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
|
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
|
4
|
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
|
5
|
Beinart FR, Gillen K. Regeneration of Lumbriculus variegatus requires post-amputation production of reactive oxygen species. Dev Growth Differ 2025; 67:104-112. [PMID: 39837571 PMCID: PMC11842891 DOI: 10.1111/dgd.12961] [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: 08/08/2024] [Revised: 01/02/2025] [Accepted: 01/04/2025] [Indexed: 01/23/2025]
Abstract
Animals vary in their ability to replace body parts lost to injury, a phenomenon known as restorative regeneration. Uncovering conserved signaling steps required for regeneration may aid regenerative medicine. Reactive oxygen species (ROS) are necessary for proper regeneration in species across a wide range of taxa, but it is unknown whether ROS are essential for annelid regeneration. As annelids are a widely used and excellent model for regeneration, we sought to determine whether ROS play a role in the regeneration of the highly regenerative annelid, Lumbriculus variegatus. Using a ROS-sensitive fluorescent probe we observed ROS accumulation at the wound site within 15 min after amputation; this ROS burst lessened by 6 h post-amputation. Chemical inhibition of this ROS burst delayed regeneration, an impairment that was partially rescued with exogenous ROS. Our results suggest that similar to other animals, annelid regeneration depends upon ROS signaling, implying a phylogenetically ancient requirement for ROS in regeneration.
Collapse
Affiliation(s)
- Freya R. Beinart
- Kenyon CollegeMolecular BiologyGambierOhioUSA
- Present address:
Washington UniversitySt. LouisMissouriUSA
| | - Kathy Gillen
- Kenyon CollegeMolecular BiologyGambierOhioUSA
- Kenyon CollegeBiologyGambierOhioUSA
| |
Collapse
|
6
|
Tan YR, Roan HY, Chen CH. Zebrafish tailfin as an in vivo model for capturing tissue-scale cell dynamics. Semin Cell Dev Biol 2025; 166:29-35. [PMID: 39724824 DOI: 10.1016/j.semcdb.2024.12.005] [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: 08/14/2024] [Revised: 12/11/2024] [Accepted: 12/13/2024] [Indexed: 12/28/2024]
Abstract
The intricate control of collective cell dynamics is crucial for enabling organismic development and tissue regeneration. Despite the availability of various in vitro and in vivo models, studies on tissue-scale cell dynamics and associated emergent properties in living systems remain methodically challenging. Here, we describe key advantages of using the adult zebrafish tailfin (caudal fin) as a robust in vivo model for dissecting millimeter-scale collective cell dynamics during regeneration and wound healing in a complex tissue. For researchers considering this model system, we briefly introduce the tailfin anatomy, as well as available transgenic reporter tools and live-imaging setups that may be utilized to study epidermal cell behaviors. To highlight the unique strengths of the zebrafish tailfin model, we present an example project that was made possible by techniques for tracking cell dynamics at a millimeter scale with single-cell resolution in live animals. Finally, we discuss the research directions at the interface of collective cell dynamics and regenerative biology that most excite us and can be examined using the tailfin model.
Collapse
Affiliation(s)
- Yue Rong Tan
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Hsiao-Yuh Roan
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Chen-Hui Chen
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan.
| |
Collapse
|
7
|
Rich A, Lu Z, Simone AD, Garcia L, Janssen J, Ando K, Ou J, Vergassola M, Poss KD, Talia SD. Decaying and expanding Erk gradients process memory of skeletal size during zebrafish fin regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.23.634576. [PMID: 39896678 PMCID: PMC11785216 DOI: 10.1101/2025.01.23.634576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Regeneration of an amputated salamander limb or fish fin restores pre-injury size and structure, illustrating the phenomenon of positional memory. Although appreciated for centuries, the identity of position-dependent cues and how they control tissue growth are not resolved. Here, we quantify Erk signaling events in whole populations of osteoblasts during zebrafish fin regeneration. We find that osteoblast Erk activity is dependent on Fgf receptor signaling and organized into millimeter-long gradients that extend from the distal tip to the amputation site. Erk activity scales with the amount of tissue amputated, predicts the likelihood of osteoblast cycling, and predicts the size of regenerated skeletal structures. Mathematical modeling suggests gradients are established by the transient deposition of long-lived ligands that are transported by tissue growth. This concept is supported by the observed scaling of expression of the essential epidermal ligand fgf20a with extents of amputation. Our work provides evidence that localized, scaled expression of pro-regenerative ligands instructs long-range signaling and cycling to control skeletal size in regenerating appendages.
Collapse
Affiliation(s)
- Ashley Rich
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
- Duke Center for Quantitative Living Systems, Duke University Medical Center, Durham, NC, USA
| | - Ziqi Lu
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
- Duke Center for Quantitative Living Systems, Duke University Medical Center, Durham, NC, USA
| | - Alessandro De Simone
- Department of Genetics and Evolution, University of Geneva, 1211 Geneva, Switzerland
| | - Lucas Garcia
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
| | | | - Kazunori Ando
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
- Duke Regeneration Center, Duke University Medical Center, Durham, NC, USA
- Morgridge Institute for Research, Madison WI USA
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI, USA
| | - Jianhong Ou
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
- Duke Regeneration Center, Duke University Medical Center, Durham, NC, USA
- Morgridge Institute for Research, Madison WI USA
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI, USA
| | - Massimo Vergassola
- Department of Physics, École Normale Supérieure, Paris 75005, France
- Department of Physics, University of California, San Diego, CA, USA
| | - Kenneth D. Poss
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
- Duke Regeneration Center, Duke University Medical Center, Durham, NC, USA
- Morgridge Institute for Research, Madison WI USA
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI, USA
| | - Stefano Di Talia
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
- Duke Center for Quantitative Living Systems, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
8
|
Mangiavacchi A, Morelli G, Reppe S, Saera-Vila A, Liu P, Eggerschwiler B, Zhang H, Bensaddek D, Casanova EA, Medina Gomez C, Prijatelj V, Della Valle F, Atinbayeva N, Izpisua Belmonte JC, Rivadeneira F, Cinelli P, Gautvik KM, Orlando V. LINE-1 RNA triggers matrix formation in bone cells via a PKR-mediated inflammatory response. EMBO J 2024; 43:3587-3603. [PMID: 38951609 PMCID: PMC11377738 DOI: 10.1038/s44318-024-00143-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: 08/08/2023] [Revised: 05/16/2024] [Accepted: 05/23/2024] [Indexed: 07/03/2024] Open
Abstract
Transposable elements (TEs) are mobile genetic modules of viral derivation that have been co-opted to become modulators of mammalian gene expression. TEs are a major source of endogenous dsRNAs, signaling molecules able to coordinate inflammatory responses in various physiological processes. Here, we provide evidence for a positive involvement of TEs in inflammation-driven bone repair and mineralization. In newly fractured mice bone, we observed an early transient upregulation of repeats occurring concurrently with the initiation of the inflammatory stage. In human bone biopsies, analysis revealed a significant correlation between repeats expression, mechanical stress and bone mineral density. We investigated a potential link between LINE-1 (L1) expression and bone mineralization by delivering a synthetic L1 RNA to osteoporotic patient-derived mesenchymal stem cells and observed a dsRNA-triggered protein kinase (PKR)-mediated stress response that led to strongly increased mineralization. This response was associated with a strong and transient inflammation, accompanied by a global translation attenuation induced by eIF2α phosphorylation. We demonstrated that L1 transfection reshaped the secretory profile of osteoblasts, triggering a paracrine activity that stimulated the mineralization of recipient cells.
Collapse
Affiliation(s)
- Arianna Mangiavacchi
- King Abdullah University of Science and Technology (KAUST), Biological Environmental Science and Engineering Division, Thuwal, 23500-6900, Kingdom of Saudi Arabia.
| | - Gabriele Morelli
- King Abdullah University of Science and Technology (KAUST), Biological Environmental Science and Engineering Division, Thuwal, 23500-6900, Kingdom of Saudi Arabia
| | - Sjur Reppe
- Oslo University Hospital, Department of Medical Biochemistry, Oslo, Norway
- Lovisenberg Diaconal Hospital, Unger-Vetlesen Institute, Oslo, Norway
- Oslo University Hospital, Department of Plastic and Reconstructive Surgery, Oslo, Norway
| | | | - Peng Liu
- King Abdullah University of Science and Technology (KAUST), Biological Environmental Science and Engineering Division, Thuwal, 23500-6900, Kingdom of Saudi Arabia
| | - Benjamin Eggerschwiler
- Department of Trauma, University Hospital Zurich, Sternwartstrasse 14, 8091, Zurich, Switzerland
- Life Science Zurich Graduate School, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Huoming Zhang
- Core Labs, King Abdullah University of Science and Technology (KAUST), Thuwal, 23500-6900, Kingdom of Saudi Arabia
| | - Dalila Bensaddek
- Core Labs, King Abdullah University of Science and Technology (KAUST), Thuwal, 23500-6900, Kingdom of Saudi Arabia
| | - Elisa A Casanova
- Department of Trauma, University Hospital Zurich, Sternwartstrasse 14, 8091, Zurich, Switzerland
| | | | - Vid Prijatelj
- Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Francesco Della Valle
- King Abdullah University of Science and Technology (KAUST), Biological Environmental Science and Engineering Division, Thuwal, 23500-6900, Kingdom of Saudi Arabia
- Altos Labs, San Diego, CA, USA
| | - Nazerke Atinbayeva
- King Abdullah University of Science and Technology (KAUST), Biological Environmental Science and Engineering Division, Thuwal, 23500-6900, Kingdom of Saudi Arabia
| | | | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Paolo Cinelli
- Department of Trauma, University Hospital Zurich, Sternwartstrasse 14, 8091, Zurich, Switzerland
- Center for Applied Biotechnology and Molecular Medicine, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | | | - Valerio Orlando
- King Abdullah University of Science and Technology (KAUST), Biological Environmental Science and Engineering Division, Thuwal, 23500-6900, Kingdom of Saudi Arabia.
| |
Collapse
|
9
|
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
|
10
|
Huang L, Ho C, Ye X, Gao Y, Guo W, Chen J, Sun J, Wen D, Liu Y, Liu Y, Zhang Y, Li Q. Mechanisms and translational applications of regeneration in limbs: From renewable animals to humans. Ann Anat 2024; 255:152288. [PMID: 38823491 DOI: 10.1016/j.aanat.2024.152288] [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: 10/02/2023] [Revised: 04/08/2024] [Accepted: 05/27/2024] [Indexed: 06/03/2024]
Abstract
BACKGROUND The regenerative capacity of organisms declines throughout evolution, and mammals lack the ability to regenerate limbs after injury. Past approaches to achieving successful restoration through pharmacological intervention, tissue engineering, and cell therapies have faced significant challenges. OBJECTIVES This review aims to provide an overview of the current understanding of the mechanisms behind animal limb regeneration and the successful translation of these mechanisms for human tissue regeneration. RESULTS Particular attention was paid to the Mexican axolotl (Ambystoma mexicanum), the only adult tetrapod capable of limb regeneration. We will explore fundamental questions surrounding limb regeneration, such as how amputation initiates regeneration, how the limb knows when to stop and which parts to regenerate, and how these findings can apply to mammalian systems. CONCLUSIONS Given the urgent need for regenerative therapies to treat conditions like diabetic foot ulcers and trauma survivors, this review provides valuable insights and ideas for researchers, clinicians, and biomedical engineers seeking to facilitate the regeneration process or elicit full regeneration from partial regeneration events.
