acid cargo between cells as a mode of intercellular communication. Although EVs can promote tissue repair in mammals, their roles in animals with greater regenerative capacity are not well understood. Planarian flatworms are capable of whole body regeneration due to pluripotent somatic stem cells called neoblasts that proliferate in response to injury. Here, using transmission electron microscopy, nanoparticle tracking analysis, and protein content examination, we showed that EVs enriched from the tissues of the planarian Schmidtea mediterranea had similar morphology and size as other eukaryotic EVs, and that these EVs carried orthologs of the conserved EV biogenesis regulators ALIX and TSG101. PKH67-labeled EVs were taken up more quickly by S/G2 neoblasts than G1 neoblasts/early progeny and differentiated cells. When injected into living planarians, EVs from regenerating tissue fragments enhanced upregulation of neoblast-associated transcripts. In addition, EV injection increased the number of F-ara-EdU-labelled cells by 49% as compared to buffer injection only. Our findings demonstrate that regenerating planarians produce EVs that promote stem cell proliferation, and suggest the planarian as an amenable in vivo model for the study of EV function during regeneration.
Collapse
Affiliation(s)
- Priscilla N. Avalos
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | - Lily L. Wong
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | - David J. Forsthoefel
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| |
Collapse
|
22
|
King HO, Owusu-Boaitey KE, Fincher CT, Reddien PW. A transcription factor atlas of stem cell fate in planarians. Cell Rep 2024; 43:113843. [PMID: 38401119 PMCID: PMC11232438 DOI: 10.1016/j.celrep.2024.113843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 12/05/2023] [Accepted: 02/06/2024] [Indexed: 02/26/2024] Open
Abstract
Whole-body regeneration requires the ability to produce the full repertoire of adult cell types. The planarian Schmidtea mediterranea contains over 125 cell types, which can be regenerated from a stem cell population called neoblasts. Neoblast fate choice can be regulated by the expression of fate-specific transcription factors (FSTFs). How fate choices are made and distributed across neoblasts versus their post-mitotic progeny remains unclear. We used single-cell RNA sequencing to systematically map fate choices made in S/G2/M neoblasts and, separately, in their post-mitotic progeny that serve as progenitors for all adult cell types. We defined transcription factor expression signatures associated with all detected fates, identifying numerous new progenitor classes and FSTFs that regulate them. Our work generates an atlas of stem cell fates with associated transcription factor signatures for most cell types in a complete adult organism.
Collapse
Affiliation(s)
- Hunter O King
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kwadwo E Owusu-Boaitey
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA; Harvard/MIT MD-PhD Program, Harvard Medical School, Boston, MA, USA
| | - Christopher T Fincher
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Peter W Reddien
- Howard Hughes Medical Institute, Chevy Chase, MD, USA; Whitehead Institute for Biomedical Research, Cambridge, MA, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
23
|
Póti Á, Szüts D, Vermezovic J. Mutational profile of the regenerative process and de novo genome assembly of the planarian Schmidtea polychroa. Nucleic Acids Res 2024; 52:1779-1792. [PMID: 38180823 PMCID: PMC10899757 DOI: 10.1093/nar/gkad1250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 12/13/2023] [Accepted: 01/03/2024] [Indexed: 01/07/2024] Open
Abstract
Planarians are organisms with a unique capacity to regenerate any part of their body. New tissues are generated in a process that requires many swift cell divisions. How costly is this process to an animal in terms of mutational load remains unknown. Using whole genome sequencing, we defined the mutational profile of the process of regeneration in the planarian species Schmidtea polychroa. We assembled de novo the genome of S. polychroa and analyzed mutations in animals that have undergone regeneration. We observed a threefold increase in the number of mutations and an altered mutational spectrum. High allele frequencies of subclonal mutations in regenerated animals suggested that most of the cells in the regenerated animal were descendants of a small number of stem cells with high expansion potential. We provide, for the first time, the draft genome assembly of S. polychroa, an estimation of the germline mutation rate for a planarian species and the mutational spectrum of the regeneration process of a living organism.
Collapse
Affiliation(s)
- Ádám Póti
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, H-1117, Hungary
| | - Dávid Szüts
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, H-1117, Hungary
| | - Jelena Vermezovic
- IFOM ETS - The AIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| |
Collapse
|
24
|
Sun Y, Huang Y, Hao Z, Zhang S, Tian Q. MRLC controls apoptotic cell death and functions to regulate epidermal development during planarian regeneration and homeostasis. Cell Prolif 2024; 57:e13524. [PMID: 37357415 PMCID: PMC10771114 DOI: 10.1111/cpr.13524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 06/02/2023] [Accepted: 06/13/2023] [Indexed: 06/27/2023] Open
Abstract
Adult stem cells (ASCs) are pluripotent cells with the capacity to self-renew and constantly replace lost cells due to physiological turnover or injury. Understanding the molecular mechanisms of the precise coordination of stem cell proliferation and proper cell fate decision is important to regeneration and organismal homeostasis. The planarian epidermis provides a highly tractable model to study ASC complex dynamic due to the distinct spatiotemporal differentiation stages during lineage development. Here, we identified the myosin regulatory light chain (MRLC) homologue in the Dugesia japonica transcriptome. We found high expression levels of MRLC in wound region during regeneration and also expressed in late epidermal progenitors as an essential regulator of the lineage from neoblasts to mature epidermal cells. We investigated the function of MRLC using in situ hybridization, real-time polymerase chain reaction and double fluorescent and uncovered the potential mechanism. Knockdown of MRLC leads to a remarkable increase in cell death, causes severe abnormalities during regeneration and homeostasis and eventually leads to animal death. The global decrease in epidermal cell in MRLC RNAi animals induces accelerated epidermal proliferation and differentiation. Additionally, we find that MRLC is co-expressed with cdc42 and acts cooperatively to control the epidermal lineage development by affecting cell death. Our results uncover an important role of MRLC, as an inhibitor of apoptosis, involves in epidermal development.
