1
|
Liu Y, Li M, Wan QL, Wang X, Mortimer M, Fang WD, Guo LH. Recent advances in bioassays for assessing the toxicity of environmental contaminants in effect-directed analysis. J Environ Sci (China) 2025; 155:343-358. [PMID: 40246470 DOI: 10.1016/j.jes.2024.07.031] [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: 02/02/2024] [Revised: 07/28/2024] [Accepted: 07/30/2024] [Indexed: 04/19/2025]
Abstract
Chemical cocktails in the environment can cause adverse impacts on ecosystems and human health even at low concentrations. Effect-directed analysis (EDA) has proven to be very valuable in identifying key toxic substances in environmental mixtures. For this, it is important to carefully select accurate bioassays from a wide range of tests for EDA when applying it to actual environmental samples. This article reviews studies published from 2014 to 2023 that have applied EDA and summarizes the bioassays and their corresponding biological effects. A total of 127 studies were selected from 591 publications evaluating the toxic effects of environmental samples, including wastewater, surface water, and sediments. Here, bioassays used in EDA are summarized, including the assays that measure specific receptor-mediated modes of action (MOA), induction of xenobiotic metabolism pathways, and induction of adaptive stress response pathways using either in vitro or in vivo bioassays. Also, the identified substances using EDA are discussed based on their MOA. The importance of EDA in establishing a comprehensive approach for the detection of environmental contaminants using bioanalytical methods is emphasized. The current limitations and benefits of using EDA in practical applications are outlined and strategies for moving forward are proposed.
Collapse
Affiliation(s)
- Yao Liu
- College of Life Sciences, China Jiliang University, Hangzhou 310018, China; Institute of Environmental and Health Sciences, China Jiliang University, Hangzhou 310018, China
| | - Minjie Li
- Institute of Environmental and Health Sciences, China Jiliang University, Hangzhou 310018, China; College of Quality and Safety Engineering, China Jiliang University, Hangzhou 310018, China
| | - Qi-Lin Wan
- Institute of Environmental and Health Sciences, China Jiliang University, Hangzhou 310018, China; College of Quality and Safety Engineering, China Jiliang University, Hangzhou 310018, China
| | - Xun Wang
- Institute of Environmental and Health Sciences, China Jiliang University, Hangzhou 310018, China; College of Quality and Safety Engineering, China Jiliang University, Hangzhou 310018, China
| | - Monika Mortimer
- Institute of Environmental and Health Sciences, China Jiliang University, Hangzhou 310018, China; College of Quality and Safety Engineering, China Jiliang University, Hangzhou 310018, China
| | - Wen-Di Fang
- Institute of Environmental and Health Sciences, China Jiliang University, Hangzhou 310018, China; College of Quality and Safety Engineering, China Jiliang University, Hangzhou 310018, China.
| | - Liang-Hong Guo
- Institute of Environmental and Health Sciences, China Jiliang University, Hangzhou 310018, China; College of Quality and Safety Engineering, China Jiliang University, Hangzhou 310018, China.
| |
Collapse
|
2
|
Chen Y, Hou Y, Zeng Q, Wang I, Shang M, Shin K, Hemauer C, Xing X, Kang J, Zhao G, Wang T. Common and specific gene regulatory programs in zebrafish caudal fin regeneration at single-cell resolution. Genome Res 2025; 35:202-218. [PMID: 39809530 PMCID: PMC11789645 DOI: 10.1101/gr.279372.124] [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: 03/18/2024] [Accepted: 11/04/2024] [Indexed: 01/16/2025]
Abstract
Following amputation, zebrafish regenerate their injured caudal fin through lineage-restricted reprogramming. Although previous studies have charted various genetic and epigenetic dimensions of this process, the intricate gene regulatory programs shared by, or unique to, different regenerating cell types remain underinvestigated. Here, we mapped the regulatory landscape of fin regeneration by applying paired snRNA-seq and snATAC-seq on uninjured and regenerating fins. This map delineates the regulatory dynamics of predominant cell populations at multiple stages of regeneration. We observe a marked increase in the accessibility of chromatin regions associated with regenerative and developmental processes at 1 dpa, followed by a gradual closure across major cell types at later stages. This pattern is distinct from that of transcriptomic dynamics, which is characterized by several waves of gene upregulation and downregulation. We identified and in vivo validated cell-type-specific and position-specific regeneration-responsive enhancers and constructed regulatory networks by cell type and stage. Our single-cell resolution transcriptomic and chromatin accessibility map across regenerative stages provides new insights into regeneration regulatory mechanisms and serves as a valuable resource for the community.
Collapse
Affiliation(s)
- Yujie Chen
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Yiran Hou
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Qinglin Zeng
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Irene Wang
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Meiru Shang
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Kwangdeok Shin
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin Madison, Madison, Wisconsin 53705, USA
| | - Christopher Hemauer
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Xiaoyun Xing
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Junsu Kang
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin Madison, Madison, Wisconsin 53705, USA
| | - Guoyan Zhao
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri 63110, USA;
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Ting Wang
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri 63110, USA;
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
3
|
Senk A, Fazzari J, Djonov V. Vascular mimicry in zebrafish fin regeneration: how macrophages build new blood vessels. Angiogenesis 2024; 27:397-410. [PMID: 38546923 PMCID: PMC11303510 DOI: 10.1007/s10456-024-09914-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 03/10/2024] [Indexed: 08/07/2024]
Abstract
Vascular mimicry has been thoroughly investigated in tumor angiogenesis. In this study, we demonstrate for the first time that a process closely resembling tumor vascular mimicry is present during physiological blood vessel formation in tissue regeneration using the zebrafish fin regeneration assay. At the fin-regenerating front, vasculature is formed by mosaic blood vessels with endothelial-like cells possessing the morphological phenotype of a macrophage and co-expressing both endothelial and macrophage markers within single cells. Our data demonstrate that the vascular segments of the regenerating tissue expand, in part, through the transformation of adjacent macrophages into endothelial-like cells, forming functional, perfused channels and contributing to the de novo formation of microvasculature. Inhibiting the formation of tubular vascular-like structures by CVM-1118 prevents vascular mimicry and network formation resulting in a 70% shorter regeneration area with 60% reduced vessel growth and a complete absence of any signs of regeneration in half of the fin area. Additionally, this is associated with a significant reduction in macrophages. Furthermore, depleting macrophages using macrophage inhibitor PLX-3397, results in impaired tissue regeneration and blood vessel formation, namely a reduction in the regeneration area and vessel network by 75% in comparison to controls.
Collapse
Affiliation(s)
- Anita Senk
- Institute of Anatomy, University of Bern, Bern, Switzerland
| | | | - Valentin Djonov
- Institute of Anatomy, University of Bern, Bern, Switzerland.
| |
Collapse
|
4
|
Nakamura M, Kyoda T, Yoshida H, Takebayashi-Suzuki K, Koike R, Takahashi E, Moriyama Y, Wlizla M, Horb ME, Suzuki A. Injury-induced cooperation of InhibinβA and JunB is essential for cell proliferation in Xenopus tadpole tail regeneration. Sci Rep 2024; 14:3679. [PMID: 38355764 PMCID: PMC10867027 DOI: 10.1038/s41598-024-54280-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 02/10/2024] [Indexed: 02/16/2024] Open
Abstract
In animal species that have the capability of regenerating tissues and limbs, cell proliferation is enhanced after wound healing and is essential for the reconstruction of injured tissue. Although the ability to induce cell proliferation is a common feature of such species, the molecular mechanisms that regulate the transition from wound healing to regenerative cell proliferation remain unclear. Here, we show that upon injury, InhibinβA and JunB cooperatively function for this transition during Xenopus tadpole tail regeneration. We found that the expression of inhibin subunit beta A (inhba) and junB proto-oncogene (junb) is induced by injury-activated TGF-β/Smad and MEK/ERK signaling in regenerating tails. Similarly to junb knockout (KO) tadpoles, inhba KO tadpoles show a delay in tail regeneration, and inhba/junb double KO (DKO) tadpoles exhibit severe impairment of tail regeneration compared with either inhba KO or junb KO tadpoles. Importantly, this impairment is associated with a significant reduction of cell proliferation in regenerating tissue. Moreover, JunB regulates tail regeneration via FGF signaling, while InhibinβA likely acts through different mechanisms. These results demonstrate that the cooperation of injury-induced InhibinβA and JunB is critical for regenerative cell proliferation, which is necessary for re-outgrowth of regenerating Xenopus tadpole tails.
Collapse
Affiliation(s)
- Makoto Nakamura
- Amphibian Research Center, Graduate School of Integrated Sciences for Life, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-8526, Japan
- Cardiovascular Research Institute, University of California San Francisco, 555 Mission Bay Boulevard South, San Francisco, CA, 94158, USA
| | - Tatsuya Kyoda
- Amphibian Research Center, Graduate School of Integrated Sciences for Life, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-8526, Japan
| | - Hitoshi Yoshida
- National Xenopus Resource and Eugene Bell Center for Regenerative Biology and Tissue Engineering, Marine Biological Laboratory, Woods Hole, MA, 02543, USA
| | - Kimiko Takebayashi-Suzuki
- Amphibian Research Center, Graduate School of Integrated Sciences for Life, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-8526, Japan
| | - Ryota Koike
- Amphibian Research Center, Graduate School of Integrated Sciences for Life, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-8526, Japan
| | - Eri Takahashi
- Amphibian Research Center, Graduate School of Integrated Sciences for Life, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-8526, Japan
| | - Yuka Moriyama
- Amphibian Research Center, Graduate School of Integrated Sciences for Life, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-8526, Japan
| | - Marcin Wlizla
- National Xenopus Resource and Eugene Bell Center for Regenerative Biology and Tissue Engineering, Marine Biological Laboratory, Woods Hole, MA, 02543, USA
- Embryology, Charles River Laboratories, Wilmington, MA, 01887, USA
| | - Marko E Horb
- National Xenopus Resource and Eugene Bell Center for Regenerative Biology and Tissue Engineering, Marine Biological Laboratory, Woods Hole, MA, 02543, USA
| | - Atsushi Suzuki
- Amphibian Research Center, Graduate School of Integrated Sciences for Life, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-8526, Japan.
| |
Collapse
|
5
|
Zhang S, Li X, Li X, Wang X, Ru S, Tian H. 17β-Trenbolone activates androgen receptor, upregulates transforming growth factor beta/bone morphogenetic protein and Wnt signaling pathways, and induces masculinization of caudal and anal fins in female guppies (Poecilia reticulata). AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2023; 263:106677. [PMID: 37677862 DOI: 10.1016/j.aquatox.2023.106677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 07/13/2023] [Accepted: 08/30/2023] [Indexed: 09/09/2023]
Abstract
Sexually mature female guppies (Poecilia reticulata) were exposed to environmentally relevant concentrations (20, 200, and 2000 ng/L) of 17β-trenbolone for four weeks. As evidenced by the increased caudal fin index and anal fins developing into gonopodium-like structures, exposed females displayed masculinized secondary sexual characteristics. Differential gene expression and subsequent pathway analysis of mRNA sequencing data revealed that the transcription of transforming growth factor beta/bone morphogenetic protein signaling pathway and Wnt signaling pathway were upregulated following 17β-trenbolone exposure. Enzyme-linked immunosorbent assays showed that the bone morphogenetic protein 7 protein content was elevated after 17β-trenbolone exposure. Finally, real-time PCR revealed that 17β-trenbolone treatment significantly increased androgen receptor mRNA levels, and molecular docking showed potent interaction between 17β-trenbolone and guppy androgen receptor. Furthermore, 17β-trenbolone-induced masculinization of caudal and anal fins in female guppies, concomitant to the upregulated expression of differentially expressed genes involved in the above-mentioned two signaling pathways, was significantly inhibited by flutamide (androgen receptor antagonist). These findings demonstrated that 17β-trenbolone masculinized fins of female guppies by activating the androgen receptor. This study revealed that 17β-trenbolone could upregulate signaling pathways related to fin growth and differentiation, and eventually cause caudal and anal fin masculinization in female guppies.