Collapse
Affiliation(s)
- Lu Huang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, China; Department of Ophthalmology, Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA.
| | - Chiakang Ho
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, China
| | - Xinran Ye
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, China
| | - Ya Gao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, China
| | - Weiming Guo
- Shanghai Key Laboratory of Stomatology, 639 Zhizaoju Road, Shanghai 200011, China; National Clinical Research Center for Oral Diseases, Shanghai 200011, China; National Center for Stomatology, Shanghai 200011, China; College of Stomatology, Shanghai Jiao Tong University, Shanghai 200011, China; Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, China
| | - Julie Chen
- Department of Ophthalmology, Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Jiaming Sun
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, China
| | - Dongsheng Wen
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, China
| | - Yangdan Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, China
| | - Yuxin Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, China
| | - Yifan Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, China.
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, China.
| |
Collapse
|
11
|
Tomasso A, Disela V, Longaker MT, Bartscherer K. Marvels of spiny mouse regeneration: cellular players and their interactions in restoring tissue architecture in mammals. Curr Opin Genet Dev 2024; 87:102228. [PMID: 39047585 DOI: 10.1016/j.gde.2024.102228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/12/2024] [Accepted: 06/30/2024] [Indexed: 07/27/2024]
Abstract
Understanding the cellular and molecular determinants of mammalian tissue regeneration and repair is crucial for developing effective therapies that restore tissue architecture and function. In this review, we focus on the cell types involved in scarless wound response and regeneration of spiny mice (Acomys). Comparative -omics approaches with scar-prone mammals have revealed species-specific peculiarities in cellular behavior during the divergent healing trajectories. We discuss the developing views on which cell types engage in restoring the architecture of spiny mouse tissues through a co-ordinated spatiotemporal response to injury. While yet at the beginning of understanding how cells interact in these fascinating animals to regenerate tissues, spiny mice hold great promise for scar prevention and anti-fibrotic treatments.
Collapse
Affiliation(s)
- Antonio Tomasso
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University - School of Medicine, Department of Surgery, Stanford, CA 94305, USA; Department of Biology/Chemistry, Osnabrück University, Osnabrück 49076, Germany; Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), Utrecht 3584CT, the Netherlands. https://twitter.com/@anto_tomasso
| | - Vanessa Disela
- Department of Biology/Chemistry, Osnabrück University, Osnabrück 49076, Germany; Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), Utrecht 3584CT, the Netherlands. https://twitter.com/@VDisela
| | - Michael T Longaker
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University - School of Medicine, Department of Surgery, Stanford, CA 94305, USA. https://twitter.com/@LongakerLab
| | - Kerstin Bartscherer
- Department of Biology/Chemistry, Osnabrück University, Osnabrück 49076, Germany.
| |
Collapse
|
12
|
Rajagopalan K, Selvan Christyraj JD, Chelladurai KS, Kalimuthu K, Das P, Chandrasekar M, Balamurugan N, Murugan K. Understanding the molecular mechanism of regeneration through apoptosis-induced compensatory proliferation studies - updates and future aspects. Apoptosis 2024:10.1007/s10495-024-01958-1. [PMID: 38581530 DOI: 10.1007/s10495-024-01958-1] [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] [Accepted: 03/10/2024] [Indexed: 04/08/2024]
Abstract
AICP is a crucial process that maintaining tissue homeostasis and regeneration. In the past, cell death was perceived merely as a means to discard cells without functional consequences. However, during regeneration, effector caspases orchestrate apoptosis, releasing signals that activate stem cells, thereby compensating for tissue loss across various animal models. Despite significant progress, the activation of Wnt3a by caspase-3 remains a focal point of research gaps in AICP mechanisms, spanning from lower to higher regenerative animals. This inquiry into the molecular intricacies of caspase-3-induced Wnt3a activation contributes to a deeper understanding of the links between regeneration and cancer mechanisms. Our report provides current updates on AICP pathways, delineating research gaps and highlighting the potential for future investigations aimed at enhancing our comprehension of this intricate process.
Collapse
Affiliation(s)
- Kamarajan Rajagopalan
- Molecular Biology and Stem Cell Research Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science and Technology (Deemed to be University), Chennai, Tamil Nadu, India
| | - Jackson Durairaj Selvan Christyraj
- Molecular Biology and Stem Cell Research Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science and Technology (Deemed to be University), Chennai, Tamil Nadu, India.
| | - Karthikeyan Subbiahanadar Chelladurai
- Molecular Biology and Stem Cell Research Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science and Technology (Deemed to be University), Chennai, Tamil Nadu, India
| | | | - Puja Das
- Molecular Biology and Stem Cell Research Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science and Technology (Deemed to be University), Chennai, Tamil Nadu, India
| | - Meikandan Chandrasekar
- Molecular Biology and Stem Cell Research Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science and Technology (Deemed to be University), Chennai, Tamil Nadu, India
| | - Nivedha Balamurugan
- Molecular Biology and Stem Cell Research Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science and Technology (Deemed to be University), Chennai, Tamil Nadu, India
| | - Karthikeyan Murugan
- Department of Biotechnology, Sri Venkateswara College of Engineering, Sriperumbudur, Tamil Nadu, India
| |
Collapse
|
13
|
Mitchell DG, Edgar A, Mateu JR, Ryan JF, Martindale MQ. The ctenophore Mnemiopsis leidyi deploys a rapid injury response dating back to the last common animal ancestor. Commun Biol 2024; 7:203. [PMID: 38374160 PMCID: PMC10876535 DOI: 10.1038/s42003-024-05901-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 02/08/2024] [Indexed: 02/21/2024] Open
Abstract
Regenerative potential is widespread but unevenly distributed across animals. However, our understanding of the molecular mechanisms underlying regenerative processes is limited to a handful of model organisms, restricting robust comparative analyses. Here, we conduct a time course of RNA-seq during whole body regeneration in Mnemiopsis leidyi (Ctenophora) to uncover gene expression changes that correspond with key events during the regenerative timeline of this species. We identified several genes highly enriched in this dataset beginning as early as 10 minutes after surgical bisection including transcription factors in the early timepoints, peptidases in the middle timepoints, and cytoskeletal genes in the later timepoints. We validated the expression of early response transcription factors by whole mount in situ hybridization, showing that these genes exhibited high expression in tissues surrounding the wound site. These genes exhibit a pattern of transient upregulation as seen in a variety of other organisms, suggesting that they may be initiators of an ancient gene regulatory network linking wound healing to the initiation of a regenerative response.
Collapse
Affiliation(s)
- Dorothy G Mitchell
- Whitney Laboratory for Marine Bioscience, University of Florida, Saint Augustine, FL, USA
- Department of Biology, University of Florida, Gainesville, FL, USA
| | - Allison Edgar
- Whitney Laboratory for Marine Bioscience, University of Florida, Saint Augustine, FL, USA
| | - Júlia Ramon Mateu
- Whitney Laboratory for Marine Bioscience, University of Florida, Saint Augustine, FL, USA
| | - Joseph F Ryan
- Whitney Laboratory for Marine Bioscience, University of Florida, Saint Augustine, FL, USA
- Department of Biology, University of Florida, Gainesville, FL, USA
| | - Mark Q Martindale
- Whitney Laboratory for Marine Bioscience, University of Florida, Saint Augustine, FL, USA.
- Department of Biology, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
14
|
Petersen M, Ebstrup E, Rodriguez E. Going through changes - the role of autophagy during reprogramming and differentiation. J Cell Sci 2024; 137:jcs261655. [PMID: 38393817 DOI: 10.1242/jcs.261655] [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: 02/25/2024] Open
Abstract
Somatic cell reprogramming is a complex feature that allows differentiated cells to undergo fate changes into different cell types. This process, which is conserved between plants and animals, is often achieved via dedifferentiation into pluripotent stem cells, which have the ability to generate all other types of cells and tissues of a given organism. Cellular reprogramming is thus a complex process that requires extensive modification at the epigenetic and transcriptional level, unlocking cellular programs that allow cells to acquire pluripotency. In addition to alterations in the gene expression profile, cellular reprogramming requires rearrangement of the proteome, organelles and metabolism, but these changes are comparatively less studied. In this context, autophagy, a cellular catabolic process that participates in the recycling of intracellular constituents, has the capacity to affect different aspects of cellular reprogramming, including the removal of protein signatures that might hamper reprogramming, mitophagy associated with metabolic reprogramming, and the supply of energy and metabolic building blocks to cells that undergo fate changes. In this Review, we discuss advances in our understanding of the role of autophagy during cellular reprogramming by drawing comparisons between plant and animal studies, as well as highlighting aspects of the topic that warrant further research.
Collapse
Affiliation(s)
- Morten Petersen
- Department of Biology, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Elise Ebstrup
- Department of Biology, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Eleazar Rodriguez
- Department of Biology, University of Copenhagen, 2200 Copenhagen N, Denmark
| |
Collapse
|
15
|
Rajagopalan K, Christyraj JDS, Chelladurai KS, Das P, Mahendran K, Nagarajan L, Gunalan S. Understanding the Multi-Functional Role of TCTP in the Regeneration Process of Earthworm, Perionyx excavatus. Tissue Eng Regen Med 2024; 21:353-366. [PMID: 37935935 PMCID: PMC10825100 DOI: 10.1007/s13770-023-00599-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 08/30/2023] [Accepted: 09/18/2023] [Indexed: 11/09/2023] Open
Abstract
BACKGROUND Regeneration is a highly complex process that requires the coordination of numerous molecular events, and identifying the key ruler that governs is important to investigate. While it has been shown that TCTP is a multi-functional protein that regulates cell proliferation, differentiation, apoptosis, anti-apoptosis, stem cell maintenance, and immune responses, but only a few studies associated to regeneration have been reported. To investigate the multi-functional role of TCTP in regeneration, the earthworm Perionyx excavatus was chosen. METHODS Through pharmacological suppression of TCTP, amputation, histology, molecular docking, and western blotting, the multi-function role of TCTP involved in regeneration is revealed. RESULTS Amputational studies show that P. excavatus is a clitellum-independent regenerating earthworm resulting in two functional worms upon amputation. Arresting cell cycle at the G1/S boundary using 2 mM Thymidine confirms that P. excavatus execute both epimorphosis and morphallaxis regeneration mode. The pharmacological suppression of TCTP using buclizine results in regeneration suppression. Following the combinatorial injection of 2 mM Thymidine and buclizine, the earthworm regeneration is completely blocked, which suggests a critical functional role of TCTP in morphallaxis. The pharmacological inhibition of TCTP also suppresses the key proteins involved in regeneration: Wnt3a (stem cell marker), PCNA (cell proliferation) and YAP1 (Hippo signalling) but augments the expression of cellular stress protein p53. CONCLUSION The collective results indicate that TCTP synchronously is involved in the process of stem cell activation, cell proliferation, morphallaxis, and organ development in the regeneration event.