Collapse
Affiliation(s)
- Yujia Sun
- School of Life SciencesZhengzhou UniversityZhengzhouHenanChina
| | - Yongding Huang
- School of Life SciencesZhengzhou UniversityZhengzhouHenanChina
| | - Zhitai Hao
- Department of Biochemistry and Molecular PharmacologyNew York University, School of MedicineNew YorkUSA
| | - Shoutao Zhang
- School of Life SciencesZhengzhou UniversityZhengzhouHenanChina
- Longhu Laboratory of Advanced ImmunologyZhengzhouHenanChina
| | - Qingnan Tian
- School of Life SciencesZhengzhou UniversityZhengzhouHenanChina
| |
Collapse
|
25
|
Molina MD, Abduljabbar D, Guixeras A, Fraguas S, Cebrià F. LIM-HD transcription factors control axial patterning and specify distinct neuronal and intestinal cell identities in planarians. Open Biol 2023; 13:230327. [PMID: 38086422 PMCID: PMC10715919 DOI: 10.1098/rsob.230327] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 11/01/2023] [Indexed: 12/18/2023] Open
Abstract
Adult planarians can regenerate the gut, eyes and even a functional brain. Proper identity and patterning of the newly formed structures require signals that guide and commit their adult stem cells. During embryogenesis, LIM-homeodomain (LIM-HD) transcription factors act in a combinatorial 'LIM code' to control cell fate determination and differentiation. However, our understanding about the role these genes play during regeneration and homeostasis is limited. Here, we report the full repertoire of LIM-HD genes in Schmidtea mediterranea. We found that lim homeobox (lhx) genes appear expressed in complementary patterns along the cephalic ganglia and digestive system of the planarian, with some of them being co-expressed in the same cell types. We have identified that Smed-islet1, -lhx1/5-1, -lhx2/9-3, -lhx6/8, -lmx1a/b-2 and -lmx1a/b-3 are essential to pattern and size the planarian brain as well as for correct regeneration of specific subpopulations of dopaminergic, serotonergic, GABAergic and cholinergic neurons, while Smed-lhx1/5.2 and -lhx2/9.2 are required for the proper expression of intestinal cell type markers, specifically the goblet subtype. LIM-HD are also involved in controlling axonal pathfinding (lhx6/8), axial patterning (islet1, lhx1/5-1, lmx1a/b-3), head/body proportions (islet2) and stem cell proliferation (lhx3/4, lhx2/9-3, lmx1a/b-2, lmx1a/b-3). Altogether, our results suggest that planarians might present a combinatorial LIM code that controls axial patterning and axonal growing and specifies distinct neuronal and intestinal cell identities.
Collapse
Affiliation(s)
- M. Dolores Molina
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain
| | - Dema Abduljabbar
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Anna Guixeras
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain
| | - Susanna Fraguas
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain
| | - Francesc Cebrià
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain
| |
Collapse
|
26
|
Abstract
Tissue regeneration is not simply a local repair event occurring in isolation from the distant, uninjured parts of the body. Rather, evidence indicates that regeneration is a whole-animal process involving coordinated interactions between different organ systems. Here, we review recent studies that reveal how remote uninjured tissues and organ systems respond to and engage in regeneration. We also discuss the need for toolkits and technological advancements to uncover and dissect organ communication during regeneration.
Collapse
Affiliation(s)
- Fei Sun
- Duke Regeneration Center, Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Kenneth D. Poss
- Duke Regeneration Center, Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
27
|
Park C, Owusu-Boaitey KE, Valdes GM, Reddien PW. Fate specification is spatially intermingled across planarian stem cells. Nat Commun 2023; 14:7422. [PMID: 37973979 PMCID: PMC10654723 DOI: 10.1038/s41467-023-43267-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 11/06/2023] [Indexed: 11/19/2023] Open
Abstract
Regeneration requires mechanisms for producing a wide array of cell types. Neoblasts are stem cells in the planarian Schmidtea mediterranea that undergo fate specification to produce over 125 adult cell types. Fate specification in neoblasts can be regulated through expression of fate-specific transcription factors. We utilize multiplexed error-robust fluorescence in situ hybridization (MERFISH) and whole-mount FISH to characterize fate choice distribution of stem cells within planarians. Fate choices are often made distant from target tissues and in a highly intermingled manner, with neighboring neoblasts frequently making divergent fate choices for tissues of different location and function. We propose that pattern formation is driven primarily by the migratory assortment of progenitors from mixed and spatially distributed fate-specified stem cells and that fate choice involves stem-cell intrinsic processes.
Collapse
Affiliation(s)
- Chanyoung Park
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kwadwo E Owusu-Boaitey
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard/MIT MD-PhD Program, Harvard Medical School, Boston, MA, USA
| | - Giselle M Valdes
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Peter W Reddien
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA.
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| |
Collapse
|
28
|
Song Q, Geng H, Zhen H, Liu H, Deng H, Yuan Z, Zhang J, Cao Z, Pang Q, Zhao B. DjFARP Contributes to the Regeneration and Maintenance of the Brain through Activation of DjRac1 in Dugesia japonica. Mol Neurobiol 2023; 60:6294-6306. [PMID: 37442859 DOI: 10.1007/s12035-023-03478-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 07/02/2023] [Indexed: 07/15/2023]
Abstract
FERM, RhoGEF, and Pleckstrin domain protein (FARP) mediated RhoGTPase pathways are involved in diverse biological processes, such as neuronal development and tumorigenesis. However, little is known about their role in neural regeneration. We uncovered for the first time that FARP-Rac1 signaling plays an important role in neural regeneration in Dugesia japonica, a planarian that possesses unparalleled regenerative capacities. The planarian FARP homolog DjFARP was primarily expressed in both intact and regenerating brain and pharynx tissue. Functional studies suggested that downregulation of DjFARP with dsRNA in Dugesia japonica led to smaller brain sizes, defects in brain lateral branches, and loss of cholinergic, GABAergic, and dopaminergic neurons in both intact and regenerating animals. Moreover, the Rho GTPase DjRac1 was shown to play a similar role in neural regeneration and maintenance. Rac1 activation assay showed that DjFARP acts as a guanine nucleotide exchange factor (GEF) for DjRac1. Together, these findings indicate that the brain defects seen in DjFARP knockdown animals may be attributable to DjRac1 inactivation. In conclusion, our study demonstrated that DjFARP-DjRac1 signaling was required for the maintenance and proper regeneration of the brain in Dugesia japonica.
Collapse
Affiliation(s)
- Qian Song
- Laboratory of Developmental and Evolutionary Biology, Shandong University of Technology, 266 Xincun West Road, Zibo, 255049, People's Republic of China
| | - Huazhi Geng
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, 255049, China
| | - Hui Zhen
- Laboratory of Developmental and Evolutionary Biology, Shandong University of Technology, 266 Xincun West Road, Zibo, 255049, People's Republic of China
| | - Hongjin Liu
- Zibo Maternal and Child Health Hospital, Zibo, 255000, China
| | - Hongkuan Deng
- Zibo Maternal and Child Health Hospital, Zibo, 255000, China
| | - Zuoqing Yuan
- Zibo Maternal and Child Health Hospital, Zibo, 255000, China
| | - Jianyong Zhang
- Zibo Maternal and Child Health Hospital, Zibo, 255000, China
| | - Zhonghong Cao
- Zibo Maternal and Child Health Hospital, Zibo, 255000, China
| | - Qiuxiang Pang
- Zibo Maternal and Child Health Hospital, Zibo, 255000, China
| | - Bosheng Zhao
- Laboratory of Developmental and Evolutionary Biology, Shandong University of Technology, 266 Xincun West Road, Zibo, 255049, People's Republic of China.