Collapse
Affiliation(s)
- Suqiu Zhang
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, Shandong province, China
| | - Xinyu Li
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, Shandong province, China
| | - Xuefu Li
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, Shandong province, China; College of Life Science, Langfang Normal University, Langfang 065000, Hebei province, China
| | - Xue Wang
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, Shandong province, China
| | - Shaoguo Ru
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, Shandong province, China
| | - Hua Tian
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, Shandong province, China.
| |
Collapse
|
6
|
Rees L, König D, Jaźwińska A. Regeneration of the dermal skeleton and wound epidermis formation depend on BMP signaling in the caudal fin of platyfish. Front Cell Dev Biol 2023; 11:1134451. [PMID: 36846592 PMCID: PMC9946992 DOI: 10.3389/fcell.2023.1134451] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 01/24/2023] [Indexed: 02/11/2023] Open
Abstract
Fin regeneration has been extensively studied in zebrafish, a genetic model organism. Little is known about regulators of this process in distant fish taxa, such as the Poeciliidae family, represented by the platyfish. Here, we used this species to investigate the plasticity of ray branching morphogenesis following either straight amputation or excision of ray triplets. This approach revealed that ray branching can be conditionally shifted to a more distal position, suggesting non-autonomous regulation of bone patterning. To gain molecular insights into regeneration of fin-specific dermal skeleton elements, actinotrichia and lepidotrichia, we localized expression of the actinodin genes and bmp2 in the regenerative outgrowth. Blocking of the BMP type-I receptor suppressed phospho-Smad1/5 immunoreactivity, and impaired fin regeneration after blastema formation. The resulting phenotype was characterized by the absence of bone and actinotrichia restoration. In addition, the wound epidermis displayed extensive thickening. This malformation was associated with expanded Tp63 expression from the basal epithelium towards more superficial layers, suggesting abnormal tissue differentiation. Our data add to the increasing evidence for the integrative role of BMP signaling in epidermal and skeletal tissue formation during fin regeneration. This expands our understanding of common mechanisms guiding appendage restoration in diverse clades of teleosts.
Collapse
Affiliation(s)
- Lana Rees
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Désirée König
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | | |
Collapse
|
7
|
Henke K, Farmer DT, Niu X, Kraus JM, Galloway JL, Youngstrom DW. Genetically engineered zebrafish as models of skeletal development and regeneration. Bone 2023; 167:116611. [PMID: 36395960 PMCID: PMC11080330 DOI: 10.1016/j.bone.2022.116611] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/01/2022] [Accepted: 11/08/2022] [Indexed: 11/16/2022]
Abstract
Zebrafish (Danio rerio) are aquatic vertebrates with significant homology to their terrestrial counterparts. While zebrafish have a centuries-long track record in developmental and regenerative biology, their utility has grown exponentially with the onset of modern genetics. This is exemplified in studies focused on skeletal development and repair. Herein, the numerous contributions of zebrafish to our understanding of the basic science of cartilage, bone, tendon/ligament, and other skeletal tissues are described, with a particular focus on applications to development and regeneration. We summarize the genetic strengths that have made the zebrafish a powerful model to understand skeletal biology. We also highlight the large body of existing tools and techniques available to understand skeletal development and repair in the zebrafish and introduce emerging methods that will aid in novel discoveries in skeletal biology. Finally, we review the unique contributions of zebrafish to our understanding of regeneration and highlight diverse routes of repair in different contexts of injury. We conclude that zebrafish will continue to fill a niche of increasing breadth and depth in the study of basic cellular mechanisms of skeletal biology.
Collapse
Affiliation(s)
- Katrin Henke
- Department of Orthopaedics, Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | - D'Juan T Farmer
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA 90095, USA; Department of Orthopaedic Surgery, University of California, Los Angeles, CA 90095, USA.
| | - Xubo Niu
- Center for Regenerative Medicine, Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| | - Jessica M Kraus
- Department of Orthopaedic Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA.
| | - Jenna L Galloway
- Center for Regenerative Medicine, Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| | - Daniel W Youngstrom
- Department of Orthopaedic Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA.
| |
Collapse
|
8
|
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: 9] [Impact Index Per Article: 3.0] [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
|
9
|
Chowdhury K, Lin S, Lai SL. Comparative Study in Zebrafish and Medaka Unravels the Mechanisms of Tissue Regeneration. Front Ecol Evol 2022. [DOI: 10.3389/fevo.2022.783818] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Tissue regeneration has been in the spotlight of research for its fascinating nature and potential applications in human diseases. The trait of regenerative capacity occurs diversely across species and tissue contexts, while it seems to decline over evolution. Organisms with variable regenerative capacity are usually distinct in phylogeny, anatomy, and physiology. This phenomenon hinders the feasibility of studying tissue regeneration by directly comparing regenerative with non-regenerative animals, such as zebrafish (Danio rerio) and mice (Mus musculus). Medaka (Oryzias latipes) is a fish model with a complete reference genome and shares a common ancestor with zebrafish approximately 110–200 million years ago (compared to 650 million years with mice). Medaka shares similar features with zebrafish, including size, diet, organ system, gross anatomy, and living environment. However, while zebrafish regenerate almost every organ upon experimental injury, medaka shows uneven regenerative capacity. Their common and distinct biological features make them a unique platform for reciprocal analyses to understand the mechanisms of tissue regeneration. Here we summarize current knowledge about tissue regeneration in these fish models in terms of injured tissues, repairing mechanisms, available materials, and established technologies. We further highlight the concept of inter-species and inter-organ comparisons, which may reveal mechanistic insights and hint at therapeutic strategies for human diseases.
Collapse
|
10
|
Riley SE, Feng Y, Hansen CG. Hippo-Yap/Taz signalling in zebrafish regeneration. NPJ Regen Med 2022; 7:9. [PMID: 35087046 PMCID: PMC8795407 DOI: 10.1038/s41536-022-00209-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 12/14/2021] [Indexed: 12/29/2022] Open
Abstract
The extent of tissue regeneration varies widely between species. Mammals have a limited regenerative capacity whilst lower vertebrates such as the zebrafish (Danio rerio), a freshwater teleost, can robustly regenerate a range of tissues, including the spinal cord, heart, and fin. The molecular and cellular basis of this altered response is one of intense investigation. In this review, we summarise the current understanding of the association between zebrafish regeneration and Hippo pathway function, a phosphorylation cascade that regulates cell proliferation, mechanotransduction, stem cell fate, and tumorigenesis, amongst others. We also compare this function to Hippo pathway activity in the regenerative response of other species. We find that the Hippo pathway effectors Yap/Taz facilitate zebrafish regeneration and that this appears to be latent in mammals, suggesting that therapeutically promoting precise and temporal YAP/TAZ signalling in humans may enhance regeneration and hence reduce morbidity.
Collapse
Affiliation(s)
- Susanna E Riley
- University of Edinburgh Centre for Inflammation Research, Institute for Regeneration and Repair, Queen's Medical Research Institute, Edinburgh bioQuarter, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Yi Feng
- University of Edinburgh Centre for Inflammation Research, Institute for Regeneration and Repair, Queen's Medical Research Institute, Edinburgh bioQuarter, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Carsten Gram Hansen
- University of Edinburgh Centre for Inflammation Research, Institute for Regeneration and Repair, Queen's Medical Research Institute, Edinburgh bioQuarter, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK.
| |
Collapse
|
11
|
Collagen fibers provide guidance cues for capillary regrowth during regenerative angiogenesis in zebrafish. Sci Rep 2021; 11:19520. [PMID: 34593884 PMCID: PMC8484481 DOI: 10.1038/s41598-021-98852-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 09/09/2021] [Indexed: 12/16/2022] Open
Abstract
Although well investigated, the importance of collagen fibers in supporting angiogenesis is not well understood. In this study, we demonstrate that extracellular collagen fibers provide guidance cues for endothelial cell migration during regenerative angiogenesis in the caudal zebrafish fin. Inhibition of collagen cross-linking by β-Aminopropionitrile results in a 70% shorter regeneration area with 50% reduced vessel growth and disintegrated collagen fibers. The disrupted collagen scaffold impedes endothelial cell migration and induces formation of abnormal angioma-like blood vessels. Treatment of the Fli//colRN zebrafish line with the prodrug Nifurpirinol, which selectively damages the active collagen-producing 1α2 cells, reduced the regeneration area and vascular growth by 50% with wider, but less inter-connected, capillary segments. The regenerated area contained larger vessels partially covered by endothelial cells embedded in atypical extracellular matrix containing cell debris and apoptotic bodies, macrophages and granulocytes. Similar experiments performed in early embryonic zebrafish suggested that collagens are important also during embryonic angiogenesis. In vitro assays revealed that collagen I allows for the most efficient endothelial cell migration, followed by collagen IV relative to the complete absence of exogenous matrix support. Our data demonstrates severe vascular defects and restricted fin regeneration when collagens are impaired. Collagen I therefore, provides support and guidance for endothelial cell migration while collagen IV is responsible for proper lumen formation and vascular integrity.
Collapse
|
12
|
Sinclair JW, Hoying DR, Bresciani E, Nogare DD, Needle CD, Berger A, Wu W, Bishop K, Elkahloun AG, Chitnis A, Liu P, Burgess SM. The Warburg effect is necessary to promote glycosylation in the blastema during zebrafish tail regeneration. NPJ Regen Med 2021; 6:55. [PMID: 34518542 PMCID: PMC8437957 DOI: 10.1038/s41536-021-00163-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 08/11/2021] [Indexed: 12/21/2022] Open
Abstract
Throughout their lifetime, fish maintain a high capacity for regenerating complex tissues after injury. We utilized a larval tail regeneration assay in the zebrafish Danio rerio, which serves as an ideal model of appendage regeneration due to its easy manipulation, relatively simple mixture of cell types, and superior imaging properties. Regeneration of the embryonic zebrafish tail requires development of a blastema, a mass of dedifferentiated cells capable of replacing lost tissue, a crucial step in all known examples of appendage regeneration. Using this model, we show that tail amputation triggers an obligate metabolic shift to promote glucose metabolism during early regeneration similar to the Warburg effect observed in tumor forming cells. Inhibition of glucose metabolism did not affect the overall health of the embryo but completely blocked the tail from regenerating after amputation due to the failure to form a functional blastema. We performed a time series of single-cell RNA sequencing on regenerating tails with and without inhibition of glucose metabolism. We demonstrated that metabolic reprogramming is required for sustained TGF-β signaling and blocking glucose metabolism largely mimicked inhibition of TGF-β receptors, both resulting in an aberrant blastema. Finally, we showed using genetic ablation of three possible metabolic pathways for glucose, that metabolic reprogramming is required to provide glucose specifically to the hexosamine biosynthetic pathway while neither glycolysis nor the pentose phosphate pathway were necessary for regeneration.
Collapse
Affiliation(s)
- Jason W Sinclair
- Translational and Functional Genomics Branch, National Human Genome Research Institute, Bethesda, MD, USA
| | - David R Hoying
- Translational and Functional Genomics Branch, National Human Genome Research Institute, Bethesda, MD, USA
| | - Erica Bresciani
- Translational and Functional Genomics Branch, National Human Genome Research Institute, Bethesda, MD, USA
| | - Damian Dalle Nogare
- Aquatic Models of Human Development Affinity Group, National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Carli D Needle
- Translational and Functional Genomics Branch, National Human Genome Research Institute, Bethesda, MD, USA
| | - Alexandra Berger
- Translational and Functional Genomics Branch, National Human Genome Research Institute, Bethesda, MD, USA
| | - Weiwei Wu
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, Bethesda, MD, USA
| | - Kevin Bishop
- Translational and Functional Genomics Branch, National Human Genome Research Institute, Bethesda, MD, USA
| | - Abdel G Elkahloun
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, Bethesda, MD, USA
| | - Ajay Chitnis
- Aquatic Models of Human Development Affinity Group, National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Paul Liu
- Translational and Functional Genomics Branch, National Human Genome Research Institute, Bethesda, MD, USA
| | - Shawn M Burgess
- Translational and Functional Genomics Branch, National Human Genome Research Institute, Bethesda, MD, USA.
| |
Collapse
|
13
|
Dagenais P, Blanchoud S, Pury D, Pfefferli C, Aegerter-Wilmsen T, Aegerter CM, Jaźwińska A. Hydrodynamic stress and phenotypic plasticity of the zebrafish regenerating fin. J Exp Biol 2021; 224:271142. [PMID: 34338301 DOI: 10.1242/jeb.242309] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 06/14/2021] [Indexed: 01/23/2023]
Abstract
Understanding how extrinsic factors modulate genetically encoded information to produce a specific phenotype is of prime scientific interest. In particular, the feedback mechanism between abiotic forces and locomotory organs during morphogenesis to achieve efficient movement is a highly relevant example of such modulation. The study of this developmental process can provide unique insights on the transduction of cues at the interface between physics and biology. Here, we take advantage of the natural ability of adult zebrafish to regenerate their amputated fins to assess its morphogenic plasticity upon external modulations. Using a variety of surgical and chemical treatments, we could induce phenotypic responses to the structure of the fin. Through the ablation of specific rays in regenerating caudal fins, we generated artificially narrowed appendages in which the fin cleft depth and the positioning of rays bifurcations were perturbed compared with normal regenerates. To dissect the role of mechanotransduction in this process, we investigated the patterns of hydrodynamic forces acting on the surface of a zebrafish fin during regeneration by using particle tracking velocimetry on a range of biomimetic hydrofoils. This experimental approach enabled us to quantitatively compare hydrodynamic stress distributions over flapping fins of varying sizes and shapes. As a result, viscous shear stress acting on the distal margin of regenerating fins and the resulting internal tension are proposed as suitable signals for guiding the regulation of ray growth dynamics and branching pattern. Our findings suggest that mechanical forces are involved in the fine-tuning of the locomotory organ during fin morphogenesis.