Collapse
Affiliation(s)
- Kamarajan Rajagopalan
- Molecular Biology and Stem Cell Research Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science and Technology (Deemed to be University), Chennai, Tamilnadu, India
| | - Jackson Durairaj Selvan Christyraj
- Molecular Biology and Stem Cell Research Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science and Technology (Deemed to be University), Chennai, Tamilnadu, India.
| | - Karthikeyan Subbiahanadar Chelladurai
- Molecular Biology and Stem Cell Research Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science and Technology (Deemed to be University), Chennai, Tamilnadu, India
- School of Health Sciences, Purdue University, 550 Stadium Mall Drive, West Lafayette, IN, 47907, USA
| | - Puja Das
- Molecular Biology and Stem Cell Research Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science and Technology (Deemed to be University), Chennai, Tamilnadu, India
| | - Karthikeyan Mahendran
- Department of Zoology and Microbiology, Thiyagarajar College, Madurai, Tamilnadu, India
| | - Logeshwari Nagarajan
- Molecular Biology and Stem Cell Research Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science and Technology (Deemed to be University), Chennai, Tamilnadu, India
| | - Saritha Gunalan
- Molecular Biology and Stem Cell Research Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science and Technology (Deemed to be University), Chennai, Tamilnadu, India
| |
Collapse
|
16
|
Zeng J, Liu J, Ni H, Zhang L, Wang J, Li Y, Jiang W, Wu Z, Zhou M. Mitochondrial transplantation reduces lower limb ischemia-reperfusion injury by increasing skeletal muscle energy and adipocyte browning. Mol Ther Methods Clin Dev 2023; 31:101152. [PMID: 38027061 PMCID: PMC10667789 DOI: 10.1016/j.omtm.2023.101152] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 11/03/2023] [Indexed: 12/01/2023]
Abstract
Recent studies have shown that mitochondrial transplantation can repair lower limb IRI, but the underlying mechanism of the repair effect remains unclear. In this study, we found that in addition to being taken up by skeletal muscle cells, human umbilical cord mesenchymal stem cells (hMSCs)-derived mitochondria were also taken up by adipocytes, which was accompanied by an increase in optic atrophy 1 (OPA1) and uncoupling protein 1. Transplantation of hMSCs-derived mitochondria could not only supplement the original damaged mitochondrial function of skeletal muscle, but also promote adipocyte browning by increasing the expression of OPA1. In this process, mitochondrial transplantation can reduce cell apoptosis and repair muscle tissue, which promotes the recovery of motor function in vivo. To the best of our knowledge, there is no study on the therapeutic mechanism of mitochondrial transplantation from this perspective, which could provide a theoretical basis.
Collapse
Affiliation(s)
- Jiaqi Zeng
- Department of Vascular Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing 210046, China
- Department of Vascular Surgery, Kunshan Traditional Chinese Medicine Hospital, Kunshan 215300, China
| | - Jianing Liu
- Department of Vascular Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing 210046, China
| | - Haiya Ni
- Department of Vascular Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing 210046, China
| | - Ling Zhang
- Department of Vascular Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing 210046, China
| | - Jun Wang
- Department of Vascular Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing 210046, China
| | - Yazhou Li
- Department of Vascular Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing 210046, China
| | - Wentao Jiang
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Ziyu Wu
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Min Zhou
- Department of Vascular Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing 210046, China
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| |
Collapse
|
17
|
Seifert AW, Duncan EM, Zayas RM. Enduring questions in regenerative biology and the search for answers. Commun Biol 2023; 6:1139. [PMID: 37945686 PMCID: PMC10636051 DOI: 10.1038/s42003-023-05505-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 10/25/2023] [Indexed: 11/12/2023] Open
Abstract
The potential for basic research to uncover the inner workings of regenerative processes and produce meaningful medical therapies has inspired scientists, clinicians, and patients for hundreds of years. Decades of studies using a handful of highly regenerative model organisms have significantly advanced our knowledge of key cell types and molecular pathways involved in regeneration. However, many questions remain about how regenerative processes unfold in regeneration-competent species, how they are curtailed in non-regenerative organisms, and how they might be induced (or restored) in humans. Recent technological advances in genomics, molecular biology, computer science, bioengineering, and stem cell research hold promise to collectively provide new experimental evidence for how different organisms accomplish the process of regeneration. In theory, this new evidence should inform the design of new clinical approaches for regenerative medicine. A deeper understanding of how tissues and organs regenerate will also undoubtedly impact many adjacent scientific fields. To best apply and adapt these new technologies in ways that break long-standing barriers and answer critical questions about regeneration, we must combine the deep knowledge of developmental and evolutionary biologists with the hard-earned expertise of scientists in mechanistic and technical fields. To this end, this perspective is based on conversations from a workshop we organized at the Banbury Center, during which a diverse cross-section of the regeneration research community and experts in various technologies discussed enduring questions in regenerative biology. Here, we share the questions this group identified as significant and unanswered, i.e., known unknowns. We also describe the obstacles limiting our progress in answering these questions and how expanding the number and diversity of organisms used in regeneration research is essential for deepening our understanding of regenerative capacity. Finally, we propose that investigating these problems collaboratively across a diverse network of researchers has the potential to advance our field and produce unexpected insights into important questions in related areas of biology and medicine.
Collapse
Affiliation(s)
- Ashley W Seifert
- Department of Biology, University of Kentucky, Lexington, KY, 40506, USA.
| | - Elizabeth M Duncan
- Department of Biology, University of Kentucky, Lexington, KY, 40506, USA.
| | - Ricardo M Zayas
- Department of Biology, San Diego State University, San Diego, CA, 92182, USA.
| |
Collapse
|
18
|
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
|
19
|
Avila-Martinez N, Gansevoort M, Verbakel J, Jayaprakash H, Araujo IM, Vitorino M, Tiscornia G, van Kuppevelt TH, Daamen WF. Matrisomal components involved in regenerative wound healing in axolotl and Acomys: implications for biomaterial development. Biomater Sci 2023; 11:6060-6081. [PMID: 37525590 DOI: 10.1039/d3bm00835e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
Achieving regeneration in humans has been a long-standing goal of many researchers. Whereas amphibians like the axolotl (Ambystoma mexicanum) are capable of regenerating whole organs and even limbs, most mammals heal their wounds via fibrotic scarring. Recently, the African spiny mouse (Acomys sp.) has been shown to be injury resistant and capable of regenerating several tissue types. A major focal point of research with Acomys has been the identification of drivers of regeneration. In this search, the matrisome components related to the extracellular matrix (ECM) are often overlooked. In this review, we compare Acomys and axolotl skin wound healing and blastema-mediated regeneration by examining their wound healing responses and comparing the expression pattern of matrisome genes, including glycosaminoglycan (GAG) related genes. The goal of this review is to identify matrisome genes that are upregulated during regeneration and could be potential candidates for inclusion in pro-regenerative biomaterials. Research papers describing transcriptomic or proteomic coverage of either skin regeneration or blastema formation in Acomys and axolotl were selected. Matrisome and GAG related genes were extracted from each dataset and the resulting lists of genes were compared. In our analysis, we found several genes that were consistently upregulated, suggesting possible involvement in regenerative processes. Most of the components have been implicated in regulation of cell behavior, extracellular matrix remodeling and wound healing. Incorporation of such pro-regenerative factors into biomaterials may help to shift pro-fibrotic processes to regenerative responses in treated wounds.
Collapse
Affiliation(s)
- Nancy Avila-Martinez
- Department of Medical BioSciences, Radboud Research Institute, Radboud university medical center, PO Box 9101, 6500 HB Nijmegen, The Netherlands.
| | - Merel Gansevoort
- Department of Medical BioSciences, Radboud Research Institute, Radboud university medical center, PO Box 9101, 6500 HB Nijmegen, The Netherlands.
| | - Juul Verbakel
- Department of Medical BioSciences, Radboud Research Institute, Radboud university medical center, PO Box 9101, 6500 HB Nijmegen, The Netherlands.
| | - Haarshaadri Jayaprakash
- Centre of Marine Sciences (CCMAR), University of Algarve, 8005-139, Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve, 8005-139, Faro, Portugal
| | - Ines Maria Araujo
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve, 8005-139, Faro, Portugal
- Algarve Biomedical Center Research Institute (ABC-RI), University of Algarve, 8005-139, Faro, Portugal
| | - Marta Vitorino
- Centre of Marine Sciences (CCMAR), University of Algarve, 8005-139, Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve, 8005-139, Faro, Portugal
- Algarve Biomedical Center Research Institute (ABC-RI), University of Algarve, 8005-139, Faro, Portugal
| | - Gustavo Tiscornia
- Centre of Marine Sciences (CCMAR), University of Algarve, 8005-139, Faro, Portugal
- Eugin Barcelona, Balmes, 236, 08006 Barcelona, Spain
| | - Toin H van Kuppevelt
- Department of Medical BioSciences, Radboud Research Institute, Radboud university medical center, PO Box 9101, 6500 HB Nijmegen, The Netherlands.
| | - Willeke F Daamen
- Department of Medical BioSciences, Radboud Research Institute, Radboud university medical center, PO Box 9101, 6500 HB Nijmegen, The Netherlands.
| |
Collapse
|
20
|
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
|
21
|
Aitken KJ, Yadav P, Sidler M, Thanabalasingam T, Ahmed T, Aggarwal P, Yip ST, Jeffrey N, Jiang JX, Siebenaller A, Sotiropoulos C, Huang R, Le DMQ, Delgado-Olguin P, Bagli D. Spontaneous urinary bladder regeneration after subtotal cystectomy increases YAP/WWTR1 signaling and downstream BDNF expression: Implications for smooth muscle injury responses. PLoS One 2023; 18:e0287205. [PMID: 37494380 PMCID: PMC10370683 DOI: 10.1371/journal.pone.0287205] [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: 12/30/2022] [Accepted: 06/01/2023] [Indexed: 07/28/2023] Open
Abstract
Rodents have the capacity for spontaneous bladder regeneration and bladder smooth muscle cell (BSMC) migration following a subtotal cystectomy (STC). YAP/WWTR1 and BDNF (Brain-derived neurotrophic factor) play crucial roles in development and regeneration. During partial bladder outlet obstruction (PBO), excessive YAP/WWTR1 signaling and BDNF expression increases BSMC hypertrophy and dysfunction. YAP/WWTR1 and expression of BDNF and CYR61 were examined in models of regeneration and wound repair. Live cell microscopy was utilized in an ex vivo model of STC to visualize cell movement and division. In Sprague-Dawley female rats, STC was performed by resection of the bladder dome sparing the trigone, followed by closure of the bladder. Smooth muscle migration and downstream effects on signaling and expression were also examined after scratch wound of BSMC with inhibitors of YAP and BDNF signaling. Sham, PBO and incision (cystotomy) were comparators for the STC model. Scratch wound in vitro increased SMC migration and expression of BDNF, CTGF and CYR61 in a YAP/WWTR1-dependent manner. Inhibition of YAP/WWTR1 and BDNF signaling reduced scratch-induced migration. BDNF and CYR61 expression was elevated during STC and PBO. STC induces discrete genes associated with endogenous de novo cell regeneration downstream of YAP/WWTR1 activation.