- Zibo Maternal and Child Health Hospital, Zibo, 255000, China.
| |
Collapse
|
29
|
Paré L, Bideau L, Baduel L, Dalle C, Benchouaia M, Schneider SQ, Laplane L, Clément Y, Vervoort M, Gazave E. Transcriptomic landscape of posterior regeneration in the annelid Platynereis dumerilii. BMC Genomics 2023; 24:583. [PMID: 37784028 PMCID: PMC10546743 DOI: 10.1186/s12864-023-09602-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 08/18/2023] [Indexed: 10/04/2023] Open
Abstract
BACKGROUND Restorative regeneration, the capacity to reform a lost body part following amputation or injury, is an important and still poorly understood process in animals. Annelids, or segmented worms, show amazing regenerative capabilities, and as such are a crucial group to investigate. Elucidating the molecular mechanisms that underpin regeneration in this major group remains a key goal. Among annelids, the nereididae Platynereis dumerilii (re)emerged recently as a front-line regeneration model. Following amputation of its posterior part, Platynereis worms can regenerate both differentiated tissues of their terminal part as well as a growth zone that contains putative stem cells. While this regeneration process follows specific and reproducible stages that have been well characterized, the transcriptomic landscape of these stages remains to be uncovered. RESULTS We generated a high-quality de novo Reference transcriptome for the annelid Platynereis dumerilii. We produced and analyzed three RNA-sequencing datasets, encompassing five stages of posterior regeneration, along with blastema stages and non-amputated tissues as controls. We included two of these regeneration RNA-seq datasets, as well as embryonic and tissue-specific datasets from the literature to produce a Reference transcriptome. We used this Reference transcriptome to perform in depth analyzes of RNA-seq data during the course of regeneration to reveal the important dynamics of the gene expression, process with thousands of genes differentially expressed between stages, as well as unique and specific gene expression at each regeneration stage. The study of these genes highlighted the importance of the nervous system at both early and late stages of regeneration, as well as the enrichment of RNA-binding proteins (RBPs) during almost the entire regeneration process. CONCLUSIONS In this study, we provided a high-quality de novo Reference transcriptome for the annelid Platynereis that is useful for investigating various developmental processes, including regeneration. Our extensive stage-specific transcriptional analysis during the course of posterior regeneration sheds light upon major molecular mechanisms and pathways, and will foster many specific studies in the future.
Collapse
Affiliation(s)
- Louis Paré
- Université Paris Cité, CNRS, Institut Jacques Monod, Paris, F-75013, France
| | - Loïc Bideau
- Université Paris Cité, CNRS, Institut Jacques Monod, Paris, F-75013, France
| | - Loeiza Baduel
- Université Paris Cité, CNRS, Institut Jacques Monod, Paris, F-75013, France
| | - Caroline Dalle
- Université Paris Cité, CNRS, Institut Jacques Monod, Paris, F-75013, France
| | - Médine Benchouaia
- Département de biologie, GenomiqueENS, Institut de Biologie de l'ENS (IBENS), École normale supérieure, CNRS, INSERM, Université PSL, Paris, 75005, France
| | - Stephan Q Schneider
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 11529, Taiwan
| | - Lucie Laplane
- Université Paris I Panthéon-Sorbonne, CNRS UMR 8590 Institut d'Histoire et de Philosophie des Sciences et des Techniques (IHPST), Paris, France
- Gustave Roussy, UMR 1287, Villejuif, France
| | - Yves Clément
- Université Paris Cité, CNRS, Institut Jacques Monod, Paris, F-75013, France
| | - Michel Vervoort
- Université Paris Cité, CNRS, Institut Jacques Monod, Paris, F-75013, France
| | - Eve Gazave
- Université Paris Cité, CNRS, Institut Jacques Monod, Paris, F-75013, France.
| |
Collapse
|
30
|
Abstract
Regeneration requires the collective effort of multiple organ systems. A recent study of planarian whole-body regeneration finds that Erk kinase activity propagates rapidly across the entire animal through longitudinal muscle cells to coordinate animal-wide wound responses and that this signal propagation is required for regeneration.
Collapse
Affiliation(s)
- Fei Sun
- Duke Regeneration Center and Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
| | - Alessandro De Simone
- Department of Genetics and Evolution, Faculty of Sciences, University of Geneva, Geneva, Switzerland
| | - Kenneth D Poss
- Duke Regeneration Center and Department of Cell Biology, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
31
|
Chen JJ, Lei K. The known, unknown, and unknown unknowns of cell-cell communication in planarian regeneration. Zool Res 2023; 44:981-992. [PMID: 37721107 PMCID: PMC10559094 DOI: 10.24272/j.issn.2095-8137.2023.044] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 09/11/2023] [Indexed: 09/19/2023] Open
Abstract
Planarians represent the most primitive bilateral triploblastic animals. Most planarian species exhibit mechanisms for whole-body regeneration, exemplified by the regeneration of their cephalic ganglion after complete excision. Given their robust whole-body regeneration capacity, planarians have been model organisms in regenerative research for more than 240 years. Advancements in research tools and techniques have progressively elucidated the mechanisms underlying planarian regeneration. Accurate cell-cell communication is recognized as a fundamental requirement for regeneration. In recent decades, mechanisms associated with such communication have been revealed at the cellular level. Notably, stem cells (neoblasts) have been identified as the source of all new cells during planarian homeostasis and regeneration. The interplay between neoblasts and somatic cells affects the identities and proportions of various tissues during homeostasis and regeneration. Here, this review outlines key discoveries regarding communication between stem cell compartments and other cell types in planarians, as well as the impact of communication on planarian regeneration. Additionally, this review discusses the challenges and potential directions of future planarian research, emphasizing the sustained impact of this field on our understanding of animal regeneration.
Collapse
Affiliation(s)
- Jia-Jia Chen
- School of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310030, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang 310024, China
| | - Kai Lei
- Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310030, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang 310024, China. E-mail:
| |
Collapse
|
32
|
Akheralie Z, Scidmore TJ, Pearson BJ. aristaless-like homeobox-3 is wound induced and promotes a low-Wnt environment required for planarian head regeneration. Development 2023; 150:dev201777. [PMID: 37681295 PMCID: PMC10560571 DOI: 10.1242/dev.201777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 08/30/2023] [Indexed: 09/09/2023]
Abstract
The planarian Schmidtea mediterranea is a well-established model of adult regeneration, which is dependent on a large population of adult stem cells called neoblasts. Upon amputation, planarians undergo transcriptional wounding programs and coordinated stem cell proliferation to give rise to missing tissues. Interestingly, the Wnt signaling pathway is key to guiding what tissues are regenerated, yet less known are the transcriptional regulators that ensure proper activation and timing of signaling pathway components. Here, we have identified an aristaless-like homeobox transcription factor, alx-3, that is enriched in a population of putative neural-fated progenitor cells at homeostasis, and is also upregulated in stem cells and muscle cells at anterior-facing wounds upon amputation. Knockdown of alx-3 results in failure of head regeneration and patterning defects in amputated tail fragments. alx-3 is required for the expression of several early wound-induced genes, including the Wnt inhibitor notum, which is required to establish anterior polarity during regeneration. Together, these findings reveal a role for alx-3 as an early wound-response transcriptional regulator in both muscle cells and stem cells that is required for anterior regeneration by promoting a low-Wnt environment.