Collapse
Affiliation(s)
- Paule Dagenais
- Physik-Institut, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Simon Blanchoud
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| | - David Pury
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| | - Catherine Pfefferli
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| | - Tinri Aegerter-Wilmsen
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Christof M Aegerter
- Physik-Institut, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland.,Department of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Anna Jaźwińska
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| |
Collapse
|
14
|
Durant F, Whited JL. Finding Solutions for Fibrosis: Understanding the Innate Mechanisms Used by Super-Regenerator Vertebrates to Combat Scarring. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100407. [PMID: 34032013 PMCID: PMC8336523 DOI: 10.1002/advs.202100407] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/12/2021] [Indexed: 05/08/2023]
Abstract
Soft tissue fibrosis and cutaneous scarring represent massive clinical burdens to millions of patients per year and the therapeutic options available are currently quite limited. Despite what is known about the process of fibrosis in mammals, novel approaches for combating fibrosis and scarring are necessary. It is hypothesized that scarring has evolved as a solution to maximize healing speed to reduce fluid loss and infection. This hypothesis, however, is complicated by regenerative animals, which have arguably the most remarkable healing abilities and are capable of scar-free healing. This review explores the differences observed between adult mammalian healing that typically results in fibrosis versus healing in regenerative animals that heal scarlessly. Each stage of wound healing is surveyed in depth from the perspective of many regenerative and fibrotic healers so as to identify the most important molecular and physiological variances along the way to disparate injury repair outcomes. Understanding how these powerful model systems accomplish the feat of scar-free healing may provide critical therapeutic approaches to the treatment or prevention of fibrosis.
Collapse
Affiliation(s)
- Fallon Durant
- Department of Stem Cell and Regenerative BiologyHarvard UniversityCambridgeMA02138USA
| | - Jessica L. Whited
- Department of Stem Cell and Regenerative BiologyHarvard UniversityCambridgeMA02138USA
- The Harvard Stem Cell InstituteCambridgeMA02138USA
| |
Collapse
|
15
|
Molina-Villa T, Ramírez-Vidal L, Mendoza V, Escalante-Alcalde D, López-Casillas F. Chordacentrum mineralization is delayed in zebrafish betaglycan-null mutants. Dev Dyn 2021; 251:213-225. [PMID: 34228380 DOI: 10.1002/dvdy.393] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 06/04/2021] [Accepted: 06/20/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The Transforming Growth Factor β (TGFβ) family is a group of related proteins that signal through a type I and type II receptors. Betaglycan, also known as the type III receptor (Tgfbr3), is a coreceptor for various ligands of the TGFβ family that participates in heart, liver and kidney development as revealed by the tgfbr3-null mouse, as well as in angiogenesis as revealed by Tgfbr3 downregulation in morphant zebrafish. RESULTS Here, we present CRISPR/Cas9-derived zebrafish Tgfbr3-null mutants, which exhibited unaltered embryonic angiogenesis and developed into fertile adults. One reproducible phenotype displayed by these Tgfbr3-null mutants is delayed chordacentra mineralization, which nonetheless does not result in vertebral abnormalities in the adult fishes. We also report that the canonical TGFβ signaling pathway is needed for proper chordacentra mineralization and that Tgfbr3 absence decreases this signal in the notochordal cells responsible for this process. CONCLUSION Betaglycan's "ligand presentation" function contributes to the optimal TGFβ signaling required for zebrafish chordacentra mineralization.
Collapse
Affiliation(s)
- Tonatiuh Molina-Villa
- Department of Cellular and Developmental Biology, Institute of Cellular Physiology, UNAM, México City, Mexico
| | - Lizbeth Ramírez-Vidal
- Department of Cellular and Developmental Biology, Institute of Cellular Physiology, UNAM, México City, Mexico
| | - Valentín Mendoza
- Department of Cellular and Developmental Biology, Institute of Cellular Physiology, UNAM, México City, Mexico
| | - Diana Escalante-Alcalde
- Division of Neurosciences, Department of Neural Development and Physiology, Institute of Cellular Physiology, UNAM, México City, Mexico
| | - Fernando López-Casillas
- Department of Cellular and Developmental Biology, Institute of Cellular Physiology, UNAM, México City, Mexico
| |
Collapse
|
16
|
Feleke M, Bennett S, Chen J, Chandler D, Hu X, Xu J. Biological insights into the rapid tissue regeneration of freshwater crayfish and crustaceans. Cell Biochem Funct 2021; 39:740-753. [PMID: 34165197 DOI: 10.1002/cbf.3653] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 06/03/2021] [Indexed: 11/12/2022]
Abstract
The freshwater crayfish is capable of regenerating limbs, following autotomy, injury and predation. In arthropod species, regeneration and moulting are two processes linked and strongly regulated by ecdysone. The regeneration of crayfish limbs is divided into wound healing, blastema formation, cellular reprogramming and tissue patterning. Limb blastema cells undergo proliferation, dedifferentiation and redifferentiation. A limb bud, containing folded segments of the regenerating limb, is encased within a cuticular sheath. The functional limb regenerates, in proecdysis, in two to three consecutive moults. Rapid tissue growth is regulated by hormones, limb nerves and local cells. The TGF-β/activin signalling pathway has been determined in the crayfish, P. fallax f. virginalis, and is suggested as a potential regulator of tissue regeneration. In this review article, we discuss current understanding of tissue regeneration in the crayfish and various crustaceans. A thorough understanding of the cellular, genetic and molecular pathways of these biological processes is promising for the development of therapeutic applications for a wide array of diseases in regenerative medicine.
Collapse
Affiliation(s)
- Mesalie Feleke
- Division of Regenerative Biology, School of Biomedical Sciences, University of Western Australia, Perth, Western Australia, Australia
| | - Samuel Bennett
- Division of Regenerative Biology, School of Biomedical Sciences, University of Western Australia, Perth, Western Australia, Australia
| | - Jiazhi Chen
- Division of Regenerative Biology, School of Biomedical Sciences, University of Western Australia, Perth, Western Australia, Australia.,Guangdong Provincial Key Laboratory of Industrial Surfactant, Guangdong Research Institute of Petrochemical and Fine Chemical Engineering, Guangdong Academy of Sciences, Guangzhou, China
| | - David Chandler
- Australian Genome Research Facility, Medical Research Foundation, Royal Perth Hospital, Perth, Western Australia, Australia
| | - Xiaoyong Hu
- Guangdong Provincial Key Laboratory of Industrial Surfactant, Guangdong Research Institute of Petrochemical and Fine Chemical Engineering, Guangdong Academy of Sciences, Guangzhou, China
| | - Jiake Xu
- Division of Regenerative Biology, School of Biomedical Sciences, University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
17
|
Nakamura M, Yoshida H, Moriyama Y, Kawakita I, Wlizla M, Takebayashi-Suzuki K, Horb ME, Suzuki A. TGF-β1 signaling is essential for tissue regeneration in the Xenopus tadpole tail. Biochem Biophys Res Commun 2021; 565:91-96. [PMID: 34102475 DOI: 10.1016/j.bbrc.2021.05.082] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 05/23/2021] [Indexed: 12/25/2022]
Abstract
Amphibians such as Xenopus tropicalis exhibit a remarkable capacity for tissue regeneration after traumatic injury. Although transforming growth factor-β (TGF-β) receptor signaling is known to be essential for tissue regeneration in fish and amphibians, the role of TGF-β ligands in this process is not well understood. Here, we show that inhibition of TGF-β1 function prevents tail regeneration in Xenopus tropicalis tadpoles. We found that expression of tgfb1 is present before tail amputation and is sustained throughout the regeneration process. CRISPR-mediated knock-out (KO) of tgfb1 retards tail regeneration; the phenotype of tgfb1 KO tadpoles can be rescued by injection of tgfb1 mRNA. Cell proliferation, a critical event for the success of tissue regeneration, is downregulated in tgfb1 KO tadpoles. In addition, tgfb1 KO reduces the expression of phosphorylated Smad2/3 (pSmad2/3) which is important for TGF-β signal-mediated cell proliferation. Collectively, our results show that TGF-β1 regulates cell proliferation through the activation of Smad2/3. We therefore propose that TGF-β1 plays a critical role in TGF-β receptor-dependent tadpole tail regeneration in Xenopus.
Collapse
Affiliation(s)
- Makoto Nakamura
- Amphibian Research Center, Graduate School of Integrated Sciences for Life, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-8526, Japan
| | - Hitoshi Yoshida
- National Xenopus Resource and Eugene Bell Center for Regenerative Biology and Tissue Engineering, Marine Biological Laboratory, Woods Hole, MA 02543, USA
| | - Yuka Moriyama
- Amphibian Research Center, Graduate School of Integrated Sciences for Life, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-8526, Japan
| | - Itsuki Kawakita
- Amphibian Research Center, School of Science, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-8526, Japan
| | - Marcin Wlizla
- National Xenopus Resource and Eugene Bell Center for Regenerative Biology and Tissue Engineering, Marine Biological Laboratory, Woods Hole, MA 02543, USA
| | - Kimiko Takebayashi-Suzuki
- Amphibian Research Center, Graduate School of Integrated Sciences for Life, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-8526, Japan
| | - Marko E Horb
- National Xenopus Resource and Eugene Bell Center for Regenerative Biology and Tissue Engineering, Marine Biological Laboratory, Woods Hole, MA 02543, USA
| | - Atsushi Suzuki
- Amphibian Research Center, Graduate School of Integrated Sciences for Life, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-8526, Japan.
| |
Collapse
|
18
|
Cloutier JK, McMann CL, Oderberg IM, Reddien PW. activin-2 is required for regeneration of polarity on the planarian anterior-posterior axis. PLoS Genet 2021; 17:e1009466. [PMID: 33780442 PMCID: PMC8057570 DOI: 10.1371/journal.pgen.1009466] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 04/20/2021] [Accepted: 03/03/2021] [Indexed: 01/16/2023] Open
Abstract
Planarians are flatworms and can perform whole-body regeneration. This ability involves a mechanism to distinguish between anterior-facing wounds that require head regeneration and posterior-facing wounds that require tail regeneration. How this head-tail regeneration polarity decision is made is studied to identify principles underlying tissue-identity specification in regeneration. We report that inhibition of activin-2, which encodes an Activin-like signaling ligand, resulted in the regeneration of ectopic posterior-facing heads following amputation. During tissue turnover in uninjured planarians, positional information is constitutively expressed in muscle to maintain proper patterning. Positional information includes Wnts expressed in the posterior and Wnt antagonists expressed in the anterior. Upon amputation, several wound-induced genes promote re-establishment of positional information. The head-versus-tail regeneration decision involves preferential wound induction of the Wnt antagonist notum at anterior-facing over posterior-facing wounds. Asymmetric activation of notum represents the earliest known molecular distinction between head and tail regeneration, yet how it occurs is unknown. activin-2 RNAi animals displayed symmetric wound-induced activation of notum at anterior- and posterior-facing wounds, providing a molecular explanation for their ectopic posterior-head phenotype. activin-2 RNAi animals also displayed anterior-posterior (AP) axis splitting, with two heads appearing in anterior blastemas, and various combinations of heads and tails appearing in posterior blastemas. This was associated with ectopic nucleation of anterior poles, which are head-tip muscle cells that facilitate AP and medial-lateral (ML) pattern at posterior-facing wounds. These findings reveal a role for Activin signaling in determining the outcome of AP-axis-patterning events that are specific to regeneration. A central problem in animal regeneration is how animals determine what body part to regenerate. Planarians are flatworms that can regenerate any missing body region, and are studied to identify mechanisms underlying regeneration. At transverse amputation planes, a poorly understood mechanism specifies regeneration of either a head or a tail. This head-versus-tail regeneration decision-making process is referred to as regeneration polarity and has been studied for over a century to identify mechanisms that specify what to regenerate. The gene notum, which encodes a Wnt antagonist, is induced within hours after injury preferentially at anterior-facing wounds, where it specifies head regeneration. We report that Activin signaling is required for regeneration polarity, and the underlying asymmetric activation of notum at anterior- over posterior-facing wounds. We propose that Activin signaling is involved in regeneration-specific responses broadly in the animal kingdom.