Collapse
Affiliation(s)
- Karen J. Aitken
- Developmental and Stem Cell Biology, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
- Institute of Medical Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Priyank Yadav
- Developmental and Stem Cell Biology, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Urology and Renal Transplantation, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
- Urology Division, Department of Surgery, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Martin Sidler
- Developmental and Stem Cell Biology, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
- Institute of Medical Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Urology Division, Department of Surgery, Hospital for Sick Children, Toronto, Ontario, Canada
- Division Chief, Paediatric and Neonatal Surgeon, University Hospital Ulm, Ulm, Baden-Württemberg, Germany
| | - Thenuka Thanabalasingam
- Developmental and Stem Cell Biology, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
- Human Biology Programme, Faculty of Arts and Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Tabina Ahmed
- Developmental and Stem Cell Biology, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
- Human Biology Programme, Faculty of Arts and Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Prateek Aggarwal
- Developmental and Stem Cell Biology, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
- Institute of Medical Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Human Biology Programme, Faculty of Arts and Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Shing Tai Yip
- Developmental and Stem Cell Biology, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Nefateri Jeffrey
- Developmental and Stem Cell Biology, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Jia-Xin Jiang
- Developmental and Stem Cell Biology, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Aliza Siebenaller
- Developmental and Stem Cell Biology, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Chris Sotiropoulos
- Developmental and Stem Cell Biology, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
- Human Biology Programme, Faculty of Arts and Sciences, University of Toronto, Toronto, Ontario, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Ryan Huang
- Human Biology Programme, Faculty of Arts and Sciences, University of Toronto, Toronto, Ontario, Canada
| | - David Minh Quynh Le
- Human Biology Programme, Faculty of Arts and Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Paul Delgado-Olguin
- Translational Medicine Program, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Molecular Genetics, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Darius Bagli
- Developmental and Stem Cell Biology, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
- Institute of Medical Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Urology Division, Department of Surgery, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
22
|
Pio-Lopez L, Bischof J, LaPalme JV, Levin M. The scaling of goals from cellular to anatomical homeostasis: an evolutionary simulation, experiment and analysis. Interface Focus 2023; 13:20220072. [PMID: 37065270 PMCID: PMC10102734 DOI: 10.1098/rsfs.2022.0072] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 03/02/2023] [Indexed: 04/18/2023] Open
Abstract
Complex living agents consist of cells, which are themselves competent sub-agents navigating physiological and metabolic spaces. Behaviour science, evolutionary developmental biology and the field of machine intelligence all seek to understand the scaling of biological cognition: what enables individual cells to integrate their activities to result in the emergence of a novel, higher-level intelligence with large-scale goals and competencies that belong to it and not to its parts? Here, we report the results of simulations based on the TAME framework, which proposes that evolution pivoted the collective intelligence of cells during morphogenesis of the body into traditional behavioural intelligence by scaling up homeostatic competencies of cells in metabolic space. In this article, we created a minimal in silico system (two-dimensional neural cellular automata) and tested the hypothesis that evolutionary dynamics are sufficient for low-level setpoints of metabolic homeostasis in individual cells to scale up to tissue-level emergent behaviour. Our system showed the evolution of the much more complex setpoints of cell collectives (tissues) that solve a problem in morphospace: the organization of a body-wide positional information axis (the classic French flag problem in developmental biology). We found that these emergent morphogenetic agents exhibit a number of predicted features, including the use of stress propagation dynamics to achieve the target morphology as well as the ability to recover from perturbation (robustness) and long-term stability (even though neither of these was directly selected for). Moreover, we observed an unexpected behaviour of sudden remodelling long after the system stabilizes. We tested this prediction in a biological system-regenerating planaria-and observed a very similar phenomenon. We propose that this system is a first step towards a quantitative understanding of how evolution scales minimal goal-directed behaviour (homeostatic loops) into higher-level problem-solving agents in morphogenetic and other spaces.
Collapse
Affiliation(s)
- Léo Pio-Lopez
- Allen Discovery Center, Tufts University, Medford, MA, USA
| | | | | | - Michael Levin
- Allen Discovery Center, Tufts University, Medford, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| |
Collapse
|
23
|
Tomasso A, Koopmans T, Lijnzaad P, Bartscherer K, Seifert AW. An ERK-dependent molecular switch antagonizes fibrosis and promotes regeneration in spiny mice ( Acomys). SCIENCE ADVANCES 2023; 9:eadf2331. [PMID: 37126559 PMCID: PMC10132760 DOI: 10.1126/sciadv.adf2331] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Although most mammals heal injured tissues and organs with scarring, spiny mice (Acomys) naturally regenerate skin and complex musculoskeletal tissues. Now, the core signaling pathways driving mammalian tissue regeneration are poorly characterized. Here, we show that, while immediate extracellular signal-regulated kinase (ERK) activation is a shared feature of scarring (Mus) and regenerating (Acomys) injuries, ERK activity is only sustained at high levels during complex tissue regeneration. Following ERK inhibition, ear punch regeneration in Acomys shifted toward fibrotic repair. Using single-cell RNA sequencing, we identified ERK-responsive cell types. Loss- and gain-of-function experiments prompted us to uncover fibroblast growth factor and ErbB signaling as upstream ERK regulators of regeneration. The ectopic activation of ERK in scar-prone injuries induced a pro-regenerative response, including cell proliferation, extracellular matrix remodeling, and hair follicle neogenesis. Our data detail an important distinction in ERK activity between regenerating and poorly regenerating adult mammals and open avenues to redirect fibrotic repair toward regenerative healing.
Collapse
Affiliation(s)
- Antonio Tomasso
- Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, Münster 48149, Germany
- Cells in Motion Cluster of Excellence-International Max Planck Research School (CiM-IMPRS Graduate Program), Münster 48149, Germany
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), Uppsalalaan 8, Utrecht 3584CT, Netherlands
- Department of Biology/Chemistry, Osnabrück University, Barbarastrasse 11, Osnabrück 49076, Germany
- Department of Biology, University of Kentucky, 101 T.H. Morgan Building, Lexington, KY 40506, USA
| | - Tim Koopmans
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), Uppsalalaan 8, Utrecht 3584CT, Netherlands
- Department of Biology/Chemistry, Osnabrück University, Barbarastrasse 11, Osnabrück 49076, Germany
| | - Philip Lijnzaad
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, Netherlands
| | - Kerstin Bartscherer
- Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, Münster 48149, Germany
- Cells in Motion Cluster of Excellence-International Max Planck Research School (CiM-IMPRS Graduate Program), Münster 48149, Germany
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), Uppsalalaan 8, Utrecht 3584CT, Netherlands
- Department of Biology/Chemistry, Osnabrück University, Barbarastrasse 11, Osnabrück 49076, Germany
| | - Ashley W Seifert
- Department of Biology, University of Kentucky, 101 T.H. Morgan Building, Lexington, KY 40506, USA
| |
Collapse
|
24
|
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
|
25
|
Comparison of the Effect of Different Conditioning Media on the Angiogenic Potential of Hypoxia Preconditioned Blood-Derived Secretomes: Towards Engineering Next-Generation Autologous Growth Factor Cocktails. Int J Mol Sci 2023; 24:ijms24065485. [PMID: 36982558 PMCID: PMC10049474 DOI: 10.3390/ijms24065485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 03/06/2023] [Accepted: 03/09/2023] [Indexed: 03/17/2023] Open
Abstract
Hypoxia Preconditioned Plasma (HPP) and Serum (HPS) are regenerative blood-derived growth factor compositions that have been extensively examined for their angiogenic and lymphangiogenic activity towards wound healing and tissue repair. Optimization of these secretomes’ growth factor profile, through adjustments of the conditioning parameters, is a key step towards clinical application. In this study, the autologous liquid components (plasma/serum) of HPP and HPS were replaced with various conditioning media (NaCl, PBS, Glucose 5%, AIM V medium) and were analyzed in terms of key pro- (VEGF-A, EGF) and anti-angiogenic (TSP-1, PF-4) protein factors, as well as their ability to promote microvessel formation in vitro. We found that media substitution resulted in changes in the concentration of the aforementioned growth factors, and also influenced their ability to induce angiogenesis. While NaCl and PBS led to a lower concentration of all growth factors examined, and consequently an inferior tube formation response, replacement with Glucose 5% resulted in increased growth factor concentrations in anticoagulated blood-derived secretomes, likely due to stimulation of platelet factor release. Medium substitution with Glucose 5% and specialized peripheral blood cell-culture AIM V medium generated comparable tube formation to HPP and HPS controls. Altogether, our data suggest that medium replacement of plasma and serum may significantly influence the growth factor profile of hypoxia-preconditioned blood-derived secretomes and, therefore, their potential application as tools for promoting therapeutic angiogenesis.
Collapse
|
26
|
Tursch A, Holstein TW. From injury to patterning—MAPKs and Wnt signaling in Hydra. Curr Top Dev Biol 2023; 153:381-417. [PMID: 36967201 DOI: 10.1016/bs.ctdb.2023.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Hydra has a regenerative capacity that is not limited to individual organs but encompasses the entire body. Various global and integrative genome, transcriptome and proteome approaches have shown that many of the signaling pathways and transcription factors present in vertebrates are already present in Cnidaria, the sister group of Bilateria, and are also activated in regeneration. It is now possible to investigate one of the central questions of regeneration biology, i.e., how does the patterning system become activated by the injury signals that initiate regeneration. This review will present the current data obtained in Hydra and draw parallels with regeneration in Bilateria. Important findings of this global analysis are that the Wnt signaling pathway has a dual function in the regeneration process. In the early phase Wnt is activated generically and in a second phase of pattern formation it is activated in a position specific manner. Thus, Wnt signaling is part of the generic injury response, in which mitogen-activated protein kinases (MAPKs) are initially activated via calcium and reactive oxygen species (ROS). The MAPKs, p38, c-Jun N-terminal kinases (JNKs) and extracellular signal-regulated kinases (ERK) are essential for Wnt activation in Hydra head and foot regenerates. Furthermore, the antagonism between the ERK signaling pathway and stress-induced MAPKs results in a balanced induction of apoptosis and mitosis. However, the early Wnt genes are activated by MAPK signaling rather than apoptosis. Early Wnt gene activity is differentially integrated with a stable, β-Catenin-based gradient along the primary body axis maintaining axial polarity and activating further Wnts in the regenerating head. Because MAPKs and Wnts are highly evolutionarily conserved, we hypothesize that this mechanism is also present in vertebrates but may be activated to different degrees at the level of early Wnt gene integration.