Collapse
Affiliation(s)
- Zaleena Akheralie
- The Hospital for Sick Children, Program in Developmental and Stem Cell Biology, Toronto, ON M5G0A4, Canada
- University of Toronto, Department of Molecular Genetics, Toronto, ON M5S1A8, Canada
| | - Tanner J. Scidmore
- The Hospital for Sick Children, Program in Developmental and Stem Cell Biology, Toronto, ON M5G0A4, Canada
- University of Toronto, Department of Molecular Genetics, Toronto, ON M5S1A8, Canada
| | - Bret J. Pearson
- The Hospital for Sick Children, Program in Developmental and Stem Cell Biology, Toronto, ON M5G0A4, Canada
- University of Toronto, Department of Molecular Genetics, Toronto, ON M5S1A8, Canada
| |
Collapse
|
33
|
Fan Y, Chai C, Li P, Zou X, Ferrell JE, Wang B. Ultrafast distant wound response is essential for whole-body regeneration. Cell 2023; 186:3606-3618.e16. [PMID: 37480850 PMCID: PMC10957142 DOI: 10.1016/j.cell.2023.06.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 01/11/2023] [Accepted: 06/23/2023] [Indexed: 07/24/2023]
Abstract
Injury induces systemic responses, but their functions remain elusive. Mechanisms that can rapidly synchronize wound responses through long distances are also mostly unknown. Using planarian flatworms capable of whole-body regeneration, we report that injury induces extracellular signal-regulated kinase (Erk) activity waves to travel at a speed 10-100 times faster than those in other multicellular tissues. This ultrafast propagation requires longitudinal body-wall muscles, elongated cells forming dense parallel tracks running the length of the organism. The morphological properties of muscles allow them to act as superhighways for propagating and disseminating wound signals. Inhibiting Erk propagation prevents tissues distant to the wound from responding and blocks regeneration, which can be rescued by a second injury to distal tissues shortly after the first injury. Our findings provide a mechanism for long-range signal propagation in large, complex tissues to coordinate responses across cell types and highlight the function of feedback between spatially separated tissues during whole-body regeneration.
Collapse
Affiliation(s)
- Yuhang Fan
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Chew Chai
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Pengyang Li
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Xinzhi Zou
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - James E Ferrell
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA; Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - Bo Wang
- Department of Bioengineering, Stanford University, Stanford, CA, USA.
| |
Collapse
|
34
|
Tajer B, Savage AM, Whited JL. The salamander blastema within the broader context of metazoan regeneration. Front Cell Dev Biol 2023; 11:1206157. [PMID: 37635872 PMCID: PMC10450636 DOI: 10.3389/fcell.2023.1206157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 07/26/2023] [Indexed: 08/29/2023] Open
Abstract
Throughout the animal kingdom regenerative ability varies greatly from species to species, and even tissue to tissue within the same organism. The sheer diversity of structures and mechanisms renders a thorough comparison of molecular processes truly daunting. Are "blastemas" found in organisms as distantly related as planarians and axolotls derived from the same ancestral process, or did they arise convergently and independently? Is a mouse digit tip blastema orthologous to a salamander limb blastema? In other fields, the thorough characterization of a reference model has greatly facilitated these comparisons. For example, the amphibian Spemann-Mangold organizer has served as an amazingly useful comparative template within the field of developmental biology, allowing researchers to draw analogies between distantly related species, and developmental processes which are superficially quite different. The salamander limb blastema may serve as the best starting point for a comparative analysis of regeneration, as it has been characterized by over 200 years of research and is supported by a growing arsenal of molecular tools. The anatomical and evolutionary closeness of the salamander and human limb also add value from a translational and therapeutic standpoint. Tracing the evolutionary origins of the salamander blastema, and its relatedness to other regenerative processes throughout the animal kingdom, will both enhance our basic biological understanding of regeneration and inform our selection of regenerative model systems.
Collapse
Affiliation(s)
| | | | - Jessica L. Whited
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, United States
| |
Collapse
|
35
|
Zhen H, Huang M, Zheng M, Gao L, Guo Z, Pang Q, Jin G, Zhou Z. WTAP regulates stem cells via TRAF6 to maintain planarian homeostasis and regeneration. Int J Biol Macromol 2023:124932. [PMID: 37268082 DOI: 10.1016/j.ijbiomac.2023.124932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 04/23/2023] [Accepted: 05/06/2023] [Indexed: 06/04/2023]
Abstract
WTAP, a highly conserved Wilms' tumor 1 interacting protein, is involved in a variety of biological processes. However, functional studies of WTAP in planarians have not been reported. In this study, we examined the spatiotemporal expression pattern of planarian DjWTAP and investigated its functions in planarians regeneration and homeostasis. Knocking-down DjWTAP resulted in severe morphological defects leading to lethality within 20 days. Silencing DjWTAP promoted the proliferation of PiwiA+ cells but impaired the lineage differentiation of epidermal, neural, digestive, and excretory cell types, suggesting a critical role for DjWTAP in stem cell self-renewal and differentiation in planarian. To further investigate the mechanisms underlying the defective differentiation, RNA-seq was employed to determine the transcriptomic alterations upon DjWTAP RNA interference. Histone 4 (H4), Histone-lysine N-methyltransferase-SETMAR like, and TNF receptor-associated factor 6 (TRAF6), were significantly upregulated in response to DjWTAP RNAi. Knocking-down TRAF6 largely rescued the defective tissue homeostasis and regeneration resulted from DjWTAP knockdown in planarians, suggesting that DjWTAP maintains planarian regeneration and homeostasis via TRAF6.
Collapse
Affiliation(s)
- Hui Zhen
- Guangdong Cardiovascular Institute, Medical Research Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Mujie Huang
- Guangdong Cardiovascular Institute, Medical Research Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Mingyue Zheng
- Guangdong Cardiovascular Institute, Medical Research Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Lili Gao
- Laboratory of Developmental and Evolutionary Biology, Shandong University of Technology, Zibo, China
| | - Zepeng Guo
- Guangdong Cardiovascular Institute, Medical Research Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Qiuxiang Pang
- Laboratory of Developmental and Evolutionary Biology, Shandong University of Technology, Zibo, China.
| | - Guoxiang Jin
- Guangdong Cardiovascular Institute, Medical Research Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Guangdong Provincial Geriatrics Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, China.
| | - Zhongjun Zhou
- Guangdong Cardiovascular Institute, Medical Research Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; School of Biomedical Sciences, The University of Hong Kong, Hong Kong; Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, CHINA.