Collapse
Affiliation(s)
- Jennifer K. Cloutier
- Whitehead Institute for Biomedical Research, Cambridge, MA, United States of America
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, United States of America
- Howard Hughes Medical Institute, Chevy Chase, MD, United States of America
- Harvard/MIT MD-PhD, Harvard Medical School, Boston, MA, United States of America
| | - Conor L. McMann
- Whitehead Institute for Biomedical Research, Cambridge, MA, United States of America
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, United States of America
- Howard Hughes Medical Institute, Chevy Chase, MD, United States of America
| | - Isaac M. Oderberg
- Whitehead Institute for Biomedical Research, Cambridge, MA, United States of America
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, United States of America
- Howard Hughes Medical Institute, Chevy Chase, MD, United States of America
| | - Peter W. Reddien
- Whitehead Institute for Biomedical Research, Cambridge, MA, United States of America
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, United States of America
- Howard Hughes Medical Institute, Chevy Chase, MD, United States of America
- * E-mail:
| |
Collapse
|
19
|
Daponte V, Tylzanowski P, Forlino A. Appendage Regeneration in Vertebrates: What Makes This Possible? Cells 2021; 10:cells10020242. [PMID: 33513779 PMCID: PMC7911911 DOI: 10.3390/cells10020242] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/18/2021] [Accepted: 01/22/2021] [Indexed: 12/26/2022] Open
Abstract
The ability to regenerate amputated or injured tissues and organs is a fascinating property shared by several invertebrates and, interestingly, some vertebrates. The mechanism of evolutionary loss of regeneration in mammals is not understood, yet from the biomedical and clinical point of view, it would be very beneficial to be able, at least partially, to restore that capability. The current availability of new experimental tools, facilitating the comparative study of models with high regenerative ability, provides a powerful instrument to unveil what is needed for a successful regeneration. The present review provides an updated overview of multiple aspects of appendage regeneration in three vertebrates: lizard, salamander, and zebrafish. The deep investigation of this process points to common mechanisms, including the relevance of Wnt/β-catenin and FGF signaling for the restoration of a functional appendage. We discuss the formation and cellular origin of the blastema and the identification of epigenetic and cellular changes and molecular pathways shared by vertebrates capable of regeneration. Understanding the similarities, being aware of the differences of the processes, during lizard, salamander, and zebrafish regeneration can provide a useful guide for supporting effective regenerative strategies in mammals.
Collapse
Affiliation(s)
- Valentina Daponte
- Biochemistry Unit, Department of Molecular Medicine, University of Pavia, via Taramelli 3/B, 27100 Pavia, Italy;
| | - Przemko Tylzanowski
- Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, University of Leuven, 3000 Leuven, Belgium;
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 20-059 Lublin, Poland
| | - Antonella Forlino
- Biochemistry Unit, Department of Molecular Medicine, University of Pavia, via Taramelli 3/B, 27100 Pavia, Italy;
- Correspondence: ; Tel.: +39-0382-987235
| |
Collapse
|
20
|
Abarca-Buis RF, Mandujano-Tinoco EA, Cabrera-Wrooman A, Krötzsch E. The complexity of TGFβ/activin signaling in regeneration. J Cell Commun Signal 2021; 15:7-23. [PMID: 33481173 DOI: 10.1007/s12079-021-00605-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 01/05/2021] [Indexed: 12/11/2022] Open
Abstract
The role of transforming growth factor β TGFβ/activin signaling in wound repair and regeneration is highly conserved in the animal kingdom. Various studies have shown that TGF-β/activin signaling can either promote or inhibit different aspects of the regeneration process (i.e., proliferation, differentiation, and re-epithelialization). It has been demonstrated in several biological systems that some of the different cellular responses promoted by TGFβ/activin signaling depend on the activation of Smad-dependent or Smad-independent signal transduction pathways. In the context of regeneration and wound healing, it has been shown that the type of R-Smad stimulated determines the different effects that can be obtained. However, neither the possible roles of Smad-independent pathways nor the interaction of the TGFβ/activin pathway with other complex signaling networks involved in the regenerative process has been studied extensively. Here, we review the important aspects concerning the TGFβ/activin signaling pathway in the regeneration process. We discuss data regarding the role of TGF-β/activin in the most common animal regenerative models to demonstrate how this signaling promotes or inhibits regeneration, depending on the cellular context.
Collapse
Affiliation(s)
- René Fernando Abarca-Buis
- Laboratory of Connective Tissue, Centro Nacional de Investigación y Atención de Quemados, Instituto Nacional de Rehabilitación "Luís Guillermo Ibarra Ibarra", Calzada México-Xochimilco No. 289, Col. Arenal de Guadalupe, Tlalpan, 14389, Mexico City, Mexico.
| | - Edna Ayerim Mandujano-Tinoco
- Laboratory of Connective Tissue, Centro Nacional de Investigación y Atención de Quemados, Instituto Nacional de Rehabilitación "Luís Guillermo Ibarra Ibarra", Calzada México-Xochimilco No. 289, Col. Arenal de Guadalupe, Tlalpan, 14389, Mexico City, Mexico
| | - Alejandro Cabrera-Wrooman
- Laboratory of Connective Tissue, Centro Nacional de Investigación y Atención de Quemados, Instituto Nacional de Rehabilitación "Luís Guillermo Ibarra Ibarra", Calzada México-Xochimilco No. 289, Col. Arenal de Guadalupe, Tlalpan, 14389, Mexico City, Mexico
| | - Edgar Krötzsch
- Laboratory of Connective Tissue, Centro Nacional de Investigación y Atención de Quemados, Instituto Nacional de Rehabilitación "Luís Guillermo Ibarra Ibarra", Calzada México-Xochimilco No. 289, Col. Arenal de Guadalupe, Tlalpan, 14389, Mexico City, Mexico
| |
Collapse
|
21
|
Abstract
Tissue or organ regeneration is a complex process with successful outcomes depending on the type of tissue and organism. Upon damage, mammals can only efficiently restore a few tissues including the liver, skin, epithelia of the lung, kidney, and gut. In contrast, lower vertebrates such as zebrafish possess an extraordinary regeneration ability, which restores the normal function of a broad spectrum of tissues including heart, fin, brain, spinal cord, and retina. This regeneration process is either mediated by the proliferation of resident stem cells, or cells that dedifferentiate into a stem cell-like. In recent years, evidence has suggested that the innate immune system can modulate stem cell activity to initiate the regenerative response to damage. This review will explore some of the newer concepts of inflammation in zebrafish regeneration in different tissues. Understanding how inflammation regulates regeneration in zebrafish would provide important clues to improve the therapeutic strategies for repairing injured mammalian tissues that do not have an inherent regenerative capacity.
Collapse
Affiliation(s)
- Maria Iribarne
- Center for Zebrafish Research, Department of Biological Sciences; Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, IN, USA
| |
Collapse
|
22
|
Wang W, Hu CK, Zeng A, Alegre D, Hu D, Gotting K, Ortega Granillo A, Wang Y, Robb S, Schnittker R, Zhang S, Alegre D, Li H, Ross E, Zhang N, Brunet A, Sánchez Alvarado A. Changes in regeneration-responsive enhancers shape regenerative capacities in vertebrates. Science 2020; 369:369/6508/eaaz3090. [PMID: 32883834 DOI: 10.1126/science.aaz3090] [Citation(s) in RCA: 135] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 03/05/2020] [Accepted: 07/07/2020] [Indexed: 12/13/2022]
Abstract
Vertebrates vary in their ability to regenerate, and the genetic mechanisms underlying such disparity remain elusive. Comparative epigenomic profiling and single-cell sequencing of two related teleost fish uncovered species-specific and evolutionarily conserved genomic responses to regeneration. The conserved response revealed several regeneration-responsive enhancers (RREs), including an element upstream to inhibin beta A (inhba), a known effector of vertebrate regeneration. This element activated expression in regenerating transgenic fish, and its genomic deletion perturbed caudal fin regeneration and abrogated cardiac regeneration altogether. The enhancer is present in mammals, shares functionally essential activator protein 1 (AP-1)-binding motifs, and responds to injury, but it cannot rescue regeneration in fish. This work suggests that changes in AP-1-enriched RREs are likely a crucial source of loss of regenerative capacities in vertebrates.
Collapse
Affiliation(s)
- Wei Wang
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA.,Howard Hughes Medical Institute, Kansas City, MO 64110, USA
| | - Chi-Kuo Hu
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - An Zeng
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Dana Alegre
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Deqing Hu
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Kirsten Gotting
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | | | - Yongfu Wang
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Sofia Robb
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | | | - Shasha Zhang
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Dillon Alegre
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Hua Li
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Eric Ross
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA.,Howard Hughes Medical Institute, Kansas City, MO 64110, USA
| | - Ning Zhang
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Anne Brunet
- Department of Genetics, Stanford University, Stanford, CA 94305, USA.,Glenn Laboratories for the Biology of Aging. Stanford University, Stanford, CA 94305, USA
| | - Alejandro Sánchez Alvarado
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA. .,Howard Hughes Medical Institute, Kansas City, MO 64110, USA
| |
Collapse
|
23
|
Lebedeva L, Zhumabayeva B, Gebauer T, Kisselev I, Aitasheva Z. Zebrafish ( Danio rerio) as a Model for Understanding the Process of Caudal Fin Regeneration. Zebrafish 2020; 17:359-372. [PMID: 33259770 DOI: 10.1089/zeb.2020.1926] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
After its introduction for scientific investigation in the 1950s, the cypriniform zebrafish, Danio rerio, has become a valuable model for the study of regenerative processes and mechanisms. Zebrafish exhibit epimorphic regeneration, in which a nondifferentiated cell mass formed after amputation is able to fully regenerate lost tissue such as limbs, heart muscle, brain, retina, and spinal cord. The process of limb regeneration in zebrafish comprises several stages characterized by the activation of specific signaling pathways and gene expression. We review current research on key factors in limb regeneration using zebrafish as a model.
Collapse
Affiliation(s)
- Lina Lebedeva
- Department of Molecular Biology and Genetics, Faculty of Biology and Biotechnology, al-Farabi Kazakh National University, Almaty, The Republic of Kazakhstan
| | - Beibitgul Zhumabayeva
- Department of Molecular Biology and Genetics, Faculty of Biology and Biotechnology, al-Farabi Kazakh National University, Almaty, The Republic of Kazakhstan
| | - Tatyana Gebauer
- South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Institute of Aquaculture and Protection of Waters, Faculty of Fisheries and Protection of Waters, University of South Bohemia in Ceske Budejovice, České Budějovice, Czech Republic
| | - Ilya Kisselev
- Institute of General Genetics and Cytology, Almaty, The Republic of Kazakhstan
| | - Zaure Aitasheva
- Department of Molecular Biology and Genetics, Faculty of Biology and Biotechnology, al-Farabi Kazakh National University, Almaty, The Republic of Kazakhstan
| |
Collapse
|
24
|
Maciuszek M, Pijanowski L, Pekala-Safinska A, Kemenade BMLVV, Chadzinska M. 17β-Estradiol affects the innate immune response in common carp. FISH PHYSIOLOGY AND BIOCHEMISTRY 2020; 46:1775-1794. [PMID: 32519008 PMCID: PMC7427712 DOI: 10.1007/s10695-020-00827-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 05/21/2020] [Indexed: 05/05/2023]
Abstract
Inflammation is the evolutionary conserved immune response to harmful stimuli such as pathogens or damaged cells. This multistep process acts by removing injurious stimuli and initiating the healing process. Therefore, it must be tightly regulated by cytokines, chemokines, and enzymes, as well as neuroendocrine mediators. In the present work, we studied the immunoregulatory properties of 17β-estradiol (E2) in common carp. We determined the in vitro effects of E2 on the activity/polarization of macrophages and the in vivo effects during Aeromonas salmonicida-induced inflammation. In vitro, E2 reduced the lipopolysaccharide (LPS)-stimulated expression of pro- and anti-inflammatory mediator genes but did not change the gene expression of the estrogen receptors and of aromatase CYP19. In contrast, in vivo in the head kidney of A. salmonicida-infected fish, E2-treated feeding induced an upregulation of gene expression of pro-inflammatory (il-12p35 and cxcb2) and anti-inflammatory (arginase 1, arginase 2, il-10, and mmp9) mediators. Moreover, in infected fish fed with E2-treated food, a higher gene expression of the estrogen receptors and of the aromatase CYP19 was found. Our results demonstrate that estrogens can modulate the carp innate immune response, though the in vitro and in vivo effects of this hormone are contrasting. This implies that estradiol not only induces a direct effect on macrophages but rather exerts immunomodulatory actions through indirect mechanisms involving other cellular targets.
Collapse
Affiliation(s)
- Magdalena Maciuszek
- Department of Evolutionary Immunology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Gronostajowa 9, PL30-387, Krakow, Poland
| | - Lukasz Pijanowski
- Department of Evolutionary Immunology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Gronostajowa 9, PL30-387, Krakow, Poland
| | - Agnieszka Pekala-Safinska
- Department of Fish Diseases, National Veterinary Research Institute, Partyzantow Avenue 57, PL24-100, Pulawy, Poland
| | | | - Magdalena Chadzinska
- Department of Evolutionary Immunology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Gronostajowa 9, PL30-387, Krakow, Poland.
| |
Collapse
|
25
|
Wound Repair, Scar Formation, and Cancer: Converging on Activin. Trends Mol Med 2020; 26:1107-1117. [PMID: 32878730 DOI: 10.1016/j.molmed.2020.07.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/24/2020] [Accepted: 07/28/2020] [Indexed: 02/07/2023]
Abstract
Wound repair is a highly regulated process that requires the interaction of various cell types. It has been shown that cancers use the mechanisms of wound healing to promote their own growth. Therefore, it is of importance to identify common regulators of wound repair and tumor formation and to unravel their functions and mechanisms of action. An exciting example is activin, which acts on multiple cell types in wounds and tumors, thereby promoting healing, but also scar formation and tumorigenesis. Here, we summarize current knowledge on the role of activin in these processes and highlight the therapeutic potential of activin or activin antagonists for the treatment of impaired healing or excessive scarring and cancer, respectively.