Collapse
|
27
|
Injury-induced MAPK activation triggers body axis formation in Hydra by default Wnt signaling. Proc Natl Acad Sci U S A 2022; 119:e2204122119. [PMID: 35994642 PMCID: PMC9436372 DOI: 10.1073/pnas.2204122119] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Hydra's almost unlimited regenerative potential is based on Wnt signaling, but so far it is unknown how the injury stimulus is transmitted to discrete patterning fates in head and foot regenerates. We previously identified mitogen-activated protein kinases (MAPKs) among the earliest injury response molecules in Hydra head regeneration. Here, we show that three MAPKs-p38, c-Jun N-terminal kinases (JNKs), and extracellular signal-regulated kinases (ERKs)-are essential to initiate regeneration in Hydra, independent of the wound position. Their activation occurs in response to any injury and requires calcium and reactive oxygen species (ROS) signaling. Phosphorylated MAPKs hereby exhibit cross talk with mutual antagonism between the ERK pathway and stress-induced MAPKs, orchestrating a balance between cell survival and apoptosis. Importantly, Wnt3 and Wnt9/10c, which are induced by MAPK signaling, can partially rescue regeneration in tissues treated with MAPK inhibitors. Also, foot regenerates can be reverted to form head tissue by a pharmacological increase of β-catenin signaling or the application of recombinant Wnts. We propose a model in which a β-catenin-based stable gradient of head-forming capacity along the primary body axis, by differentially integrating an indiscriminate injury response, determines the fate of the regenerating tissue. Hereby, Wnt signaling acquires sustained activation in the head regenerate, while it is transient in the presumptive foot tissue. Given the high level of evolutionary conservation of MAPKs and Wnts, we assume that this mechanism is deeply embedded in our genome.
Collapse
|
28
|
Rajagopalan K, Christyraj JDS, Chelladurai KS, Gnanaraja JKJS, Christyraj JRSS. Comparative analysis of the survival and regeneration potential of juvenile and matured earthworm, Eudrilus eugeniae, upon in vivo and in vitro maintenance. In Vitro Cell Dev Biol Anim 2022; 58:587-598. [PMID: 35920958 DOI: 10.1007/s11626-022-00706-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 07/08/2022] [Indexed: 11/29/2022]
Abstract
Eudrilus eugeniae is a clitellum-dependent earthworm that requires intact clitellum segments for its survival and regeneration. The present study aims to interconnect the survival and regeneration ability that varies between in vivo and in vitro maintenance upon different sites of amputation. The amputated portion of the worm that possesses intact clitellum (13th-18th segments) survived and had the potential to regenerate, whereas worms with partial or without clitellum segments only survived and were unable to regenerate. Besides segment length and clitellum segments, clitellum factors also determined the survival, blastemal initiation and differentiation potential. The survivability and regeneration potential of worms were augmented upon in vitro maintenance. Notably, the amputated segments (1st-10th segments) and posterior segments of similar length, which usually die within the 4th day in vivo, survived for more than 60 days in vitro but lacked the regeneration ability. On the other hand, the amputated posterior segments (30th to 37th segments) from juvenile worms, maintained in in vitro condition, survived and initiated blastema with multiple buds but lacked the ability to regenerate. Interestingly, the equal half of adult worm blastema that is maintained in in vitro conditions were able to form the blastema-like structure with the help of a unique stick. The anterior blastema failed to retain the regenerative structure but the posterior portion of the amputated blastema, which is also associated with a small portion of the body segment, showed the ability to retain the regenerative structure. Our results conclude that the survivability is enhanced upon in vitro maintenance and this condition favours the adult dedifferentiated blastemal and stem cell-enriched juvenile posterior segments to form a regenerative blastema.
Collapse
Affiliation(s)
- Kamarajan Rajagopalan
- Regeneration and Stem Cell Biology Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science and Technology (Deemed to Be University), Chennai, Tamil Nadu, 621306, India
| | - Jackson Durairaj Selvan Christyraj
- Regeneration and Stem Cell Biology Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science and Technology (Deemed to Be University), Chennai, Tamil Nadu, 621306, India.
| | - Karthikeyan Subbiahanadar Chelladurai
- Regeneration and Stem Cell Biology Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science and Technology (Deemed to Be University), Chennai, Tamil Nadu, 621306, India.,School of Health Sciences, Purdue University, West Lafayette, USA
| | | | - Johnson Retnaraj Samuel Selvan Christyraj
- Regeneration and Stem Cell Biology Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science and Technology (Deemed to Be University), Chennai, Tamil Nadu, 621306, India.
| |
Collapse
|
29
|
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
|
30
|
Sehring I, Weidinger G. Zebrafish Fin: Complex Molecular Interactions and Cellular Mechanisms Guiding Regeneration. Cold Spring Harb Perspect Biol 2022; 14:a040758. [PMID: 34649924 PMCID: PMC9248819 DOI: 10.1101/cshperspect.a040758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The zebrafish caudal fin has become a popular model to study cellular and molecular mechanisms of regeneration due to its high regenerative capacity, accessibility for experimental manipulations, and relatively simple anatomy. The formation of a regenerative epidermis and blastema are crucial initial events and tightly regulated. Both the regenerative epidermis and the blastema are highly organized structures containing distinct domains, and several signaling pathways regulate the formation and interaction of these domains. Bone is the major tissue regenerated from the progenitor cells of the blastema. Several cellular mechanisms can provide source cells for blastemal (pre-)osteoblasts, including dedifferentiation of differentiated osteoblasts and de novo formation from other cell types, providing intriguing examples of cellular plasticity. In recent years, omics analyses and single-cell approaches have elucidated genetic and epigenetic regulation, increasing our knowledge of the surprisingly complex coordination of various mechanisms to achieve successful restoration of a seemingly simple structure.
Collapse
Affiliation(s)
- Ivonne Sehring
- Institute of Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Gilbert Weidinger
- Institute of Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| |
Collapse
|
31
|
Sehring IM, Mohammadi HF, Haffner-Luntzer M, Ignatius A, Huber-Lang M, Weidinger G. Zebrafish fin regeneration involves generic and regeneration-specific osteoblast injury responses. eLife 2022; 11:77614. [PMID: 35748539 PMCID: PMC9259016 DOI: 10.7554/elife.77614] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 06/23/2022] [Indexed: 11/13/2022] Open
Abstract
Successful regeneration requires the coordinated execution of multiple cellular responses to injury. In amputated zebrafish fins, mature osteoblasts dedifferentiate, migrate towards the injury and form proliferative osteogenic blastema cells. We show that osteoblast migration is preceded by cell elongation and alignment along the proximodistal axis, which require actomyosin, but not microtubule turnover. Surprisingly, osteoblast dedifferentiation and migration can be uncoupled. Using pharmacological and genetic interventions, we found that NF-ĸB and retinoic acid signalling regulate dedifferentiation without affecting migration, while the complement system and actomyosin dynamics affect migration but not dedifferentiation. Furthermore, by removing bone at two locations within a fin ray, we established an injury model containing two injury sites. We found that osteoblasts dedifferentiate at and migrate towards both sites, while accumulation of osteogenic progenitor cells and regenerative bone formation only occur at the distal-facing injury. Together, these data indicate that osteoblast dedifferentiation and migration represent generic injury responses that are differentially regulated and can occur independently of each other and of regenerative growth. We conclude that successful fin bone regeneration appears to involve the coordinated execution of generic and regeneration-specific responses of osteoblasts to injury.
Collapse
Affiliation(s)
| | | | | | - Anita Ignatius
- Institute of Orthopaedic Research and Biomechanics, University Hospital Ulm, Ulm, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology (ITI), University Hospital Ulm, Ulm, Germany
| | - Gilbert Weidinger
- Institute of Biochemistry and Molecular Biology, University of Ulm, Ulm, Germany
| |
Collapse
|
32
|
Scimone ML, Cloutier JK, Maybrun CL, Reddien PW. The planarian wound epidermis gene equinox is required for blastema formation in regeneration. Nat Commun 2022; 13:2726. [PMID: 35585061 PMCID: PMC9117669 DOI: 10.1038/s41467-022-30412-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 04/09/2022] [Indexed: 11/25/2022] Open
Abstract
Regeneration often involves the formation of a blastema, an outgrowth or regenerative bud formed at the plane of injury where missing tissues are produced. The mechanisms that trigger blastema formation are therefore fundamental for regeneration. Here, we identify a gene, which we named equinox, that is expressed within hours of injury in the planarian wound epidermis. equinox encodes a predicted secreted protein that is conserved in many animal phyla. Following equinox inhibition, amputated planarians fail to maintain wound-induced gene expression and to subsequently undergo blastema outgrowth. Associated with these defects is an inability to reestablish lost positional information needed for missing tissue specification. Our findings link the planarian wound epidermis, through equinox, to regeneration of positional information and blastema formation, indicating a broad regulatory role of the wound epidermis in diverse regenerative contexts.
Collapse
Affiliation(s)
- M Lucila Scimone
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Whitehead Institute for Biomedical Research, Cambridge, MA, 02142, USA
| | - Jennifer K Cloutier
- Whitehead Institute for Biomedical Research, Cambridge, MA, 02142, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Harvard/MIT MD-PhD, Harvard Medical School, Boston, MA, 02115, USA
| | - Chloe L Maybrun
- Whitehead Institute for Biomedical Research, Cambridge, MA, 02142, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Peter W Reddien
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
- Whitehead Institute for Biomedical Research, Cambridge, MA, 02142, USA.
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
| |
Collapse
|
33
|
Reddien PW. Positional Information and Stem Cells Combine to Result in Planarian Regeneration. Cold Spring Harb Perspect Biol 2022; 14:a040717. [PMID: 34518341 PMCID: PMC9121904 DOI: 10.1101/cshperspect.a040717] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The capacity for regeneration is broad in the animal kingdom. Planarians are flatworms that can regenerate any missing body part and their regenerative powers have combined with ease of experimentation to make them a classic regeneration model for more than a century. Pluripotent stem cells called neoblasts generate missing planarian tissues. Fate specification happens in the neoblasts, and this can occur in response to regeneration instructions in the form of positional information. Fate specification can lead to differentiating cells in single steps rather than requiring a long lineage hierarchy. Planarians display constitutive expression of positional information from muscle cells, which is required for patterned maintenance of tissues in tissue turnover. Amputation leads to the rapid resetting of positional information in a process triggered by wound signaling and the resetting of positional information is required for regeneration. These findings suggest a model for planarian regeneration in which adult positional information resets after injury to regulate stem cells to bring about the replacement of missing parts.
