1
|
Le Bleu HK, Kioussi RG, Henner AL, Lewis VM, Stewart S, Stankunas K. Voltage-gated calcium channels generate blastema Ca 2+ fluxes restraining zebrafish fin regenerative outgrowth. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.08.21.608903. [PMID: 39229087 PMCID: PMC11370486 DOI: 10.1101/2024.08.21.608903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Adult zebrafish fins regenerate to their original size regardless of damage extent, providing a tractable model of organ size and scale control. Gain-of-function of voltage-gated K + channels expressed in fibroblast-lineage blastema cells promotes excessive fin outgrowth, leading to a long-finned phenotype. Similarly, inhibition of the Ca 2+ -dependent phosphatase calcineurin during regeneration causes dramatic fin overgrowth. However, Ca 2+ fluxes and their potential origins from dynamic membrane voltages have not been explored or linked to fin size restoration. We used fibroblast-lineage GCaMP imaging of regenerating adult fins to identify widespread, heterogeneous Ca 2+ transients in distal blastema cells. Membrane depolarization of isolated regenerating fin fibroblasts triggered Ca 2+ spikes dependent on voltage-gated Ca 2+ channel activity. Single cell transcriptomics identified the voltage-gated Ca 2+ channels cacna1c (L-type channel), cacna1ba (N-type), and cacna1g (T-type) as candidate mediators of fibroblast-lineage Ca 2+ signaling. Small molecule inhibition revealed L- and/or N-type voltage-gated Ca 2+ channels act during regenerative outgrowth to restore fins to their original scale. Strikingly, cacna1g homozygous mutant zebrafish regenerated extraordinarily long fins due to prolonged outgrowth. The regenerated fins far exceeded their original length but with otherwise normal ray skeletons. Therefore, cacna1g mutants uniquely provide a genetic loss-of-function long-finned model that decouples developmental and regenerative fin outgrowth. Live GCaMP imaging of regenerating fins showed T-type Cacna1g channels enable Ca 2+ dynamics in distal fibroblast-lineage blastemal mesenchyme during the outgrowth phase. We conclude "bioelectricity" for fin size control likely entirely reflects voltage-modulated Ca 2+ dynamics in fibroblast-lineage blastemal cells that specifically and steadily decelerates outgrowth at a rate tuned to restore the original fin size.
Collapse
|
2
|
Allen RS, Biswas SK, Seifert AW. Ear pinna growth and differentiation is conserved in murids and requires BMP signaling for chondrocyte proliferation. Development 2025; 152:DEV204560. [PMID: 39846506 PMCID: PMC11883244 DOI: 10.1242/dev.204560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 01/14/2025] [Indexed: 01/24/2025]
Abstract
Despite being a major target of reconstructive surgery, development of the ear pinna remains poorly studied. Here, we provide a cellular characterization of late gestational and postnatal ear pinna development in two rodents and investigate the role of BMP5 in expansion and differentiation of auricular elastic cartilage. We find that ear pinna development is largely conserved between Mus musculus and the highly regenerative Acomys dimidiatus. The pattern of pre-cartilaginous cells is established early in development. These cells are specified into chondroblasts before ear unfolding and then undergo extensive proliferation before maturation. The elastic cartilage, connective tissue fibroblasts, dermal papilla and sheath cells, and adipocytes in the adult pinna are derived from cranial neural crest. Cellular analysis using the naturally occurring short ear mouse mutant shows that loss of BMP5 does not prevent specification of chondroblasts, but does impair chondroblast proliferation. Finally, chondroblast proliferation remains impaired in the adult mid-distal ear pinna of these mutants. Together, these data establish the developmental basis for differentiation of ear pinna tissues.
Collapse
Affiliation(s)
- Robyn S. Allen
- Department of Biology, University of Kentucky, Lexington, KY 40506, USA
| | - Shishir K. Biswas
- Department of Biology, University of Kentucky, Lexington, KY 40506, USA
| | - Ashley W. Seifert
- Department of Biology, University of Kentucky, Lexington, KY 40506, USA
| |
Collapse
|
3
|
Tan YR, Roan HY, Chen CH. Zebrafish tailfin as an in vivo model for capturing tissue-scale cell dynamics. Semin Cell Dev Biol 2025; 166:29-35. [PMID: 39724824 DOI: 10.1016/j.semcdb.2024.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 12/11/2024] [Accepted: 12/13/2024] [Indexed: 12/28/2024]
Abstract
The intricate control of collective cell dynamics is crucial for enabling organismic development and tissue regeneration. Despite the availability of various in vitro and in vivo models, studies on tissue-scale cell dynamics and associated emergent properties in living systems remain methodically challenging. Here, we describe key advantages of using the adult zebrafish tailfin (caudal fin) as a robust in vivo model for dissecting millimeter-scale collective cell dynamics during regeneration and wound healing in a complex tissue. For researchers considering this model system, we briefly introduce the tailfin anatomy, as well as available transgenic reporter tools and live-imaging setups that may be utilized to study epidermal cell behaviors. To highlight the unique strengths of the zebrafish tailfin model, we present an example project that was made possible by techniques for tracking cell dynamics at a millimeter scale with single-cell resolution in live animals. Finally, we discuss the research directions at the interface of collective cell dynamics and regenerative biology that most excite us and can be examined using the tailfin model.