Collapse
|
26
|
Midkine-a functions as a universal regulator of proliferation during epimorphic regeneration in adult zebrafish. PLoS One 2020; 15:e0232308. [PMID: 32530962 PMCID: PMC7292404 DOI: 10.1371/journal.pone.0232308] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 05/28/2020] [Indexed: 12/20/2022] Open
Abstract
Zebrafish have the ability to regenerate damaged cells and tissues by activating quiescent stem and progenitor cells or reprogramming differentiated cells into regeneration-competent precursors. Proliferation among the cells that will functionally restore injured tissues is a fundamental biological process underlying regeneration. Midkine-a is a cytokine growth factor, whose expression is strongly induced by injury in a variety of tissues across a range of vertebrate classes. Using a zebrafish Midkine-a loss of function mutant, we evaluated regeneration of caudal fin, extraocular muscle and retinal neurons to investigate the function of Midkine-a during epimorphic regeneration. In wildtype zebrafish, injury among these tissues induces robust proliferation and rapid regeneration. In Midkine-a mutants, the initial proliferation in each of these tissues is significantly diminished or absent. Regeneration of the caudal fin and extraocular muscle is delayed; regeneration of the retina is nearly completely absent. These data demonstrate that Midkine-a is universally required in the signaling pathways that convert tissue injury into the initial burst of cell proliferation. Further, these data highlight differences in the molecular mechanisms that regulate epimorphic regeneration in zebrafish.
Collapse
|
27
|
Lan Y, Pan H, Li C, Banks KM, Sam J, Ding B, Elemento O, Goll MG, Evans T. TETs Regulate Proepicardial Cell Migration through Extracellular Matrix Organization during Zebrafish Cardiogenesis. Cell Rep 2020; 26:720-732.e4. [PMID: 30650362 PMCID: PMC6366638 DOI: 10.1016/j.celrep.2018.12.076] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 10/30/2018] [Accepted: 12/18/2018] [Indexed: 12/22/2022] Open
Abstract
Ten-eleven translocation (Tet) enzymes (Tet1/2/3) mediate 5-methylcytosine (5mC) hydroxylation, which can facilitate DNA demethylation and thereby impact gene expression. Studied mostly for how mutant isoforms impact cancer, the normal roles for Tet enzymes during organogenesis are largely unknown. By analyzing compound mutant zebrafish, we discovered a requirement for Tet2/3 activity in the embryonic heart for recruitment of epicardial progenitors, associated with development of the atrial-ventricular canal (AVC). Through a combination of methylation, hydroxymethylation, and transcript profiling, the genes encoding the activin A subunit Inhbaa (in endocardium) and Sox9b (in myocardium) were implicated as demethylation targets of Tet2/3 and critical for organization of AVC-localized extracellular matrix (ECM), facilitating migration of epicardial progenitors onto the developing heart tube. This study elucidates essential DNA demethylation modifications that govern gene expression changes during cardiac development with striking temporal and lineage specificities, highlighting complex interactions in multiple cell populations during development of the vertebrate heart. Lan et al. show that zebrafish larvae mutant for tet2 and tet3 fail to demethylate genes encoding Inhbaa (in endocardium) and Sox9b (in myocardium), leading to defects in ECM needed to form valves and to recruit epicardial progenitors onto the heart tube.
Collapse
Affiliation(s)
- Yahui Lan
- Department of Surgery, Weill Cornell Medical College, New York, NY 10065, USA
| | - Heng Pan
- Department of Physiology and Biophysics, Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Cheng Li
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Program in Biochemistry and Structural Biology, Cell and Developmental Biology, and Molecular Biology, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY 10065, USA
| | - Kelly M Banks
- Department of Surgery, Weill Cornell Medical College, New York, NY 10065, USA
| | - Jessica Sam
- Department of Surgery, Weill Cornell Medical College, New York, NY 10065, USA
| | - Bo Ding
- Bonacept, LLC, 7699 Palmilla Drive, Apt. 3312, San Diego, CA 92122, USA
| | - Olivier Elemento
- Department of Physiology and Biophysics, Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Mary G Goll
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Todd Evans
- Department of Surgery, Weill Cornell Medical College, New York, NY 10065, USA.
| |
Collapse
|
28
|
Conedera FM, Quintela Pousa AM, Presby DM, Mercader N, Enzmann V, Tschopp M. Diverse Signaling by TGFβ Isoforms in Response to Focal Injury is Associated with Either Retinal Regeneration or Reactive Gliosis. Cell Mol Neurobiol 2020; 41:43-62. [PMID: 32219603 PMCID: PMC7811507 DOI: 10.1007/s10571-020-00830-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 03/11/2020] [Indexed: 12/13/2022]
Abstract
Müller cells may have stem cell-like capability as they regenerate photoreceptor loss upon injury in some vertebrates, but not in mammals. Indeed, mammalian Müller cells undergo major cellular and molecular changes summarized as reactive gliosis. Transforming growth factor beta (TGFβ) isoforms are multifunctional cytokines that play a central role, both in wound healing and in tissue repair. Here, we studied the role of TGFβ isoforms and their signaling pathways in response to injury induction during tissue regeneration in zebrafish and scar formation in mouse. Our transcriptome analysis showed a different activation of canonical and non-canonical signaling pathways and how they shaped the injury response. In particular, TGFβ3 promotes retinal regeneration via Smad-dependent canonical pathway upon regulation of junb gene family and mycb in zebrafish Müller cells. However, in mice, TGFβ1 and TGFβ2 evoke the p38MAPK signaling pathway. The activation of this non-canonical pathway leads to retinal gliosis. Thus, the regenerative versus reparative effect of the TGFβ pathway observed may rely on the activation of different signaling cascades. This provides one explanation of the different injury response in zebrafish and mouse retina.
Collapse
Affiliation(s)
- Federica Maria Conedera
- Department of Ophthalmology, University Hospital of Bern, University of Bern, Bern, Switzerland.,Department of BioMedical Research, University of Bern, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Ana Maria Quintela Pousa
- Department of Ophthalmology, University Hospital of Bern, University of Bern, Bern, Switzerland.,Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - David Mikal Presby
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Nadia Mercader
- Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Volker Enzmann
- Department of Ophthalmology, University Hospital of Bern, University of Bern, Bern, Switzerland. .,Department of BioMedical Research, University of Bern, Bern, Switzerland.
| | - Markus Tschopp
- Department of Ophthalmology, University Hospital of Bern, University of Bern, Bern, Switzerland. .,Department of Ophthalmology, Cantonal Hospital Aarau, Aarau, Switzerland.
| |
Collapse
|
29
|
Bensimon-Brito A, Ramkumar S, Boezio GLM, Guenther S, Kuenne C, Helker CSM, Sánchez-Iranzo H, Iloska D, Piesker J, Pullamsetti S, Mercader N, Beis D, Stainier DYR. TGF-β Signaling Promotes Tissue Formation during Cardiac Valve Regeneration in Adult Zebrafish. Dev Cell 2019; 52:9-20.e7. [PMID: 31786069 DOI: 10.1016/j.devcel.2019.10.027] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 09/17/2019] [Accepted: 10/28/2019] [Indexed: 12/14/2022]
Abstract
Cardiac valve disease can lead to severe cardiac dysfunction and is thus a frequent cause of morbidity and mortality. Its main treatment is valve replacement, which is currently greatly limited by the poor recellularization and tissue formation potential of the implanted valves. As we still lack suitable animal models to identify modulators of these processes, here we used adult zebrafish and found that, upon valve decellularization, they initiate a rapid regenerative program that leads to the formation of new functional valves. After injury, endothelial and kidney marrow-derived cells undergo cell cycle re-entry and differentiate into new extracellular matrix-secreting valve cells. The TGF-β signaling pathway promotes the regenerative process by enhancing progenitor cell proliferation as well as valve cell differentiation. These findings reveal a key role for TGF-β signaling in cardiac valve regeneration and establish the zebrafish as a model to identify and test factors promoting cardiac valve recellularization and growth.
Collapse
Affiliation(s)
- Anabela Bensimon-Brito
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany.
| | - Srinath Ramkumar
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
| | - Giulia L M Boezio
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
| | - Stefan Guenther
- Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
| | - Carsten Kuenne
- Bioinformatics Core Unit, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
| | - Christian S M Helker
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
| | - Héctor Sánchez-Iranzo
- Cell Biology and Biophysics Research Unit, EMBL Heidelberg, Heidelberg 69117, Germany
| | - Dijana Iloska
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
| | - Janett Piesker
- Scientific Service Group Microscopy, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
| | - Soni Pullamsetti
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
| | - Nadia Mercader
- Institute of Anatomy, University of Bern, Bern 3012, Switzerland; Centro Nacional de Investigaciones Cardiovasculares, CNIC, Madrid 28049, Spain
| | - Dimitris Beis
- Developmental Biology, Biomedical Research Foundation of the Academy of Athens, Athens 11527, Greece
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany.
| |
Collapse
|
30
|
König D, Dagenais P, Senk A, Djonov V, Aegerter CM, Jaźwińska A. Distribution and Restoration of Serotonin-Immunoreactive Paraneuronal Cells During Caudal Fin Regeneration in Zebrafish. Front Mol Neurosci 2019; 12:227. [PMID: 31616250 PMCID: PMC6763699 DOI: 10.3389/fnmol.2019.00227] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 09/04/2019] [Indexed: 12/22/2022] Open
Abstract
Aquatic vertebrates possess diverse types of sensory cells in their skin to detect stimuli in the water. In the adult zebrafish, a common model organism, the presence of such cells in fins has only rarely been studied. Here, we identified scattered serotonin (5-HT)-positive cells in the epidermis of the caudal fin. These cells were distinct from keratinocytes as revealed by their low immunoreactivity for cytokeratin and desmosome markers. Instead, they were detected by Calretinin (Calbindin-2) and Synaptic vesicle glycoprotein 2 (SV2) antibodies, indicating a calcium-regulated neurosecretory activity. Consistently, electron microscopy revealed abundant secretory organelles in desmosome-negative cells in the fin epidermis. Based on the markers, 5-HT, Calretinin and SV2, we referred to these cells as HCS-cells. We found that HCS-cells were spread throughout the entire caudal fin at an average density of 140 cells per mm2 on each fin surface. These cells were strongly enriched at ray bifurcations in wild type fins, as well as in elongated fins of another longfin mutant fish. To determine whether hydrodynamics play a role in the distribution of HCS-cells, we used an interdisciplinary approach and performed kinematic analysis. Measurements of particle velocity with a fin model revealed differences in fluid velocities between bifurcated rods and adjacent non-bifurcated regions. Therefore the accumulation of HCS-cells near bone bifurcations may be a biological adaptation for sensing of water parameters. The significance of this HCS-cell pattern is reinforced by the fact, that it is reestablished in the regenerated fin after amputation. Regeneration of HCS-cells was not impaired by the chemical inhibition of serotonin synthesis, suggesting that this neurotransmitter is not essential for the restorative process. In conclusion, our study identified a specific population of solitary paraneurons in the zebrafish fin, whose distribution correlates with fluid dynamics.
Collapse
Affiliation(s)
- Désirée König
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Paule Dagenais
- Physik-Institut, University of Zurich, Zurich, Switzerland
| | - Anita Senk
- Institute of Anatomy, University of Bern, Bern, Switzerland
| | | | | | - Anna Jaźwińska
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
31
|
Flanagan-Steet H, Christian C, Lu PN, Aarnio-Peterson M, Sanman L, Archer-Hartmann S, Azadi P, Bogyo M, Steet RA. TGF-ß Regulates Cathepsin Activation during Normal and Pathogenic Development. Cell Rep 2019. [PMID: 29539424 PMCID: PMC6247414 DOI: 10.1016/j.celrep.2018.02.066] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Cysteine cathepsins play roles during development and disease beyond their function in lysosomal protein turnover. Here, we leverage a fluorescent activity-based probe (ABP), BMV109, to track cysteine cathepsins in normal and diseased zebrafish embryos. Using this probe in a model of mucolipidosis II, we show that loss of carbohydrate-dependent lysosomal sorting alters the activity of several cathepsin proteases. The data support a pathogenic mechanism where TGF-β signals enhance the proteolytic processing of pro-Ctsk by modulating the expression of chondroitin 4-sulfate (C4-S). In MLII, elevated C4-S corresponds with TGF-β-mediated increases in chst11 expression. Inhibiting chst11 impairs the proteolytic activation of Ctsk and alleviates the MLII phenotypes. These findings uncover a regulatory loop between TGF-β signaling and Ctsk activation that is altered in the context of lysosomal disease. This work highlights the power of ABPs to identify mechanisms underlying pathogenic development in living animals. Chondroitin sulfate is a known regulator of cathepsin protease activity. Flanagan-Steet et al. identify a positive feedback mechanism whereby cathepsins secreted from chondrocytes upon loss of lysosomal targeting activate TGF-β signaling in developing cartilage. This increased signaling, in turn, stimulates chondroitin-4 sulfation and enhances cathepsin activity.