| |
Collapse
|
36
|
Hulett RE, Gehrke AR, Gompers A, Rivera-López C, Srivastava M. A wound-induced differentiation trajectory for neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.10.540286. [PMID: 37214981 PMCID: PMC10197691 DOI: 10.1101/2023.05.10.540286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Animals capable of whole-body regeneration can replace any missing cell type and regenerate fully-functional new organs, de novo . The regeneration of a new brain requires the formation of diverse neuronal cell types and their assembly into an organized structure and correctly-wired circuits. Recent work in various regenerative animals has revealed transcriptional programs required for the differentiation of distinct neuronal subpopulations, however how these transcriptional programs are initiated upon amputation remains unknown. Here, we focused on the highly regenerative acoel worm, Hofstenia miamia , to study wound-induced transcriptional regulatory events that lead to the production of neurons. Footprinting analysis using chromatin accessibility data on an improved genome assembly revealed that binding sites for the NFY transcription factor complex were significantly bound during regeneration, showing a dynamic increase in binding within one hour upon amputation specifically in tail fragments, which will regenerate a new brain. Strikingly, NFY targets were highly enriched for genes with neuronal functional. Single-cell transcriptome analysis combined with functional studies identified sox4 + stem cells as the likely progenitor population for multiple neuronal subtypes. Further, we found that wound-induced sox4 expression is likely under direct transcriptional control by NFY, uncovering a mechanism for how early wound-induced binding of a transcriptional regulator results in the initiation of a neuronal differentiation pathway. Highlights A new chromosome-scale assembly for Hofstenia enables comprehensive analysis of transcription factor binding during regeneration NFY motifs become dynamically bound by 1hpa in regenerating tail fragments, particularly in the loci of neural genes A sox4 + neural-specialized stem cell is identified using scRNA-seq sox4 is wound-induced and required for differentiation of multiple neural cell types NFY regulates wound-induced expression of sox4 during regeneration.
Collapse
|
37
|
Ye K, Liu X, Li D, Gao L, Zheng K, Qu J, Xing N, Yang F, Liu B, Li A, Pang Q. Extracellular matrix-regulator MMPA is required for the orderly proliferation of neoblasts and differentiation of ectodermal progenitor cells in the planarian Dugesia japonica. Biochem Biophys Res Commun 2023; 659:1-9. [PMID: 37030019 DOI: 10.1016/j.bbrc.2023.03.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/09/2023] [Accepted: 03/14/2023] [Indexed: 04/05/2023]
Abstract
Matrix metalloproteinases (MMPs) are members of a family of zinc-dependent metallopeptidase proteins that are widely found in plants, animals, and microorganisms. As the regulators of the extracellular matrix and basement membrane, MMPs play an important role in embryogenesis, development, innate immunity, and regeneration. However, the function of MMP family in planarian, a model for regeneration research, is still ambiguous. Here, we cloned 5 MMPs genes from Dugesia japonica and found that DjMMPA was associated with the process of regeneration, neoblasts cell maintenance confusion and destruction. Loss of DjMMPA led to homeostasis confusion and eventually death, owing to neoblasts proliferation disorder. Additionally, DjMMPA RNAi-treated animals had impaired regeneration after amputation. Furthermore, knockdown of DjMMPA had noticeable defects in cell differentiation of ectoderm, especially in eyes and neural progenitor cells, possibly by inhibiting Wnt signaling. Our results suggest that extracellular matrix-regulator MMPA is required for the orderly proliferation of neoblasts and differentiation of ectodermal progenitor cells in the planarian, which provide valuable information for further explorations into the molecular mechanism of MMPS, stem cells, and regeneration.
Collapse
|
38
|
Fan Y, Chai C, Li P, Zou X, Ferrell JE, Wang B. Ultrafast and long-range coordination of wound responses is essential for whole-body regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.15.532844. [PMID: 36993633 PMCID: PMC10055111 DOI: 10.1101/2023.03.15.532844] [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: 05/23/2023]
Abstract
Injury induces systemic, global responses whose functions remain elusive. In addition, mechanisms that rapidly synchronize wound responses through long distances across the organismal scale are mostly unknown. Using planarians, which have extreme regenerative ability, we report that injury induces Erk activity to travel in a wave-like manner at an unexpected speed (∼1 mm/h), 10-100 times faster than those measured in other multicellular tissues. This ultrafast signal propagation requires longitudinal body-wall muscles, elongated cells forming dense parallel tracks running the length of the organism. Combining experiments and computational models, we show that the morphological properties of muscles allow them to minimize the number of slow intercellular signaling steps and act as bidirectional superhighways for propagating wound signals and instructing responses in other cell types. Inhibiting Erk propagation prevents cells distant to the wound from responding and blocks regeneration, which can be rescued by a second injury to distal tissues within a narrow time window after the first injury. These results suggest that rapid responses in uninjured tissues far from wounds are essential for regeneration. Our findings provide a mechanism for long-range signal propagation in large and complex tissues to coordinate cellular responses across diverse cell types, and highlights the function of feedback between spatially separated tissues during whole-body regeneration.
Collapse
Affiliation(s)
- Yuhang Fan
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Chew Chai
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Pengyang Li
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Xinzhi Zou
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - James E. Ferrell
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - Bo Wang
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| |
Collapse
|
39
|
Rennolds CW, Bely AE. Integrative biology of injury in animals. Biol Rev Camb Philos Soc 2023; 98:34-62. [PMID: 36176189 PMCID: PMC10087827 DOI: 10.1111/brv.12894] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 07/29/2022] [Accepted: 08/02/2022] [Indexed: 01/12/2023]
Abstract
Mechanical injury is a prevalent challenge in the lives of animals with myriad potential consequences for organisms, including reduced fitness and death. Research on animal injury has focused on many aspects, including the frequency and severity of wounding in wild populations, the short- and long-term consequences of injury at different biological scales, and the variation in the response to injury within or among individuals, species, ontogenies, and environmental contexts. However, relevant research is scattered across diverse biological subdisciplines, and the study of the effects of injury has lacked synthesis and coherence. Furthermore, the depth of knowledge across injury biology is highly uneven in terms of scope and taxonomic coverage: much injury research is biomedical in focus, using mammalian model systems and investigating cellular and molecular processes, while research at organismal and higher scales, research that is explicitly comparative, and research on invertebrate and non-mammalian vertebrate species is less common and often less well integrated into the core body of knowledge about injury. The current state of injury research presents an opportunity to unify conceptually work focusing on a range of relevant questions, to synthesize progress to date, and to identify fruitful avenues for future research. The central aim of this review is to synthesize research concerning the broad range of effects of mechanical injury in animals. We organize reviewed work by four broad and loosely defined levels of biological organization: molecular and cellular effects, physiological and organismal effects, behavioural effects, and ecological and evolutionary effects of injury. Throughout, we highlight the diversity of injury consequences within and among taxonomic groups while emphasizing the gaps in taxonomic coverage, causal understanding, and biological endpoints considered. We additionally discuss the importance of integrating knowledge within and across biological levels, including how initial, localized responses to injury can lead to long-term consequences at the scale of the individual animal and beyond. We also suggest important avenues for future injury biology research, including distinguishing better between related yet distinct injury phenomena, expanding the subjects of injury research to include a greater variety of species, and testing how intrinsic and extrinsic conditions affect the scope and sensitivity of injury responses. It is our hope that this review will not only strengthen understanding of animal injury but will contribute to building a foundation for a more cohesive field of 'injury biology'.