Collapse
Affiliation(s)
- Peter W Reddien
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts 02142, USA
- Department of Biology, MIT, Cambridge, Massachusetts 02139, USA
- Department of Biology, MIT, Cambridge, Massachusetts 02139, USA
| |
Collapse
|
34
|
Avalos PN, Forsthoefel DJ. An Emerging Frontier in Intercellular Communication: Extracellular Vesicles in Regeneration. Front Cell Dev Biol 2022; 10:849905. [PMID: 35646926 PMCID: PMC9130466 DOI: 10.3389/fcell.2022.849905] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 03/28/2022] [Indexed: 12/12/2022] Open
Abstract
Regeneration requires cellular proliferation, differentiation, and other processes that are regulated by secreted cues originating from cells in the local environment. Recent studies suggest that signaling by extracellular vesicles (EVs), another mode of paracrine communication, may also play a significant role in coordinating cellular behaviors during regeneration. EVs are nanoparticles composed of a lipid bilayer enclosing proteins, nucleic acids, lipids, and other metabolites, and are secreted by most cell types. Upon EV uptake by target cells, EV cargo can influence diverse cellular behaviors during regeneration, including cell survival, immune responses, extracellular matrix remodeling, proliferation, migration, and differentiation. In this review, we briefly introduce the history of EV research and EV biogenesis. Then, we review current understanding of how EVs regulate cellular behaviors during regeneration derived from numerous studies of stem cell-derived EVs in mammalian injury models. Finally, we discuss the potential of other established and emerging research organisms to expand our mechanistic knowledge of basic EV biology, how injury modulates EV biogenesis, cellular sources of EVs in vivo, and the roles of EVs in organisms with greater regenerative capacity.
Collapse
Affiliation(s)
- Priscilla N. Avalos
- Department of Cell Biology, College of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - David J. Forsthoefel
- Department of Cell Biology, College of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| |
Collapse
|
35
|
Sarkar SR, Dubey VK, Jahagirdar A, Lakshmanan V, Haroon MM, Sowndarya S, Sowdhamini R, Palakodeti D. DDX24 is required for muscle fiber organization and the suppression of wound-induced Wnt activity necessary for pole re-establishment during planarian regeneration. Dev Biol 2022; 488:11-29. [PMID: 35523320 DOI: 10.1016/j.ydbio.2022.04.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 04/06/2022] [Accepted: 04/28/2022] [Indexed: 12/14/2022]
Abstract
Planarians have a remarkable ability to undergo whole-body regeneration. Successful regeneration outcome is determined by processes like polarity establishment at the wound site, which is followed by pole (organizer) specification. Interestingly, these determinants are almost exclusively expressed by muscles in these animals. However, the molecular toolkit that enables the functional versatility of planarian muscles remains poorly understood. Here we report that SMED_DDX24, a D-E-A-D Box RNA helicase, is necessary for planarian survival and regeneration. We found that DDX24 is enriched in muscles and its knockdown disrupts muscle fiber organization. This leads to defective pole specification, which in turn results in misregulation of many positional control genes specifically during regeneration. ddx24 RNAi also upregulates wound-induced Wnt signalling. Suppressing this ectopic Wnt activity rescues the knockdown phenotype by enabling better anterior pole regeneration. To summarize, our work highlights the role of an RNA helicase in muscle fiber organization, and modulating amputation-induced wnt levels, both of which seem critical for pole re-organization, thereby regulating whole-body regeneration.
Collapse
Affiliation(s)
- Souradeep R Sarkar
- National Centre for Biological Sciences (NCBS), Tata Institute of Fundamental Research (TIFR), Bengaluru, 560065, India; Integrative Chemical Biology (ICB), Institute for Stem Cell Science and Regenerative Medicine (inStem), Bengaluru, 560065, India
| | - Vinay Kumar Dubey
- Integrative Chemical Biology (ICB), Institute for Stem Cell Science and Regenerative Medicine (inStem), Bengaluru, 560065, India; Manipal Academy of Higher Education, Manipal, 576104, India
| | - Anusha Jahagirdar
- Integrative Chemical Biology (ICB), Institute for Stem Cell Science and Regenerative Medicine (inStem), Bengaluru, 560065, India
| | - Vairavan Lakshmanan
- Integrative Chemical Biology (ICB), Institute for Stem Cell Science and Regenerative Medicine (inStem), Bengaluru, 560065, India
| | - Mohamed Mohamed Haroon
- Integrative Chemical Biology (ICB), Institute for Stem Cell Science and Regenerative Medicine (inStem), Bengaluru, 560065, India; SASTRA University, Thanjavur, 613401, India
| | - Sai Sowndarya
- Integrative Chemical Biology (ICB), Institute for Stem Cell Science and Regenerative Medicine (inStem), Bengaluru, 560065, India
| | - Ramanathan Sowdhamini
- National Centre for Biological Sciences (NCBS), Tata Institute of Fundamental Research (TIFR), Bengaluru, 560065, India
| | - Dasaradhi Palakodeti
- Integrative Chemical Biology (ICB), Institute for Stem Cell Science and Regenerative Medicine (inStem), Bengaluru, 560065, India.
| |
Collapse
|
36
|
Wu W, Liu S, Wu H, Chen M, Gao L, Zhao B, Liu B, Pang Q. DjPtpn11 is an essential modulator of planarian (Dugesia japonica) regeneration. Int J Biol Macromol 2022; 209:1054-1064. [PMID: 35452697 DOI: 10.1016/j.ijbiomac.2022.04.095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 04/11/2022] [Accepted: 04/13/2022] [Indexed: 11/19/2022]
Abstract
Freshwater planarian Dugesia japonica is an excellent model organism for investigating stem cell behavior during regeneration. Despite studies showing that numerous genetic factors are involved in regeneration, much more research is required to fully understand the molecular mechanisms that orchestrate regeneration. In this study, we identified an evolutionarily conserved gene DjPtpn11(DjShp2). DjPtpn11 transcripts are expressed in neoblasts and some differentiated cells, with a high expression at the newly formed blastema. Its silencing by RNA interference (RNAi) affected anterior regeneration and inhibited the regeneration of posterior regions, including cholinergic and serotonergic neuron regeneration. In adult planarians, DjPtpn11 knockdown did not affect neoblast survival and proliferation but might prevent the stem cell migration and differentiation through ERK signaling. DjPtpn11 was demonstrated to be necessary for the anterior blastema cell differentiation partially via regulating ERK-DjMkpA activity. DjPtpn11 also influenced posterior specification via DjIslet, suggesting that DjPtpn11 may be involved in regulating the Wnt signaling pathway during the development of posterior blastema. Together, these data identified that DjPtpn11 is an essential modulator for the regeneration of planarians, and it may influence the appropriate differentiation of blastema cells.
Collapse
Affiliation(s)
- Weiwei Wu
- Anti-aging & Regenerative Medicine Research Institution, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China
| | - Shuo Liu
- Anti-aging & Regenerative Medicine Research Institution, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China
| | - Hao Wu
- Anti-aging & Regenerative Medicine Research Institution, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China
| | - Meishan Chen
- Anti-aging & Regenerative Medicine Research Institution, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China
| | - Lili Gao
- Anti-aging & Regenerative Medicine Research Institution, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China
| | - Bosheng Zhao
- Laboratory of Developmental and Evolutionary Biology, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China
| | - Baohua Liu
- Anti-aging & Regenerative Medicine Research Institution, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China.; Shenzhen University of Health Science Center, Shenzhen 518060, China
| | - Qiuxiang Pang
- Anti-aging & Regenerative Medicine Research Institution, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China.; Laboratory of Developmental and Evolutionary Biology, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China.
| |
Collapse
|
37
|
Coronel-Córdoba P, Molina MD, Cardona G, Fraguas S, Pascual-Carreras E, Saló E, Cebrià F, Adell T. FoxK1 is Required for Ectodermal Cell Differentiation During Planarian Regeneration. Front Cell Dev Biol 2022; 10:808045. [PMID: 35273960 PMCID: PMC8901602 DOI: 10.3389/fcell.2022.808045] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 01/25/2022] [Indexed: 11/24/2022] Open
Abstract
Forkhead box (Fox) genes belong to the “winged helix” transcription factor superfamily. The function of some Fox genes is well known, such as the role of foxO in controlling metabolism and longevity and foxA in controlling differentiation of endodermal tissues. However, the role of some Fox factors is not yet well characterized. Such is the case of FoxK genes, which are mainly studied in mammals and have been implicated in diverse processes including cell proliferation, tissue differentiation and carcinogenesis. Planarians are free-living flatworms, whose importance in biomedical research lies in their regeneration capacity. Planarians possess a wide population of pluripotent adult stem cells, called neoblasts, which allow them to regenerate any body part after injury. In a recent study, we identified three foxK paralogs in the genome of Schmidtea mediterranea. In this study, we demonstrate that foxK1 inhibition prevents regeneration of the ectodermal tissues, including the nervous system and the epidermis. These results correlate with foxK1 expression in neoblasts and in neural progenitors. Although the triggering of wound genes expression, polarity reestablishment and proliferation was not affected after foxK1 silencing, the apoptotic response was decreased. Altogether, these results suggest that foxK1 would be required for differentiation and maintenance of ectodermal tissues.
Collapse
Affiliation(s)
- Pablo Coronel-Córdoba
- 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
| | - 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
| | - Gemma Cardona
- 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
| | - Eudald Pascual-Carreras
- 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
| | - Emili Saló
- 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
| | - Teresa Adell
- 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
|
38
|
Leigh ND, Currie JD. Re-building limbs, one cell at a time. Dev Dyn 2022; 251:1389-1403. [PMID: 35170828 PMCID: PMC9545806 DOI: 10.1002/dvdy.463] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 01/23/2022] [Accepted: 01/25/2022] [Indexed: 11/24/2022] Open
Abstract
New techniques for visualizing and interrogating single cells hold the key to unlocking the underlying mechanisms of salamander limb regeneration.