Collapse
Affiliation(s)
| | - Courtney Christian
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30606, USA
| | - Po-Nien Lu
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30606, USA
| | | | - Laura Sanman
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305-5324, USA
| | | | - Parastoo Azadi
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30606, USA
| | - Matthew Bogyo
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305-5324, USA
| | - Richard A Steet
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30606, USA
| |
Collapse
|
32
|
Garland MA, Sengupta S, Mathew LK, Truong L, de Jong E, Piersma AH, La Du J, Tanguay RL. Glucocorticoid receptor-dependent induction of cripto-1 ( one-eyed pinhead) inhibits zebrafish caudal fin regeneration. Toxicol Rep 2019; 6:529-537. [PMID: 31249786 PMCID: PMC6584771 DOI: 10.1016/j.toxrep.2019.05.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 05/27/2019] [Accepted: 05/28/2019] [Indexed: 12/15/2022] Open
Abstract
We previously used a chemical genetics approach with the larval zebrafish to identify small molecule inhibitors of tissue regeneration. This led to the discovery that glucocorticoids (GC) block early stages of tissue regeneration by the inappropriate activation of the glucocorticoid receptor (GR). We performed a microarray analysis to identify the changes in gene expression associated with beclomethasone dipropionate (BDP) exposure during epimorphic fin regeneration. Oncofetal cripto-1 showed > eight-fold increased expression in BDP-treated regenerates. We hypothesized that the mis-expression of cripto-1 was essential for BDP to block regeneration. Expression of cripto-1 was not elevated in GR morphants in the presence of BDP indicating that cripto-1 induction was GR-dependent. Partial translational suppression of Cripto-1 in the presence of BDP restored tissue regeneration. Retinoic acid exposure prevented increased cripto-1 expression and permitted regeneration in the presence of BDP. We demonstrated that BDP exposure increased cripto-1 expression in mouse embryonic stem cells and that regulation of cripto-1 by GCs is conserved in mammals.
Collapse
Key Words
- AEC, apical epithelial cap
- BDP, beclomethasone dipropionate
- Beclomethasone dipropionate
- Cripto-1
- DMSO, dimethyl sulfoxide
- EB, embryoid body
- ECM, extracellular matrix
- EMT, epithelial-to-mesenchymal transition
- ERK, extracellular signal-regulated kinase
- Epimorphic regeneration
- FGF, fibroblast growth factor
- GC, glucocorticoid
- GR, glucocorticoid receptor
- Glucocorticoids
- ISH, in situ hybridization
- MIAME, Minimum Information About a Microarray Experiment
- MO, morpholino oligonucleotide
- One-eyed pinhead
- RA, retinoic acid
- SEM, standard error of the mean
- TGF-β, transforming growth factor beta
- Zebrafish
- dpa, days post-amputation
- dpf, days post-fertilization
- eSC, embryonic stem cell
- hpa, hours post-amputation
- hpf, hours post-fertilization
- mLIF, murine leukemia inhibitory factor
- qRT-PCR, quantitative reverse transcription polymerase chain reaction
- zf, zebrafish
Collapse
Affiliation(s)
| | - Sumitra Sengupta
- Department of Environmental and Molecular Toxicology, United States
| | - Lijoy K Mathew
- Department of Environmental and Molecular Toxicology, United States
| | - Lisa Truong
- Department of Environmental and Molecular Toxicology, United States
| | - Esther de Jong
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, the Netherlands.,National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Aldert H Piersma
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, the Netherlands.,National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Jane La Du
- Department of Environmental and Molecular Toxicology, United States
| | - Robert L Tanguay
- Department of Environmental and Molecular Toxicology, United States
| |
Collapse
|
33
|
Paredes LC, Olsen Saraiva Camara N, Braga TT. Understanding the Metabolic Profile of Macrophages During the Regenerative Process in Zebrafish. Front Physiol 2019; 10:617. [PMID: 31178754 PMCID: PMC6543010 DOI: 10.3389/fphys.2019.00617] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 05/01/2019] [Indexed: 12/14/2022] Open
Abstract
In contrast to mammals, lower vertebrates, including zebrafish (Danio rerio), have the ability to regenerate damaged or lost tissues, such as the caudal fin, which makes them an ideal model for tissue and organ regeneration studies. Since several diseases involve the process of transition between fibrosis and tissue regeneration, it is necessary to attain a better understanding of these processes. It is known that the cells of the immune system, especially macrophages, play essential roles in regeneration by participating in the removal of cellular debris, release of pro- and anti-inflammatory factors, remodeling of components of the extracellular matrix and alteration of oxidative patterns during proliferation and angiogenesis. Immune cells undergo phenotypical and functional alterations throughout the healing process due to growth factors and cytokines that are produced in the tissue microenvironment. However, some aspects of the molecular mechanisms through which macrophages orchestrate the formation and regeneration of the blastema remain unclear. In the present review, we outline how macrophages orchestrate the regenerative process in zebrafish and give special attention to the redox balance in the context of tail regeneration.
Collapse
Affiliation(s)
| | - Niels Olsen Saraiva Camara
- Department of Immunology, Institute of Biomedical Sciences IV, University of São Paulo, São Paulo, Brazil.,Nephrology Division, Federal University of São Paulo, São Paulo, Brazil.,Renal Pathophysiology Laboratory, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | | |
Collapse
|
34
|
König D, Jaźwińska A. Zebrafish fin regeneration involves transient serotonin synthesis. Wound Repair Regen 2019; 27:375-385. [DOI: 10.1111/wrr.12719] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 03/13/2019] [Accepted: 04/02/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Désirée König
- Department of BiologyUniversity of Fribourg Chemin du Musée 10, 1700, Fribourg Switzerland
| | - Anna Jaźwińska
- Department of BiologyUniversity of Fribourg Chemin du Musée 10, 1700, Fribourg Switzerland
| |
Collapse
|
35
|
Nourian Dehkordi A, Mirahmadi Babaheydari F, Chehelgerdi M, Raeisi Dehkordi S. Skin tissue engineering: wound healing based on stem-cell-based therapeutic strategies. Stem Cell Res Ther 2019; 10:111. [PMID: 30922387 PMCID: PMC6440165 DOI: 10.1186/s13287-019-1212-2] [Citation(s) in RCA: 305] [Impact Index Per Article: 50.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Normal wound healing is a dynamic and complex multiple phase process involving coordinated interactions between growth factors, cytokines, chemokines, and various cells. Any failure in these phases may lead wounds to become chronic and have abnormal scar formation. Chronic wounds affect patients' quality of life, since they require repetitive treatments and incur considerable medical costs. Thus, much effort has been focused on developing novel therapeutic approaches for wound treatment. Stem-cell-based therapeutic strategies have been proposed to treat these wounds. They have shown considerable potential for improving the rate and quality of wound healing and regenerating the skin. However, there are many challenges for using stem cells in skin regeneration. In this review, we present some sets of the data published on using embryonic stem cells, induced pluripotent stem cells, and adult stem cells in healing wounds. Additionally, we will discuss the different angles whereby these cells can contribute to their unique features and show the current drawbacks.
Collapse
Affiliation(s)
- Azar Nourian Dehkordi
- Department of Stem Cell and Regenerative Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Fatemeh Mirahmadi Babaheydari
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mohammad Chehelgerdi
- Biotechnology Research Center, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | | |
Collapse
|
36
|
Liu Y, Wang C, Su G. Cellular Signaling in Müller Glia: Progenitor Cells for Regenerative and Neuroprotective Responses in Pharmacological Models of Retinal Degeneration. J Ophthalmol 2019; 2019:5743109. [PMID: 31016037 PMCID: PMC6444254 DOI: 10.1155/2019/5743109] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 02/28/2019] [Indexed: 12/13/2022] Open
Abstract
Retinal degenerative diseases are a leading cause of visual impairment or blindness. There are many therapies for delaying the progression of vision loss but no curative strategies currently. Stimulating intrinsic neuronal regeneration is a potential approach to therapy in retinal degenerative diseases. In contrast to stem cells, as embryonic/pluripotent stem cell-derived retinal progenitor cell or mesenchymal stem cells, Müller glia provided an endogenous cellular source for regenerative therapy in the retina. Müller glia are a major component of the retina and considerable evidence suggested these cells can be induced to produce the lost neurons in several species. Understanding the specific characteristic of Müller glia to generate lost neurons will inspire an attractive and alternative therapeutic strategy for treating visual impairment with regenerative research. This review briefly provides the different signal transduction mechanisms which are underlying Müller cell-mediated neuroprotection and neuron regeneration and discusses recent advances about regeneration from Müller glia-derived progenitors.
Collapse
Affiliation(s)
- Yang Liu
- Eye Center, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun, Jilin 130021, China
| | - Chenguang Wang
- Eye Center, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun, Jilin 130021, China
| | - Guanfang Su
- Eye Center, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun, Jilin 130021, China
| |
Collapse
|
37
|
Sun L, Gu L, Tan H, Liu P, Gao G, Tian L, Chen H, Lu T, Qian H, Fu Z, Pan X. Effects of 17α‑ethinylestradiol on caudal fin regeneration in zebrafish larvae. THE SCIENCE OF THE TOTAL ENVIRONMENT 2019; 653:10-22. [PMID: 30390549 DOI: 10.1016/j.scitotenv.2018.10.275] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 10/19/2018] [Accepted: 10/19/2018] [Indexed: 06/08/2023]
Abstract
The ability to restore tissue function and morphology after injury is a key advantage of many fish for a greater chance of survival. The tissue regeneration process is regulated by multiple pathways, and it can therefore be hypothesized that environmental contaminants targeting components of these signaling pathways, may disrupt the fish's capability to repair or regenerate. This could lead to higher mortality and eventually even to a decline in populations. In this study, the effects of 17α‑ethinylestradiol (EE2), a synthetic estrogen, were assessed on the regenerative capacity of larval zebrafish. Zebrafish aged 2 hour post fertilization (hpf) were exposed to 1, 10, or 100 ng/L EE2, and the caudal fins were amputated at 72 hpf. It was found that EE2 exposure significantly inhibited fin regeneration and changed locomotor behavior. The transcription levels for most of the genes involved in the signaling networks regulating the fin regeneration, such as axin2, fgfr1, bmp2b and igf2b, were down-regulated in the amputated fish in response to EE2 exposure, which was in contrast to their increased patterns in the vehicle-exposed control fish. Additionally, the mRNA levels of several immune-related genes, such as il-1β, il-6, il-10 and nf-κb2, were significantly decreased after EE2 exposure, accompanied by a lower density of neutrophils migrated into the wound site. In conclusion, the present study indicated for the first time that estrogenic endocrine disrupting chemicals (EEDCs) could inhibit the regenerative capacity of zebrafish, and this effect was speculated to be mediated through the alteration in regeneration-related signaling pathways and immune competence. This work expands our knowledge of the potential effects of EEDCs on injured aquatic organisms, and highlights the ecotoxicological significance of relationships between regenerative process and endocrine system. This study also implies the potential application of fin regeneration assay for assessing immunotoxicity in ecotoxicological risk assessment.
Collapse
Affiliation(s)
- Liwei Sun
- College of Environment, Zhejiang University of Technology, Hangzhou 310032, PR China; Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310032, PR China
| | - Linqi Gu
- College of Environment, Zhejiang University of Technology, Hangzhou 310032, PR China
| | - Hana Tan
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, PR China
| | - Pan Liu
- College of Environment, Zhejiang University of Technology, Hangzhou 310032, PR China
| | - Gan Gao
- College of Environment, Zhejiang University of Technology, Hangzhou 310032, PR China
| | - Li Tian
- College of Environment, Zhejiang University of Technology, Hangzhou 310032, PR China
| | - Hui Chen
- Department of Food Science and Technology, Ocean College, Zhejiang University of Technology, Hangzhou 310032, PR China
| | - Tao Lu
- College of Environment, Zhejiang University of Technology, Hangzhou 310032, PR China
| | - Haifeng Qian
- College of Environment, Zhejiang University of Technology, Hangzhou 310032, PR China
| | - Zhengwei Fu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, PR China
| | - Xiangliang Pan
- College of Environment, Zhejiang University of Technology, Hangzhou 310032, PR China.
| |
Collapse
|
38
|
Morales RA, Allende ML. Peripheral Macrophages Promote Tissue Regeneration in Zebrafish by Fine-Tuning the Inflammatory Response. Front Immunol 2019; 10:253. [PMID: 30891030 PMCID: PMC6413720 DOI: 10.3389/fimmu.2019.00253] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 01/29/2019] [Indexed: 12/14/2022] Open
Abstract
The role of macrophages during regeneration in zebrafish has been well-documented. Nevertheless, new evidence indicates that zebrafish macrophages are a heterogeneous population of cells, and that they can play different roles during immune responses and in tissue restoration after damage and infection. In this work, we first aimed to classify zebrafish macrophages according to their distribution in the larva during homeostasis and after tissue damage, distinguishing peripheral, and hematopoietic tissue resident macrophages. We discovered differences between the migratory behavior of these two macrophage populations both before and after tissue damage, triggered by the amputation of the tail fin. Further, we found a specific role for peripheral tissue-resident macrophages, and we propose that these cells contribute to tail fin regeneration by down-regulating inflammatory mediators such as interleukin-1b (il1b) and by diminishing reactive oxygen species (ROS) in the damage site. Our work suggests that specific macrophage populations recruited after tissue damage in zebrafish larvae can display different functions during both inflammation and tissue regeneration.