Collapse
|
40
|
Liu X, Sun Y, Wang S, Zhang S, Tian Q. Actin restricts cell proliferation and promotes differentiation during planarian regeneration. Biochem Biophys Res Commun 2023; 640:150-156. [PMID: 36508928 DOI: 10.1016/j.bbrc.2022.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 12/03/2022] [Indexed: 12/11/2022]
Abstract
Actin is an integral component of the cytoskeleton, which plays an important role in various fundamental cellular processes, such as affecting the polarity of embryonic cells during embryonic development in various model organisms. Meanwhile, previous studies have demonstrated that the polymerization of the actin cytoskeleton can affect cell migration, proliferation, and differentiation. Actin polymerization state regulated osteogenic differentiation and affected cell proliferation. However, the function of actin in regenerative biology has not been thoroughly elucidated. The planarian flatworm, which contains a large number of adult somatic stem cells (neoblasts), is an ideal model organism to study regenerative biology. Here, we identified a homolog of actin in planarian Dugesia japonica and found that RNAi targeting actin during planarian regeneration results in the formation of protrusions on the dorsal side, where the division of phospho-H3 mitotic cells is increased. In addition, a decrease in differentiation is observed in regenerating tissues after Djactin RNAi. These results indicate that Djactin functions in proliferation and differentiation control in planarian regeneration.
Collapse
Affiliation(s)
- Xiaomai Liu
- School of LifeSciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Yujia Sun
- School of LifeSciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Shaocong Wang
- School of LifeSciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Shoutao Zhang
- School of LifeSciences, Zhengzhou University, Zhengzhou, Henan, China; Longhu Laboratory of Advanced Immunology, Zhengzhou, Henan, China.
| | - Qingnan Tian
- School of LifeSciences, Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
41
|
Drees L, Rink JC. The planarian flatworm Schmidtea mediterranea. Nat Methods 2023; 20:3-5. [PMID: 36635538 DOI: 10.1038/s41592-022-01727-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Leonard Drees
- Department of Tissue Dynamics and Regeneration, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Jochen C Rink
- Department of Tissue Dynamics and Regeneration, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.
| |
Collapse
|
42
|
Yang Y, Wang L, Zhao Y, Ma F, Lin Z, Liu Y, Dong Z, Chen G, Liu D. PBDEs disrupt homeostasis maintenance and regeneration of planarians due to DNA damage, proliferation and apoptosis anomaly. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 248:114287. [PMID: 36371889 DOI: 10.1016/j.ecoenv.2022.114287] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 11/03/2022] [Accepted: 11/09/2022] [Indexed: 06/16/2023]
Abstract
Polybrominated diphenyl ethers (PBDEs) are widely used as brominated flame retardants in the manufacturing industry, belonging to persistent organic pollutants in the environment. Planarians are the freshwater worms, with strong regenerative ability and extreme sensitivity to environmental toxicants. This study aimed to evaluate the potential acute comprehensive effects of PBDE-47/-209 on freshwater planarians. Methods to detect the effects include: detection of oxidative stress, observation of morphology and histology, detection of DNA fragmentation, and detection of cell proliferation and apoptosis. In the PBDE-47 treatment group, planarians showed increased oxidative stress intensity, severe tissue damage, increased DNA fragmentation level, and increased cell proliferation and apoptosis. In the PBDE-209 treatment group, planarians showed decreased oxidative stress intensity, slight tissue damage, almost unchanged DNA fragmentation level and apoptosis, proliferation increased only on the first day after treatment. In conclusion, both PBDE-47 and PBDE-209 are dangerous environmental hazardous material that can disrupt planarians homeostasis, while the toxicity of PBDE-47 is sever than PBDE-209 that PBDE-47 can lead to the death of planarians.
Collapse
Affiliation(s)
- Yibo Yang
- College of Life Science, Henan Normal University, Xinxiang City, 453007 Henan, China
| | - Lei Wang
- College of Life Science, Henan Normal University, Xinxiang City, 453007 Henan, China
| | - Yuhao Zhao
- College of Life Science, Henan Normal University, Xinxiang City, 453007 Henan, China
| | - Fuhao Ma
- College of Life Science, Henan Normal University, Xinxiang City, 453007 Henan, China
| | - Ziyi Lin
- College of Life Science, Henan Normal University, Xinxiang City, 453007 Henan, China
| | - Yingyu Liu
- College of Life Science, Henan Normal University, Xinxiang City, 453007 Henan, China
| | - Zimei Dong
- College of Life Science, Henan Normal University, Xinxiang City, 453007 Henan, China.
| | - Guangwen Chen
- College of Life Science, Henan Normal University, Xinxiang City, 453007 Henan, China.
| | - Dezeng Liu
- College of Life Science, Henan Normal University, Xinxiang City, 453007 Henan, China
| |
Collapse
|
43
|
Hendin N, Gordon T, Shenkar N, Wurtzel O. Molecular characterization of the immediate wound response of the solitary ascidian Polycarpa mytiligera. Dev Dyn 2022; 251:1968-1981. [PMID: 36001356 PMCID: PMC10087333 DOI: 10.1002/dvdy.526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 06/16/2022] [Accepted: 07/05/2022] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Injury response is key to successful regeneration. Yet, transcriptome analyses of injury response were performed only on a handful of regenerative organisms. Here, we studied the injury response of the solitary ascidian Polycarpa mytiligera, an emerging model system, capable of regenerating any body part. We used the siphon as a model for studying transcriptional changes following injury, and identified genes that were activated in the initial 24 hours post amputation (hpa). RESULTS Highly conserved genes, such as bone morphogenetic protein-1 (BMP1), growth hormone secretagogue receptor (GHSR) and IL-17, were upregulated by 12 hpa, yet their expression was sustained only in non-regenerating tissue fragments. We optimized fluorescent in situ hybridization, and found that the majority of BMP1+ cells were localized to the rigid tunic that covers the animal. This highlights the importance of this tissue, particularly during injury response. BMP1 was overexpressed following injuries to other body regions, suggesting that it was a part of a common injury-induced program. CONCLUSION Our study suggests that, initially, specific injury-induced genes were upregulated in P. mytiligera organs, yet, later, a unique transcriptional profile was observed only in regenerating tissues. These findings highlight the importance of studying diverse regenerating and non-regenerating organisms for complete understanding of regeneration.