Collapse
Affiliation(s)
- Nicholas D Leigh
- Molecular Medicine and Gene Therapy, Wallenberg Centre for Molecular Medicine, Lund Stem Cell Center, Lund University, Sweden
| | - Joshua D Currie
- Department of Biology, Wake Forest University, 455 Vine Street, Winston-Salem, USA
| |
Collapse
|
39
|
Wen X, Jiao L, Tan H. MAPK/ERK Pathway as a Central Regulator in Vertebrate Organ Regeneration. Int J Mol Sci 2022; 23:ijms23031464. [PMID: 35163418 PMCID: PMC8835994 DOI: 10.3390/ijms23031464] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/24/2022] [Accepted: 01/25/2022] [Indexed: 02/06/2023] Open
Abstract
Damage to organs by trauma, infection, diseases, congenital defects, aging, and other injuries causes organ malfunction and is life-threatening under serious conditions. Some of the lower order vertebrates such as zebrafish, salamanders, and chicks possess superior organ regenerative capacity over mammals. The extracellular signal-regulated kinases 1 and 2 (ERK1/2), as key members of the mitogen-activated protein kinase (MAPK) family, are serine/threonine protein kinases that are phylogenetically conserved among vertebrate taxa. MAPK/ERK signaling is an irreplaceable player participating in diverse biological activities through phosphorylating a broad variety of substrates in the cytoplasm as well as inside the nucleus. Current evidence supports a central role of the MAPK/ERK pathway during organ regeneration processes. MAPK/ERK signaling is rapidly excited in response to injury stimuli and coordinates essential pro-regenerative cellular events including cell survival, cell fate turnover, migration, proliferation, growth, and transcriptional and translational activities. In this literature review, we recapitulated the multifaceted MAPK/ERK signaling regulations, its dynamic spatio-temporal activities, and the profound roles during multiple organ regeneration, including appendages, heart, liver, eye, and peripheral/central nervous system, illuminating the possibility of MAPK/ERK signaling as a critical mechanism underlying the vastly differential regenerative capacities among vertebrate species, as well as its potential applications in tissue engineering and regenerative medicine.
Collapse
|
40
|
Wu YC, Franzenburg S, Ribes M, Pita L. Wounding response in Porifera (sponges) activates ancestral signaling cascades involved in animal healing, regeneration, and cancer. Sci Rep 2022; 12:1307. [PMID: 35079031 PMCID: PMC8789774 DOI: 10.1038/s41598-022-05230-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 01/06/2022] [Indexed: 11/24/2022] Open
Abstract
Upon injury, the homeostatic balance that ensures tissue function is disrupted. Wound-induced signaling triggers the recovery of tissue integrity and offers a context to understand the molecular mechanisms for restoring tissue homeostasis upon disturbances. Marine sessile animals are particularly vulnerable to chronic wounds caused by grazers that can compromise prey's health. Yet, in comparison to other stressors like warming or acidification, we know little on how marine animals respond to grazing. Marine sponges (Phylum Porifera) are among the earliest-diverging animals and play key roles in the ecosystem; but they remain largely understudied. Here, we investigated the transcriptomic responses to injury caused by a specialist spongivorous opisthobranch (i.e., grazing treatment) or by clipping with a scalpel (i.e., mechanical damage treatment), in comparison to control sponges. We collected samples 3 h, 1 d, and 6 d post-treatment for differential gene expression analysis on RNA-seq data. Both grazing and mechanical damage activated a similar transcriptomic response, including a clotting-like cascade (e.g., with genes annotated as transglutaminases, metalloproteases, and integrins), calcium signaling, and Wnt and mitogen-activated protein kinase signaling pathways. Wound-induced gene expression signature in sponges resembles the initial steps of whole-body regeneration in other animals. Also, the set of genes responding to wounding in sponges included putative orthologs of cancer-related human genes. Further insights can be gained from taking sponge wound healing as an experimental system to understand how ancient genes and regulatory networks determine healthy animal tissues.
Collapse
Affiliation(s)
- Yu-Chen Wu
- Research Unit Marine Microbiology, Department Marine Ecology, GEOMAR Helmholtz Centre for Ocean Research, Kiel, Germany
- Christian-Albrechts University of Kiel, Kiel, Germany
| | - Soeren Franzenburg
- Institute of Clinical Molecular Biology (IKMB), Christian-Albrechts University of Kiel, Kiel, Germany
| | - Marta Ribes
- Department Marine Biology and Oceanography, Institute of Marine Sciences (ICM-CSIC), Barcelona, Spain
| | - Lucía Pita
- Research Unit Marine Microbiology, Department Marine Ecology, GEOMAR Helmholtz Centre for Ocean Research, Kiel, Germany.
- Department Marine Biology and Oceanography, Institute of Marine Sciences (ICM-CSIC), Barcelona, Spain.
| |
Collapse
|
41
|
Le D, Sabry Z, Chandra A, Kristan WB, Collins EMS, Kristan WB. Planarian fragments behave as whole animals. Curr Biol 2021; 31:5111-5117.e4. [PMID: 34624209 DOI: 10.1016/j.cub.2021.09.056] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/08/2021] [Accepted: 09/22/2021] [Indexed: 12/11/2022]
Abstract
Behavioral responses of freshwater planarians have been studied for over a century.1 In recent decades, behavior has been used as a readout to study planarian development and regeneration,2-6 wound healing,7,8 molecular evolution,4,9,10 neurotoxicology,11-13 and learning and memory.14-17The planarian nervous system is among the simplest of the bilaterally symmetric animals,18 with an anterior brain attached to two ventral nerve cords interconnected by multiple commissures. We found that, in response to mechanical and near-UV stimulation, head stimulation produces turning, tail stimulation produces contraction, and trunk stimulation produces midbody elongation in the planarian Dugesia japonica. When cut into two or three pieces, the anterior end of each headless piece switched its behavior to turning instead of elongation; i.e., it responded as though it were the head. In addition, posterior ends of the head and midbody pieces sometimes produced contraction instead of elongation. Thus, each severed piece acts like an intact animal, with each midbody region having nearly complete behavioral capabilities. These observations show that each midbody region reads the global state of the organism and adapts its response to incoming signals from the remaining tissue. Selective lateral incisions showed that the changes in behavior are not due to nonselective pain responses and that the ventral nerve cords and cross-connectives are responsible for coordinating local behaviors. Our findings highlight a fast functional reorganization of the planarian nervous system that complements the slower repairs provided by regeneration. This reorganization provides needed behavioral responses for survival as regeneration proceeds.
Collapse
Affiliation(s)
- Dylan Le
- Section of Neurobiology, Division of Biological Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Ziad Sabry
- Department of Biology, Swarthmore College, 500 College Avenue, Swarthmore, PA 19081, USA
| | - Aarav Chandra
- The Bishop's School, 7607 La Jolla Boulevard, La Jolla, CA 92037, USA
| | - William B Kristan
- Department of Biological Sciences, California State University San Marcos, 333 South Twin Oaks Valley Road, San Marcos, CA 92096, USA
| | - Eva-Maria S Collins
- Department of Biology, Swarthmore College, 500 College Avenue, Swarthmore, PA 19081, USA; Department of Physics and Astronomy, 500 College Avenue, Swarthmore College, Swarthmore, PA 19081, USA; Department of Physics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, 415 Curie Boulevard, Philadelphia, PA, USA.
| | - William B Kristan
- Section of Neurobiology, Division of Biological Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| |
Collapse
|
42
|
Wnt/β-catenin signaling acts cell-autonomously to promote cardiomyocyte regeneration in the zebrafish heart. Dev Biol 2021; 481:226-237. [PMID: 34748730 DOI: 10.1016/j.ydbio.2021.11.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 10/15/2021] [Accepted: 11/02/2021] [Indexed: 12/22/2022]
Abstract
Zebrafish can achieve scar-free healing of heart injuries, and robustly replace all cardiomyocytes lost to injury via dedifferentiation and proliferation of mature cardiomyocytes. Previous studies suggested that Wnt/β-catenin signaling is active in the injured zebrafish heart, where it induces fibrosis and prevents cardiomyocyte cell cycling. Here, via targeting the destruction complex of the Wnt/β-catenin pathway with pharmacological and genetic tools, we demonstrate that Wnt/β-catenin activity is required for cardiomyocyte proliferation and dedifferentiation, as well as for maturation of the scar during regeneration. Using cardiomyocyte-specific conditional inhibition of the pathway, we show that Wnt/β-catenin signaling acts cell-autonomously to promote cardiomyocyte proliferation. Our results stand in contrast to previous reports and rather support a model in which Wnt/β-catenin signaling plays a positive role during heart regeneration in zebrafish.
Collapse
|
43
|
Lin YF, Sam J, Evans T. Sirt1 promotes tissue regeneration in zebrafish through regulating the mitochondrial unfolded protein response. iScience 2021; 24:103118. [PMID: 34622167 PMCID: PMC8479786 DOI: 10.1016/j.isci.2021.103118] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 08/12/2021] [Accepted: 09/09/2021] [Indexed: 11/16/2022] Open
Abstract
The mitochondrial unfolded protein response (UPRmt) is an organellar stress signaling pathway that functions to detect and restore disruption of mitochondrial proteostasis. The UPRmt is involved in a wide range of physiological and disease conditions, including aging, stem cell maintenance, innate immunity, neurodegeneration, and cancer. Here we report that the UPRmt is integral to zebrafish fin regeneration. Taking advantage of a novel zebrafish UPRmt reporter, we observed that UPRmt activation occurs in regenerating fin tissue shortly after injury. Through chemical and genetic approaches, we discovered that the Sirt1-UPRmt pathway, best known for its role in promoting lifespan extension, is crucial for fin regeneration. The metabolism of NAD+ is an important contributor to Sirt1 activity in this context. We propose that Sirt1 activation induces mitochondrial biogenesis in injured fin tissue, which leads to UPRmt activation and promotes tissue regeneration.
Collapse
Affiliation(s)
- Yi-Fan Lin
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, LC-708, New York, NY 10065, USA
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, 30013, Taiwan
- Department of Life Science, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Jessica Sam
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, LC-708, New York, NY 10065, USA
| | - Todd Evans
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, LC-708, New York, NY 10065, USA
| |
Collapse
|
44
|
Molina MD, Cebrià F. Decoding Stem Cells: An Overview on Planarian Stem Cell Heterogeneity and Lineage Progression. Biomolecules 2021; 11:1532. [PMID: 34680165 PMCID: PMC8533874 DOI: 10.3390/biom11101532] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 01/26/2023] Open
Abstract
Planarians are flatworms capable of whole-body regeneration, able to regrow any missing body part after injury or amputation. The extraordinary regenerative capacity of planarians is based upon the presence in the adult of a large population of somatic pluripotent stem cells. These cells, called neoblasts, offer a unique system to study the process of stem cell specification and differentiation in vivo. In recent years, FACS-based isolation of neoblasts, RNAi functional analyses as well as high-throughput approaches such as single-cell sequencing have allowed a rapid progress in our understanding of many different aspects of neoblast biology. Here, we summarize our current knowledge on the molecular signatures that define planarian neoblasts heterogeneity, which includes a percentage of truly pluripotent stem cells, and guide the commitment of pluripotent neoblasts into lineage-specific progenitor cells, as well as their differentiation into specific planarian cell types.