Collapse
Affiliation(s)
- Rodrigo A Morales
- FONDAP Center for Genome Regulation, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Miguel L Allende
- FONDAP Center for Genome Regulation, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| |
Collapse
|
39
|
Shinji J, Gotoh H, Miyanishi H, Lavine MD, Lavine LC. The activin signaling transcription factor Smox is an essential regulator of appendage size during regeneration after autotomy in the crayfish. Evol Dev 2018; 21:44-55. [DOI: 10.1111/ede.12277] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Junpei Shinji
- Department of Entomology; Washington State University; Pullman Washington
| | - Hiroki Gotoh
- Graduate School of Bioagricultural Sciences; Nagoya University, Furo-cho, Chikusa-ku; Nagoya Japan
| | - Hiroshi Miyanishi
- Faculty of Agriculture; University of Miyazaki, Gakuen-kibanadai-nishi; Miyazaki Japan
| | - Mark D. Lavine
- Department of Entomology; Washington State University; Pullman Washington
| | | |
Collapse
|
40
|
LaBonty M, Pray N, Yelick PC. Injury of Adult Zebrafish Expressing Acvr1l Q204D Does Not Result in Heterotopic Ossification. Zebrafish 2018; 15:536-545. [PMID: 30183553 DOI: 10.1089/zeb.2018.1611] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Fibrodysplasia Ossificans Progressiva (FOP) is a rare, autosomal dominant genetic disorder in humans characterized by the gradual ossification of fibrous tissues, including skeletal muscle, tendons, and ligaments. In humans, mutations in the Type I BMP/TGFβ family member receptor gene, ACVR1, are associated with FOP. Zebrafish acvr1l, previously known as alk8, is the functional ortholog of human ACVR1. We previously created and characterized the first adult zebrafish model for FOP by generating animals harboring heat shock-inducible mCherry-tagged constitutively active Acvr1l (Q204D). Since injury is a known trigger for heterotopic ossification (HO) development in human FOP patients, in this study, we investigated several injury models in Acvr1lQ204D-expressing zebrafish and the subsequent formation of HO. We performed studies of Activin A injection, cardiotoxin (CTX) injection, and caudal fin clip injury. We found that none of these methods resulted in HO formation at the site of injury. However, some of the cardiotoxin-injected and caudal fin-clipped animals did exhibit HO at distant sites, including the body cavity and along the spine. We describe these results in the context of new and exciting reports on FOP, and discuss future studies to better understand the etiology and progression of this disease.
Collapse
Affiliation(s)
- Melissa LaBonty
- 1 Program in Cell, Molecular, and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine , Boston, Massachusetts.,2 Division of Craniofacial and Molecular Genetics, Department of Orthodontics, Tufts University School of Dental Medicine , Boston, Massachusetts
| | - Nicholas Pray
- 2 Division of Craniofacial and Molecular Genetics, Department of Orthodontics, Tufts University School of Dental Medicine , Boston, Massachusetts
| | - Pamela C Yelick
- 1 Program in Cell, Molecular, and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine , Boston, Massachusetts.,2 Division of Craniofacial and Molecular Genetics, Department of Orthodontics, Tufts University School of Dental Medicine , Boston, Massachusetts
| |
Collapse
|
41
|
Subramaniam N, Petrik JJ, Vickaryous MK. VEGF, FGF-2 and TGFβ expression in the normal and regenerating epidermis of geckos: implications for epidermal homeostasis and wound healing in reptiles. J Anat 2018; 232:768-782. [PMID: 29417581 PMCID: PMC5879961 DOI: 10.1111/joa.12784] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/05/2018] [Indexed: 01/17/2023] Open
Abstract
The skin is a bilayered organ that serves as a key barrier between an organism and its environment. In addition to protecting against microbial invasion, physical trauma and environmental damage, skin participates in maintaining homeostasis. Skin is also capable of spontaneous self-repair following injury. These functions are mediated by numerous pleiotrophic growth factors, including members of the vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF), and transforming growth factor β (TGFβ) families. Although growth factor expression has been well documented in mammals, particularly during wound healing, for groups such as reptiles less is known. Here, we investigate the spatio-temporal pattern of expression of multiple growth factors in normal skin and following a full-thickness cutaneous injury in the representative lizard Eublepharis macularius, the leopard gecko. Unlike mammals, leopard geckos can heal cutaneous wounds without scarring. We demonstrate that before, during and after injury, keratinocytes of the epidermis express a diverse panel of growth factor ligands and receptors, including: VEGF, VEGFR1, VEGFR2, and phosphorylated VEGFR2; FGF-2 and FGFR1; and phosphorylated SMAD2, TGFβ1, and activin βA. Unexpectedly, only the tyrosine kinase receptors VEGFR1 and FGFR1 were dynamically expressed, and only during the earliest phases of re-epithelization; otherwise all the proteins of interest were constitutively present. We propose that the ubiquitous pattern of growth factor expression by keratinocytes is associated with various roles during tissue homeostasis, including protection against ultraviolet photodamage and coordinated body-wide skin shedding.
Collapse
Affiliation(s)
- Noeline Subramaniam
- Department of Biomedical SciencesOntario Veterinary CollegeUniversity of GuelphGuelphONCanada
- Institute of Medical ScienceFaculty of MedicineUniversity of TorontoTorontoONCanada
- Keenan Research Centre in the Li Ka Shing Knowledge InstituteSt. Michael's HospitalDepartment of MedicineUniversity of TorontoTorontoONCanada
| | - James J. Petrik
- Department of Biomedical SciencesOntario Veterinary CollegeUniversity of GuelphGuelphONCanada
| | - Matthew K. Vickaryous
- Department of Biomedical SciencesOntario Veterinary CollegeUniversity of GuelphGuelphONCanada
| |
Collapse
|
42
|
Murawala H, Ranadive I, Patel S, Desai I, Balakrishnan S. Protein expression pattern and analysis of differentially expressed peptides during various stages of tail regeneration in Hemidactylus flaviviridis. Mech Dev 2018; 150:1-9. [DOI: 10.1016/j.mod.2018.02.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 02/01/2018] [Indexed: 12/13/2022]
|
43
|
Omar NN, Rashed RR, El-Hazek RM, El-Sabbagh WA, Rashed ER, El-Ghazaly MA. Platelet-rich plasma-induced feedback inhibition of activin A/follistatin signaling: A mechanism for tumor-low risk skin rejuvenation in irradiated rats. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2018; 180:17-24. [PMID: 29413698 DOI: 10.1016/j.jphotobiol.2018.01.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 01/17/2018] [Accepted: 01/22/2018] [Indexed: 01/14/2023]
Abstract
BACKGROUND Platelet-rich plasma (PRP) is a source of natural growth factors and is emerging as a treatment modality to mitigate radiotherapy- induced adverse effects. Activin A (ACTA) is a member of the transforming growth factor-β (TGF-β) superfamily, which has been shown to modulate the inflammatory response and macrophages polarization between different phenotypes. The aim of this study is to determine the value of PRP in preventing radiation-induced malignancies in light of the cross-talk between PRP and activin A type II receptors (ActR-IIA)/follistatin (FST) signaling pathways where the inflammatory responses at 2 different time points were evaluated. MATERIAL AND METHODS Male albino rats were exposed to radiation and given PRP over the course of 6 days. Rats were sacrificed on day 7 or day 28 post radiation. RESULTS Quantitative real-time reverse transcriptase polymerase chain reaction (QRT-PCR) and western-blot showed that after 7 days of administrating of PRP, ActR-IIA/FST signaling was markedly induced and was associated with the expressions of inflammatory, natural killer and M1 macrophages markers, TNF-α, IL-1β, IFN-γ and IL-12. By contrast, on day 28 of PRP administration, ActR-IIA/FST signaling and the expressions of proinflammatory cytokines were downregulated in parallel with inducing M2 macrophages phenotype as indicated by arginase-1, IL-10 and dectin-1. CONCLUSION The suppression of inflammation and induction of M2 macrophages phenotype in response to PRP administration were found significantly linked to ActR-IIA/FST signaling downregulation. Furthermore, the specific M2 macrophage subtype was found to express dectin-1 receptors which have high affinity for tumor cells thereby is expected to reduce the potential for developing tumors after radiotherapy.
Collapse
Affiliation(s)
- Nesreen Nabil Omar
- Department of Biochemistry, Faculty of Pharmacy, Modern University for Technology and Information, Cairo, Egypt.
| | - Rasha R Rashed
- Department of Drug Radiation Research, National Centre for Radiation Research and Technology, Atomic Energy Authority, Cairo, Egypt
| | - Rania M El-Hazek
- Department of Drug Radiation Research, National Centre for Radiation Research and Technology, Atomic Energy Authority, Cairo, Egypt
| | - Walaa A El-Sabbagh
- Department of Drug Radiation Research, National Centre for Radiation Research and Technology, Atomic Energy Authority, Cairo, Egypt
| | - Engy R Rashed
- Department of Drug Radiation Research, National Centre for Radiation Research and Technology, Atomic Energy Authority, Cairo, Egypt.
| | - Mona A El-Ghazaly
- Department of Drug Radiation Research, National Centre for Radiation Research and Technology, Atomic Energy Authority, Cairo, Egypt
| |
Collapse
|
44
|
Murciano C, Cazorla-Vázquez S, Gutiérrez J, Hijano JA, Ruiz-Sánchez J, Mesa-Almagro L, Martín-Reyes F, Fernández TD, Marí-Beffa M. Widening control of fin inter-rays in zebrafish and inferences about actinopterygian fins. J Anat 2018; 232:783-805. [PMID: 29441573 DOI: 10.1111/joa.12785] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/05/2018] [Indexed: 01/03/2023] Open
Abstract
The amputation of a teleost fin rapidly triggers an intricate maze of hierarchically regulated signalling processes which ultimately reconstruct the diverse tissues of the appendage. Whereas the generation of the fin pattern along the proximodistal axis brings with it several well-known developmental regulators, the mechanisms by which the fin widens along its dorsoventral axis remain poorly understood. Utilizing the zebrafish as an experimental model of fin regeneration and studying more than 1000 actinopterygian species, we hypothesized a connection between specific inter-ray regulatory mechanisms and the morphological variability of inter-ray membranes found in nature. To tackle these issues, both cellular and molecular approaches have been adopted and our results suggest the existence of two distinguishable inter-ray areas in the zebrafish caudal fin, a marginal and a central region. The present work associates the activity of the cell membrane potassium channel kcnk5b, the fibroblast growth factor receptor 1 and the sonic hedgehog pathway to the control of several cell functions involved in inter-ray wound healing or dorsoventral regeneration of the zebrafish caudal fin. This ray-dependent regulation controls cell migration, cell-type patterning and gene expression. The possibility that modifications of these mechanisms are responsible for phenotypic variations found in euteleostean species, is discussed.