Collapse
Affiliation(s)
- Noam Hendin
- The School of Neurobiology, Biochemistry & Biophysics, George S. Wise Faculty of Life SciencesTel Aviv UniversityTel AvivIsrael
| | - Tal Gordon
- The School of Neurobiology, Biochemistry & Biophysics, George S. Wise Faculty of Life SciencesTel Aviv UniversityTel AvivIsrael
| | - Noa Shenkar
- School of Zoology, George S. Wise Faculty of Life SciencesTel Aviv UniversityTel AvivIsrael
- The Steinhardt Museum of Natural History, Israel National Center for Biodiversity StudiesTel‐Aviv UniversityTel‐AvivIsrael
| | - Omri Wurtzel
- The School of Neurobiology, Biochemistry & Biophysics, George S. Wise Faculty of Life SciencesTel Aviv UniversityTel AvivIsrael
- Sagol School of NeuroscienceTel Aviv UniversityTel AvivIsrael
| |
Collapse
|
44
|
Angileri KM, Bagia NA, Feschotte C. Transposon control as a checkpoint for tissue regeneration. Development 2022; 149:dev191957. [PMID: 36440631 PMCID: PMC10655923 DOI: 10.1242/dev.191957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 10/03/2022] [Indexed: 11/29/2022]
Abstract
Tissue regeneration requires precise temporal control of cellular processes such as inflammatory signaling, chromatin remodeling and proliferation. The combination of these processes forms a unique microenvironment permissive to the expression, and potential mobilization of, transposable elements (TEs). Here, we develop the hypothesis that TE activation creates a barrier to tissue repair that must be overcome to achieve successful regeneration. We discuss how uncontrolled TE activity may impede tissue restoration and review mechanisms by which TE activity may be controlled during regeneration. We posit that the diversification and co-evolution of TEs and host control mechanisms may contribute to the wide variation in regenerative competency across tissues and species.
Collapse
Affiliation(s)
- Krista M. Angileri
- Department of Molecular Biology and Genetics, Cornell University, 526 Campus Rd, Ithaca, NY 14850, USA
| | - Nornubari A. Bagia
- Department of Molecular Biology and Genetics, Cornell University, 526 Campus Rd, Ithaca, NY 14850, USA
| | - Cedric Feschotte
- Department of Molecular Biology and Genetics, Cornell University, 526 Campus Rd, Ithaca, NY 14850, USA
| |
Collapse
|
45
|
Dagan Y, Yesharim Y, Bonneau AR, Frankovits T, Schwartz S, Reddien PW, Wurtzel O. m6A is required for resolving progenitor identity during planarian stem cell differentiation. EMBO J 2022; 41:e109895. [PMID: 35971838 PMCID: PMC9627665 DOI: 10.15252/embj.2021109895] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 07/10/2022] [Accepted: 07/12/2022] [Indexed: 12/13/2022] Open
Abstract
Regeneration and tissue homeostasis require accurate production of missing cell lineages. Cell production is driven by changes to gene expression, which is shaped by multiple layers of regulation. Here, we find that the ubiquitous mRNA base-modification, m6A, is required for proper cell fate choice and cellular maturation in planarian stem cells (neoblasts). We mapped m6A-enriched regions in 7,600 planarian genes and found that perturbation of the m6A pathway resulted in progressive deterioration of tissues and death. Using single-cell RNA sequencing of >20,000 cells following perturbation of the m6A pathway, we identified an increase in expression of noncanonical histone variants, and that inhibition of the pathway resulted in accumulation of undifferentiated cells throughout the animal in an abnormal transcriptional state. Analysis of >1,000 planarian gene expression datasets revealed that the inhibition of the chromatin modifying complex NuRD had almost indistinguishable consequences, unraveling an unappreciated link between m6A and chromatin modifications. Our findings reveal that m6A is critical for planarian stem cell homeostasis and gene regulation in tissue maintenance and regeneration.
Collapse
Affiliation(s)
- Yael Dagan
- The George S. Wise Faculty of Life Sciences, School of Neurobiology, Biochemistry, and BiophysicsTel Aviv UniversityTel AvivIsrael
| | - Yarden Yesharim
- The George S. Wise Faculty of Life Sciences, School of Neurobiology, Biochemistry, and BiophysicsTel Aviv UniversityTel AvivIsrael
| | - Ashley R Bonneau
- Whitehead Institute for Biomedical ResearchCambridgeMAUSA
- Department of BiologyMassachusetts Institute of TechnologyCambridgeMAUSA
- Howard Hughes Medical InstituteChevy ChaseMDUSA
| | - Tamar Frankovits
- The George S. Wise Faculty of Life Sciences, School of Neurobiology, Biochemistry, and BiophysicsTel Aviv UniversityTel AvivIsrael
| | - Schraga Schwartz
- Department of Molecular GeneticsWeizmann Institute of ScienceRehovotIsrael
| | - Peter W Reddien
- Whitehead Institute for Biomedical ResearchCambridgeMAUSA
- Department of BiologyMassachusetts Institute of TechnologyCambridgeMAUSA
- Howard Hughes Medical InstituteChevy ChaseMDUSA
| | - Omri Wurtzel
- The George S. Wise Faculty of Life Sciences, School of Neurobiology, Biochemistry, and BiophysicsTel Aviv UniversityTel AvivIsrael
- Sagol School of NeuroscienceTel Aviv UniversityTel AvivIsrael
| |
Collapse
|
46
|
Quiroga-Artigas G, de Jong D, Schnitzler CE. GNL3 is an evolutionarily conserved stem cell gene influencing cell proliferation, animal growth and regeneration in the hydrozoan Hydractinia. Open Biol 2022; 12:220120. [PMID: 36069077 PMCID: PMC9449814 DOI: 10.1098/rsob.220120] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Nucleostemin (NS) is a vertebrate gene preferentially expressed in stem and cancer cells, which acts to regulate cell cycle progression, genome stability and ribosome biogenesis. NS and its paralogous gene, GNL3-like (GNL3L), arose in the vertebrate clade after a duplication event from their orthologous gene, G protein Nucleolar 3 (GNL3). Research on invertebrate GNL3, however, has been limited. To gain a greater understanding of the evolution and functions of the GNL3 gene, we have performed studies in the hydrozoan cnidarian Hydractinia symbiolongicarpus, a colonial hydroid that continuously generates pluripotent stem cells throughout its life cycle and presents impressive regenerative abilities. We show that Hydractinia GNL3 is expressed in stem and germline cells. The knockdown of GNL3 reduces the number of mitotic and S-phase cells in Hydractinia larvae of different ages. Genome editing of Hydractinia GNL3 via CRISPR/Cas9 resulted in colonies with reduced growth rates, polyps with impaired regeneration capabilities, gonadal morphological defects, and low sperm motility. Collectively, our study shows that GNL3 is an evolutionarily conserved stem cell and germline gene involved in cell proliferation, animal growth, regeneration and sexual reproduction in Hydractinia, and sheds new light into the evolution of GNL3 and of stem cell systems.
Collapse
Affiliation(s)
- Gonzalo Quiroga-Artigas
- Whitney Laboratory for Marine Bioscience, University of Florida, St Augustine, FL 32080, USA
| | - Danielle de Jong
- Whitney Laboratory for Marine Bioscience, University of Florida, St Augustine, FL 32080, USA
| | - Christine E Schnitzler
- Whitney Laboratory for Marine Bioscience, University of Florida, St Augustine, FL 32080, USA.,Department of Biology, University of Florida, Gainesville, FL, USA
| |
Collapse
|
47
|
Wyss LS, Bray SR, Wang B. Cellular diversity and developmental hierarchy in the planarian nervous system. Curr Opin Genet Dev 2022; 76:101960. [PMID: 35878572 DOI: 10.1016/j.gde.2022.101960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/14/2022] [Accepted: 06/21/2022] [Indexed: 12/01/2022]
Abstract
Our ability to dissect cell type diversity, development, and plasticity in the nervous system has been transformed by the recent surge of massive sequencing studies at the single-cell level. A large body of this work has focused primarily on organisms with nervous systems established early in development. Using planarian flatworms in which neurons are constantly respecified, replenished, and regenerated, we analyze several existing single-cell transcriptomic datasets and observe features in neuron identity, differentiation, maturation, and function that may provide the planarian nervous system with high levels of adaptability required to respond to various cues including injury. This analysis allows us to place many prior observations made by functional characterizations in a general framework and provide additional hypothesis and predictions to test in future investigations.