Collapse
Affiliation(s)
- M. Dolores Molina
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, 08028 Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), 08028 Barcelona, Spain
| | - Francesc Cebrià
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, 08028 Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), 08028 Barcelona, Spain
| |
Collapse
|
45
|
Zheng H, Liu H, Xu Q, Wang W, Li L, Ye G, Wen X, Chen F, Yu Y. PI3K Plays an Essential Role in Planarian Regeneration and Tissue Maintenance. Front Cell Dev Biol 2021; 9:649656. [PMID: 34422792 PMCID: PMC8377419 DOI: 10.3389/fcell.2021.649656] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 07/19/2021] [Indexed: 11/13/2022] Open
Abstract
Phosphatidylinositol 3-kinase (PI3K) signaling plays a central role in various biological processes, and its abnormality leads to a broad spectrum of human diseases, such as cancer, fibrosis, and immunological disorders. However, the mechanisms by which PI3K signaling regulates the behavior of stem cells during regeneration are poorly understood. Planarian flatworms possess abundant adult stem cells (called neoblasts) allowing them to develop remarkable regenerative capabilities, thus the animals represent an ideal model for studying stem cells and regenerative medicine in vivo. In this study, the spatiotemporal expression pattern of Djpi3k, a PI3K ortholog in the planarian Dugesia japonica, was investigated and suggests its potential role in wound response and tissue regeneration. A loss-of-function study was conducted using small molecules and RNA interference technique, providing evidence that PI3K signaling is required for blastema regrowth and cilia maintenance during planarian regeneration and homeostasis. Interestingly, the mitotic and apoptotic responses to amputation are substantially abated in PI3K inhibitor-treated regenerating animals, while knockdown of Djpi3k alleviates the mitotic response and postpones the peak of apoptotic cell death, which may contribute to the varying degrees of regenerative defects induced by the pharmacological and genetic approaches. These observations reveal novel roles for PI3K signaling in the regulation of the cellular responses to amputation during planarian regeneration and provide insights for investigating the disease-related genes in the regeneration-competent organism in vivo.
Collapse
Affiliation(s)
- Hanxue Zheng
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Hongbo Liu
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Qian Xu
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Wenjun Wang
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Linfeng Li
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Gang Ye
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Xiaomin Wen
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Fulin Chen
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China.,Provincial Key Laboratory of Biotechnology of Shaanxi, Northwest University, Xi'an, China.,Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, China
| | - Yuan Yu
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China.,Provincial Key Laboratory of Biotechnology of Shaanxi, Northwest University, Xi'an, China.,Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, China
| |
Collapse
|
46
|
Nakamura A, Goto Y, Kondo Y, Aoki K. Shedding light on developmental ERK signaling with genetically encoded biosensors. Development 2021; 148:271153. [PMID: 34338283 DOI: 10.1242/dev.199767] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The extracellular signal-regulated kinase (ERK) pathway governs cell proliferation, differentiation and migration, and therefore plays key roles in various developmental and regenerative processes. Recent advances in genetically encoded fluorescent biosensors have unveiled hitherto unrecognized ERK activation dynamics in space and time and their functional importance mainly in cultured cells. However, ERK dynamics during embryonic development have still only been visualized in limited numbers of model organisms, and we are far from a sufficient understanding of the roles played by developmental ERK dynamics. In this Review, we first provide an overview of the biosensors used for visualization of ERK activity in live cells. Second, we highlight the applications of the biosensors to developmental studies of model organisms and discuss the current understanding of how ERK dynamics are encoded and decoded for cell fate decision-making.
Collapse
Affiliation(s)
- Akinobu Nakamura
- Division of Quantitative Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan.,Quantitative Biology Research Group, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan
| | - Yuhei Goto
- Division of Quantitative Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan.,Quantitative Biology Research Group, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan.,Department of Basic Biology, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan
| | - Yohei Kondo
- Division of Quantitative Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan.,Quantitative Biology Research Group, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan.,Department of Basic Biology, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan
| | - Kazuhiro Aoki
- Division of Quantitative Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan.,Quantitative Biology Research Group, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan.,Department of Basic Biology, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan.,IRCC International Research Collaboration Center, National Institutes of Natural Sciences, 4-3-13 Toranomon, Minato-ku, Tokyo 105-0001, Japan
| |
Collapse
|
47
|
Srivastava M. Beyond Casual Resemblances: Rigorous Frameworks for Comparing Regeneration Across Species. Annu Rev Cell Dev Biol 2021; 37:415-440. [PMID: 34288710 DOI: 10.1146/annurev-cellbio-120319-114716] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The majority of animal phyla have species that can regenerate. Comparing regeneration across animals can reconstruct the molecular and cellular evolutionary history of this process. Recent studies have revealed some similarity in regeneration mechanisms, but rigorous comparative methods are needed to assess whether these resemblances are ancestral pathways (homology) or are the result of convergent evolution (homoplasy). This review aims to provide a framework for comparing regeneration across animals, focusing on gene regulatory networks (GRNs), which are substrates for assessing process homology. The homology of the wound-induced activation of Wnt signaling and of adult stem cells are discussed as examples of ongoing studies of regeneration that enable comparisons in a GRN framework. Expanding the study of regeneration GRNs in currently studied species and broadening taxonomic sampling for these approaches will identify processes that are unifying principles of regeneration biology across animals. These insights are important both for evolutionary studies of regeneration and for human regenerative medicine. Expected final online publication date for the Annual Review of Cell and Developmental Biology, Volume 37 is October 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Mansi Srivastava
- Department of Organismic and Evolutionary Biology and Museum of Comparative Zoology, Harvard University, Cambridge, Massachusetts 02138, USA;
| |
Collapse
|
48
|
Gambino G, Ippolito C, Evangelista M, Salvetti A, Rossi L. Sub-Lethal 5-Fluorouracil Dose Challenges Planarian Stem Cells Promoting Transcriptional Profile Changes in the Pluripotent Sigma-Class Neoblasts. Biomolecules 2021; 11:biom11070949. [PMID: 34206807 PMCID: PMC8301986 DOI: 10.3390/biom11070949] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/14/2021] [Accepted: 06/22/2021] [Indexed: 11/16/2022] Open
Abstract
Under physiological conditions, the complex planarian neoblast system is a composite of hierarchically organized stem cell sub-populations with sigma-class neoblasts, including clonogenic neoblasts, endowed with larger self-renewal and differentiation capabilities, thus generating all the other sub-populations and dominating the regenerative process. This complex system responds to differentiated tissue demands, ensuring a continuous cell turnover in a way to replace aged specialized cells and maintain tissue functionality. Potency of the neoblast system can be appreciated under challenging conditions in which these stem cells are massively depleted and the few remaining repopulate the entire body, ensuring animal resilience. These challenging conditions offer the possibility to deepen the relationships among different neoblast sub-populations, allowing to expose uncanonical properties that are negligible under physiological conditions. In this paper, we employ short, sub-lethal 5-fluorouracil treatment to specifically affect proliferating cells passing through the S phase and demonstrate that S-phase slowdown triggers a shift in the transcriptional profile of sigma neoblasts, which reduces the expression of their hallmark sox-P1. Later, some cells reactivate sox-P1 expression, suggesting that some neoblasts in the earlier steps of commitment could modulate their expression profile, reacquiring a wider differentiative potential.
Collapse
Affiliation(s)
- Gaetana Gambino
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (G.G.); (C.I.); (L.R.)
| | - Chiara Ippolito
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (G.G.); (C.I.); (L.R.)
| | | | - Alessandra Salvetti
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (G.G.); (C.I.); (L.R.)
- Correspondence: ; Tel.: +39-0502219108
| | - Leonardo Rossi
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (G.G.); (C.I.); (L.R.)
| |
Collapse
|
49
|
Bohr TE, Shiroor DA, Adler CE. Planarian stem cells sense the identity of the missing pharynx to launch its targeted regeneration. eLife 2021; 10:e68830. [PMID: 34156924 PMCID: PMC8219383 DOI: 10.7554/elife.68830] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 05/22/2021] [Indexed: 12/25/2022] Open
Abstract
In order to regenerate tissues successfully, stem cells must detect injuries and restore missing cell types through largely unknown mechanisms. Planarian flatworms have an extensive stem cell population responsible for regenerating any organ after amputation. Here, we compare planarian stem cell responses to different injuries by either amputation of a single organ, the pharynx, or removal of tissues from other organs by decapitation. We find that planarian stem cells adopt distinct behaviors depending on what tissue is missing to target progenitor and tissue production towards missing tissues. Loss of non-pharyngeal tissues only increases non-pharyngeal progenitors, while pharynx removal selectively triggers division and expansion of pharynx progenitors. By pharmacologically inhibiting either mitosis or activation of the MAP kinase ERK, we identify a narrow window of time during which stem cell division and ERK signaling produces pharynx progenitors necessary for regeneration. These results indicate that planarian stem cells can tailor their output to match the regenerative needs of the animal.
Collapse
Affiliation(s)
- Tisha E Bohr
- Department of Molecular Medicine, Cornell University College of Veterinary MedicineIthacaUnited States
| | - Divya A Shiroor
- Department of Molecular Medicine, Cornell University College of Veterinary MedicineIthacaUnited States
| | - Carolyn E Adler
- Department of Molecular Medicine, Cornell University College of Veterinary MedicineIthacaUnited States
| |
Collapse
|
50
|
Ramirez AN, Loubet-Senear K, Srivastava M. A Regulatory Program for Initiation of Wnt Signaling during Posterior Regeneration. Cell Rep 2021; 32:108098. [PMID: 32877680 DOI: 10.1016/j.celrep.2020.108098] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 06/03/2020] [Accepted: 08/11/2020] [Indexed: 12/17/2022] Open
Abstract
Whole-body regeneration relies on the re-establishment of body axes for patterning of new tissue. Wnt signaling is required to correctly regenerate tissues along the primary axis in many animals. However, the causal mechanisms that first launch Wnt signaling during regeneration are poorly characterized. We use the acoel worm Hofstenia miamia to identify processes that initiate Wnt signaling during posterior regeneration and find that the ligand wnt-3 is upregulated early in posterior-facing wounds. Functional studies reveal that wnt-3 is required to regenerate posterior tissues. wnt-3 is expressed in stem cells, it is needed for their proliferation, and its function is stem cell dependent. Chromatin accessibility data reveal that wnt-3 activation requires input from the general wound response. In addition, the expression of a different Wnt ligand, wnt-1, before amputation is required for wound-induced activation of wnt-3. Our study establishes a gene regulatory network for initiating Wnt signaling in posterior tissues in a bilaterian.
Collapse
Affiliation(s)
- Alyson N Ramirez
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Kaitlyn Loubet-Senear
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Mansi Srivastava
- Department of Organismic and Evolutionary Biology, Museum of Comparative Zoology, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|