Collapse
Affiliation(s)
- Carmen Murciano
- Department of Cell Biology, Genetics and Physiology, Biomedical Research Institute of Málaga (IBIMA), Faculty of Science, University of Málaga, Málaga, Spain
| | - Salvador Cazorla-Vázquez
- Department of Cell Biology, Genetics and Physiology, Biomedical Research Institute of Málaga (IBIMA), Faculty of Science, University of Málaga, Málaga, Spain
| | - Javier Gutiérrez
- Department of Cell Biology, Genetics and Physiology, Biomedical Research Institute of Málaga (IBIMA), Faculty of Science, University of Málaga, Málaga, Spain
| | - Juan Antonio Hijano
- Department of Cell Biology, Genetics and Physiology, Biomedical Research Institute of Málaga (IBIMA), Faculty of Science, University of Málaga, Málaga, Spain
| | - Josefa Ruiz-Sánchez
- Department of Cell Biology, Genetics and Physiology, Biomedical Research Institute of Málaga (IBIMA), Faculty of Science, University of Málaga, Málaga, Spain
| | - Laura Mesa-Almagro
- Department of Cell Biology, Genetics and Physiology, Biomedical Research Institute of Málaga (IBIMA), Faculty of Science, University of Málaga, Málaga, Spain
| | - Flores Martín-Reyes
- Department of Cell Biology, Genetics and Physiology, Biomedical Research Institute of Málaga (IBIMA), Faculty of Science, University of Málaga, Málaga, Spain
| | | | - Manuel Marí-Beffa
- Department of Cell Biology, Genetics and Physiology, Biomedical Research Institute of Málaga (IBIMA), Faculty of Science, University of Málaga, Málaga, Spain.,Networking Research Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Málaga, Spain
| |
Collapse
|
45
|
Li X, Song L, Hu X, Liu C, Shi J, Wang H, Zhan L, Song H. Inhibition of Epithelial-Mesenchymal Transition and Tissue Regeneration by Waterborne Titanium Dioxide Nanoparticles. ACS APPLIED MATERIALS & INTERFACES 2018; 10:3449-3458. [PMID: 29318884 DOI: 10.1021/acsami.7b18986] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Titanium dioxide nanoparticles (TiO2NPs) are among the most widely manufactured nanomaterials with broad applications in food industry, cosmetics, and medicine. Although the toxicity of TiO2NPs at high doses has been extensively explored, the potential health risks of TiO2NPs exposure at nontoxic concentrations remain poorly understood. Epithelial-mesenchymal transition (EMT) plays pivotal roles in a diversity of physiological and pathological processes, including tissue regeneration and cancer metastasis. In this study, we find that the cellular uptake of TiO2NPs inhibits EMT-mediated cell remodeling and cell migration without exhibiting cytotoxicity. Further investigation reveals that TiO2NPs suppress the process of EMT through the blockade of transforming growth factor-β (TGFβ) signaling. Particularly, TiO2NPs interact with the TGFβ receptor TβRI/II complex, induce its lysosomal degradation, and thereby downregulate expression of TGFβ target genes. Moreover, we show that waterborne TiO2NPs do not elicit toxicity in healthy tissues but hamper EMT-mediated wound healing in two animal models. Long-term exposure of TiO2NPs in environmental water and drinking water impede the regeneration of amputated fin in zebrafish and the recovery of intestinal mucosal damage in colitic mice. Our results reveal the previously unknown effects of TiO2NPs during tissue remodeling and repair, which have significant implications in their risk assessment and management.
Collapse
Affiliation(s)
- Xiaojiao Li
- Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences , Shanghai 200031, China
| | - Lele Song
- Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences , Shanghai 200031, China
| | - Xingjie Hu
- School of Public Health, Guangzhou Medical University , Guangdong 511436, China
| | - Chang Liu
- Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences , Shanghai 200031, China
| | - Jiye Shi
- UCB Pharma , Slough SL2 3WE, United Kingdom
| | - Hui Wang
- School of Public Health, Shanghai Jiao Tong University , Shanghai 200025, China
| | - Lixing Zhan
- Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences , Shanghai 200031, China
| | - Haiyun Song
- Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences , Shanghai 200031, China
- School of Public Health, Shanghai Jiao Tong University , Shanghai 200025, China
| |
Collapse
|
46
|
Shibata E, Ando K, Murase E, Kawakami A. Heterogeneous fates and dynamic rearrangement of regenerative epidermis-derived cells during zebrafish fin regeneration. Development 2018; 145:dev.162016. [DOI: 10.1242/dev.162016] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Accepted: 03/20/2018] [Indexed: 01/14/2023]
Abstract
The regenerative epidermis (RE) is a specialized tissue that plays an essential role in tissue regeneration. However, the fate of the RE during and after regeneration is unknown. In this study, we performed Cre-loxP-mediated cell fate tracking and revealed the fates of major population of regenerative epidermis cells that express fibronectin 1b (fn1b) during zebrafish fin regeneration. Our study showed that these RE cells are mainly recruited from the inter-ray epidermis, and that they follow heterogeneous cell fates. Early recruited cells contribute to initial wound healing and soon disappear by apoptosis, while the later recruited cells contribute to the regenerated epidermis. Intriguingly, many of these cells were also expelled from the regenerated tissue by a dynamic caudal movement of the epidermis over time, and in turn the loss of epidermal cells was replenished by a global self-replication of basal and suprabasal cells in fin. De-differentiation of non-basal epidermal cells into the basal epidermal cells did not occur during regeneration. Overall, our study revealed heterogeneous fates of RE cells and a dynamic rearrangement of the epidermis during and after regeneration.
Collapse
Affiliation(s)
- Eri Shibata
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta, Midori-ku, Yokohama 226-8501, Japan
| | - Kazunori Ando
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta, Midori-ku, Yokohama 226-8501, Japan
| | - Emiko Murase
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta, Midori-ku, Yokohama 226-8501, Japan
| | - Atsushi Kawakami
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta, Midori-ku, Yokohama 226-8501, Japan
| |
Collapse
|
47
|
Sato K, Umesono Y, Mochii M. A transgenic reporter under control of an es1 promoter/enhancer marks wound epidermis and apical epithelial cap during tail regeneration in Xenopus laevis tadpole. Dev Biol 2017; 433:404-415. [PMID: 29291984 DOI: 10.1016/j.ydbio.2017.08.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Revised: 05/22/2017] [Accepted: 08/09/2017] [Indexed: 11/17/2022]
Abstract
Rapid wound healing and subsequent formation of the apical epithelial cap (AEC) are believed to be required for successful appendage regeneration in amphibians. Despite the significant role of AEC in limb regeneration, its role in tail regeneration and the mechanisms that regulate the wound healing and AEC formation are not well understood. We previously identified Xenopus laevis es1, which is preferentially expressed in wounded regions, including the AEC after tail regeneration. In this study we established and characterized transgenic Xenopus laevis lines harboring the enhanced green fluorescent protein (EGFP) gene under control of an es1 gene regulatory sequence (es1:egfp). The EGFP reporter expression was clearly seen in several regions of the embryo and then declined to an undetectable level in larvae, recapitulating the endogenous es1 expression. After amputation of the tadpole tail, EGFP expression was re-activated at the edge of the stump epidermis and then increased in the wound epidermis (WE) covering the amputation surface. As the stump started to regenerate, the EGFP expression became restricted to the most distal epidermal region, including the AEC. EGFP was preferentially expressed in the basal or deep cells but not in the superficial cells of the WE and AEC. We performed a small-scale pharmacological screening for chemicals that affected the expression of EGFP in the stump epidermis after tail amputation. The EGFP expression was attenuated by treatment with an inhibitor for ERK, TGF-β or reactive oxygen species (ROS) signaling. These treatments also impaired wound closure of the amputation surface, suggesting that the three signaling activities are required for es1 expression in the WE and successful wound healing after tail amputation. These findings showed that es1:egfp Xenopus laevis should be a useful tool to analyze molecular mechanisms regulating wound healing and appendage regeneration.
Collapse
Affiliation(s)
- Kentaro Sato
- Graduate School of Life Science, University of Hyogo, 3-2-1 Koto, Kamigori, Akou, Hyogo 678-1297, Japan
| | - Yoshihiko Umesono
- Graduate School of Life Science, University of Hyogo, 3-2-1 Koto, Kamigori, Akou, Hyogo 678-1297, Japan
| | - Makoto Mochii
- Graduate School of Life Science, University of Hyogo, 3-2-1 Koto, Kamigori, Akou, Hyogo 678-1297, Japan.
| |
Collapse
|
48
|
Dogra D, Ahuja S, Kim HT, Rasouli SJ, Stainier DYR, Reischauer S. Opposite effects of Activin type 2 receptor ligands on cardiomyocyte proliferation during development and repair. Nat Commun 2017; 8:1902. [PMID: 29196619 PMCID: PMC5711791 DOI: 10.1038/s41467-017-01950-1] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 10/27/2017] [Indexed: 01/14/2023] Open
Abstract
Zebrafish regenerate damaged myocardial tissue very effectively. Hence, insights into the molecular networks underlying zebrafish heart regeneration might help develop alternative strategies to restore human cardiac performance. While TGF-β signaling has been implicated in zebrafish cardiac regeneration, the role of its individual ligands remains unclear. Here, we report the opposing expression response during zebrafish heart regeneration of two genes, mstnb and inhbaa, which encode TGF-β family ligands. Using gain-of-function (GOF) and loss-of-function (LOF) approaches, we show that these ligands mediate inverse effects on cardiac regeneration and specifically on cardiomyocyte (CM) proliferation. Notably, we find that Inhbaa functions as a CM mitogen and that its overexpression leads to accelerated cardiac recovery and scar clearance after injury. In contrast, mstnb GOF and inhbaa LOF both lead to unresolved scarring after cardiac injury. We further show that Mstnb and Inhbaa inversely control Smad2 and Smad3 transcription factor activities through alternate Activin type 2 receptors.
Collapse
Affiliation(s)
- Deepika Dogra
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231, Bad Nauheim, Germany
| | - Suchit Ahuja
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231, Bad Nauheim, Germany
| | - Hyun-Taek Kim
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231, Bad Nauheim, Germany
| | - S Javad Rasouli
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231, Bad Nauheim, Germany
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231, Bad Nauheim, Germany
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, 94158, CA, USA
| | - Sven Reischauer
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231, Bad Nauheim, Germany.
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, 94158, CA, USA.
| |
Collapse
|
49
|
Jacyniak K, McDonald RP, Vickaryous MK. Tail regeneration and other phenomena of wound healing and tissue restoration in lizards. J Exp Biol 2017; 220:2858-2869. [DOI: 10.1242/jeb.126862] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
ABSTRACT
Wound healing is a fundamental evolutionary adaptation with two possible outcomes: scar formation or reparative regeneration. Scars participate in re-forming the barrier with the external environment and restoring homeostasis to injured tissues, but are well understood to represent dysfunctional replacements. In contrast, reparative regeneration is a tissue-specific program that near-perfectly replicates that which was lost or damaged. Although regeneration is best known from salamanders (including newts and axolotls) and zebrafish, it is unexpectedly widespread among vertebrates. For example, mice and humans can replace their digit tips, while many lizards can spontaneously regenerate almost their entire tail. Whereas the phenomenon of lizard tail regeneration has long been recognized, many details of this process remain poorly understood. All of this is beginning to change. This Review provides a comparative perspective on mechanisms of wound healing and regeneration, with a focus on lizards as an emerging model. Not only are lizards able to regrow cartilage and the spinal cord following tail loss, some species can also regenerate tissues after full-thickness skin wounds to the body, transections of the optic nerve and even lesions to parts of the brain. Current investigations are advancing our understanding of the biological requirements for successful tissue and organ repair, with obvious implications for biomedical sciences and regenerative medicine.
Collapse
Affiliation(s)
- Kathy Jacyniak
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada N1G 2W1
| | - Rebecca P. McDonald
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada N1G 2W1
| | - Matthew K. Vickaryous
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada N1G 2W1
| |
Collapse
|
50
|
König D, Page L, Chassot B, Jaźwińska A. Dynamics of actinotrichia regeneration in the adult zebrafish fin. Dev Biol 2017; 433:416-432. [PMID: 28760345 DOI: 10.1016/j.ydbio.2017.07.024] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 07/25/2017] [Accepted: 07/27/2017] [Indexed: 01/21/2023]
Abstract
The skeleton of adult zebrafish fins comprises lepidotrichia, which are dermal bones of the rays, and actinotrichia, which are non-mineralized spicules at the distal margin of the appendage. Little is known about the regenerative dynamics of the actinotrichia-specific structural proteins called Actinodins. Here, we used immunofluorescence analysis to determine the contribution of two paralogous Actinodin proteins, And1/2, in regenerating fins. Both proteins were detected in the secretory organelles in the mesenchymal cells of the blastema, but only And1 was detected in the epithelial cells of the wound epithelium. The analysis of whole mount fins throughout the entire regenerative process and longitudinal sections revealed that And1-positive fibers are complementary to the lepidotrichia. The analysis of another longfin fish, a gain-of-function mutation in the potassium channel kcnk5b, revealed that the long-fin phenotype is associated with an extended size of actinotrichia during homeostasis and regeneration. Finally, we investigated the role of several signaling pathways in actinotrichia formation and maintenance. This revealed that the pulse-inhibition of either TGFβ/Activin-βA or FGF are sufficient to impair deposition of Actinodin during regeneration. Thus, the dynamic turnover of Actinodin during fin regeneration is regulated by multiple factors, including the osteoblasts, growth rate in a potassium channel mutant, and instructive signaling networks between the epithelium and the blastema of the regenerating fin.
Collapse
Affiliation(s)
- Désirée König
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| | - Lionel Page
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| | - Bérénice Chassot
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| | - Anna Jaźwińska
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland.
| |
Collapse
|