Collapse
Affiliation(s)
- Livia S Wyss
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Samuel R Bray
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Bo Wang
- Department of Bioengineering, Stanford University, Stanford, CA, USA.
| |
Collapse
|
48
|
Djptpn11 is indispensable for planarian regeneration by affecting early wound response genes expression and the Wnt pathway. Biochimie 2022; 201:184-195. [PMID: 35868605 DOI: 10.1016/j.biochi.2022.07.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 07/14/2022] [Accepted: 07/15/2022] [Indexed: 12/22/2022]
Abstract
Planarian is an ideal model system of studying regeneration. Stem cell system and positional control genes (PCGs) are two important factors for perfect regeneration of planarians and they combine to promote their regeneration. Even so, how wounds regulate proliferation and neoblast fate is still important areas to address. Ptpn11 (Protein tyrosine phosphatase non-receptor type 11), one of PTP (Protein tyrosine phosphatase) family members, plays an important role in cellular processes including cell survival, proliferation, differentiation and apoptosis. Nevertheless, the role of ptpn11 in the planarian regeneration has not been fully studied. In this study, we identify the Djptpn11 gene to observe its function in planarian regeneration. The results reveal that the regeneration is severely inhibited and cause the disorder homeostasis in planarians. Furthermore, the stem cells proliferation and differentiation decreases while the apoptosis increases following Djptpn11 RNAi. At the same time, Djptpn11 affects the expression levels of early wound response genes (Djegr2, Dj1-jun, Djrunt1, Djwnt1 and Djnotum). Djwnt1 and Djnotum are two key Wnt signaling pathway genes and Djptpn11 affects the expression levels of Djwnt1 and Djnotum in the early and late stages of planarian regeneration. In general, Djptpn11 is indispensable for the homeostasis and regeneration of planarian by affecting the stem cells, early wound response genes and the Wnt pathway.
Collapse
|
49
|
Ge XY, Han X, Zhao YL, Cui GS, Yang YG. An insight into planarian regeneration. Cell Prolif 2022; 55:e13276. [PMID: 35811385 PMCID: PMC9436907 DOI: 10.1111/cpr.13276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/22/2022] [Accepted: 05/17/2022] [Indexed: 11/29/2022] Open
Abstract
Background Planarian has attracted increasing attentions in the regeneration field for its usefulness as an important biological model organism attributing to its strong regeneration ability. Both the complexity of multiple regulatory networks and their coordinate functions contribute to the maintenance of normal cellular homeostasis and the process of regeneration in planarian. The polarity, size, location and number of regeneration tissues are regulated by diverse mechanisms. In this review we summarize the recent advances about the importance genetic and molecular mechanisms for regeneration control on various tissues in planarian. Methods A comprehensive literature search of original articles published in recent years was performed in regards to the molecular mechanism of each cell types during the planarian regeneration, including neoblast, nerve system, eye spot, excretory system and epidermal. Results Available molecular mechanisms gave us an overview of regeneration process in every tissue. The sense of injuries and initiation of regeneration is regulated by diverse genes like follistatin and ERK signaling. The Neoblasts differentiate into tissue progenitors under the regulation of genes such as egfr‐3. The regeneration polarity is controlled by Wnt pathway, BMP pathway and bioelectric signals. The neoblast within the blastema differentiate into desired cell types and regenerate the missing tissues. Those tissue specific genes regulate the tissue progenitor cells to differentiate into desired cell types to complete the regeneration process. Conclusion All tissue types in planarian participate in the regeneration process regulated by distinct molecular factors and cellular signaling pathways. The neoblasts play vital roles in tissue regeneration and morphology maintenance. These studies provide new insights into the molecular mechanisms for regulating planarian regeneration.
Collapse
Affiliation(s)
- Xin-Yang Ge
- CAS Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, College of Future Technology, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China.,Sino-Danish College, University of Chinese Academy of Sciences, Beijing, China.,China National Center for Bioinformation, Beijing, China
| | - Xiao Han
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yong-Liang Zhao
- CAS Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, College of Future Technology, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China.,Sino-Danish College, University of Chinese Academy of Sciences, Beijing, China.,China National Center for Bioinformation, Beijing, China
| | - Guan-Shen Cui
- CAS Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, College of Future Technology, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China.,China National Center for Bioinformation, Beijing, China
| | - Yun-Gui Yang
- CAS Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, College of Future Technology, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China.,Sino-Danish College, University of Chinese Academy of Sciences, Beijing, China.,China National Center for Bioinformation, Beijing, China
| |
Collapse
|
50
|
Wong LL, Bruxvoort CG, Cejda NI, Delaney MR, Otero JR, Forsthoefel DJ. Intestine-enriched apolipoprotein b orthologs are required for stem cell progeny differentiation and regeneration in planarians. Nat Commun 2022; 13:3803. [PMID: 35778403 PMCID: PMC9249923 DOI: 10.1038/s41467-022-31385-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 06/16/2022] [Indexed: 02/08/2023] Open
Abstract
Lipid metabolism plays an instructive role in regulating stem cell state and differentiation. However, the roles of lipid mobilization and utilization in stem cell-driven regeneration are unclear. Planarian flatworms readily restore missing tissue due to injury-induced activation of pluripotent somatic stem cells called neoblasts. Here, we identify two intestine-enriched orthologs of apolipoprotein b, apob-1 and apob-2, which mediate transport of neutral lipid stores from the intestine to target tissues including neoblasts, and are required for tissue homeostasis and regeneration. Inhibition of apob function by RNAi causes head regression and lysis in uninjured animals, and delays body axis re-establishment and regeneration of multiple organs in amputated fragments. Furthermore, apob RNAi causes expansion of the population of differentiating neoblast progeny and dysregulates expression of genes enriched in differentiating and mature cells in eight major cell type lineages. We conclude that intestine-derived lipids serve as a source of metabolites required for neoblast progeny differentiation.
Collapse
Affiliation(s)
- Lily L Wong
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Christina G Bruxvoort
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Veteran Affairs Medical Center - Research Services, Oklahoma City, OK, USA
| | - Nicholas I Cejda
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Center for Biomedical Data Science, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Matthew R Delaney
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Jannette Rodriguez Otero
- Howard Hughes Medical Institute, Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Department of Education, Universidad Interamericana de Puerto Rico, San Juan, Puerto Rico, USA
| | - David J Forsthoefel
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA.
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|