Collapse
Affiliation(s)
- Yue Rong Tan
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Hsiao-Yuh Roan
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Chen-Hui Chen
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan.
| |
Collapse
|
4
|
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
|
5
|
Ranadive I, Patel S, Pai S, Khaire K, Balakrishnan S. Disruption of BMP and FGF signaling prior to blastema formation causes permanent bending and skeletal malformations in Poecilia latipinna tail fin. ZOOLOGY 2025; 168:126237. [PMID: 39827581 DOI: 10.1016/j.zool.2025.126237] [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: 07/27/2024] [Revised: 01/07/2025] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
Teleost fish, such as Poecilia latipinna, exhibit remarkable regenerative capabilities, making them excellent models for studying tissue regrowth. They regenerate body parts like the tail fin through epimorphic regeneration, involving wound healing, blastema formation (a pool of proliferative cells), and tissue differentiation. Bone Morphogenetic Protein (BMP) and Fibroblast Growth Factor (FGF) signaling pathways play crucial roles in this process, but their specific functions during blastema formation remain unclear. To explore this, BMP and FGF signaling were inhibited using targeted drug treatments prior to blastema formation in amputated tail fins. The treatment group of P. latipinna received drugs at set intervals, and analyses were conducted using skeletal staining, gene expression via quantitative real-time PCR, and protein analysis with Western blotting to assess blastema formation, extracellular matrix (ECM) remodeling, and skeletal patterning. Dual inhibition of BMP and FGF pathways led to significant regenerative defects, including bent blastema and disrupted bone structure, along with downregulation of essential patterning genes like sonic hedgehog (Shh) and bmp2b. Additionally, ECM remodeling and epithelial-to-mesenchymal transition (EMT) were impaired, as shown by reduced matrix metalloproteinases (MMP2 and MMP9), hindering cell migration and blastema stability. Cell proliferation was markedly decreased, as evidenced by reduced proliferating cell nuclear antigen (PCNA) expression and bromodeoxyuridine (BrdU) incorporation, while apoptosis increased, with elevated markers like caspase 3 (casp3) and higher DNA fragmentation. These findings indicate that BMP and FGF signaling are essential for blastema formation and skeletal patterning, with their inhibition causing major regenerative abnormalities.
Collapse
Affiliation(s)
- Isha Ranadive
- Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Sonam Patel
- Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Siddharth Pai
- Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Kashmira Khaire
- Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Suresh Balakrishnan
- Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India.
| |
Collapse
|
6
|
Duca S, Xia Y, Abd Elmagid L, Bakis I, Qiu M, Cao Y, Guo Y, Eichenbaum JV, McCain ML, Kang J, Harrison MRM, Cao J. Differential vegfc expression dictates lymphatic response during zebrafish heart development and regeneration. Development 2024; 151:dev202947. [PMID: 39514676 PMCID: PMC11607685 DOI: 10.1242/dev.202947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024]
Abstract
Vascular endothelial growth factor C (Vegfc) is crucial for lymphatic and blood vessel development, yet its cellular sources and specific functions in heart development remain unclear. To address this, we created a vegfc reporter and an inducible overexpression line in zebrafish. We found vegfc expression in large coronary arteries, circulating thrombocytes, cardiac adipocytes, and outflow tract smooth muscle cells. Notably, although coronary lymphangiogenesis aligns with Vegfc-expressing arteries in juveniles, it occurs only after coronary artery formation. Vegfc overexpression induced ectopic lymphatics on the ventricular surface prior to arterial formation, indicating that Vegfc abundance, rather than arterial presence, drives lymphatic development. However, this overexpression did not affect coronary artery coverage, suggesting a specific role for Vegfc in lymphatic, rather than arterial, development. Thrombocytes emerged as the initial Vegfc source during inflammation following heart injuries, transitioning to endocardial and myocardial expression during regeneration. Lower Vegfc levels in an amputation model corresponded with a lack of lymphatic expansion. Importantly, Vegfc overexpression enhanced lymphatic expansion and promoted scar resolution without affecting cardiomyocyte proliferation, highlighting its role in regulating lymphangiogenesis and promoting heart regeneration.
Collapse
Affiliation(s)
- Sierra Duca
- Cardiovascular Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
| | - Yu Xia
- Cardiovascular Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
| | - Laila Abd Elmagid
- Cardiovascular Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
| | - Isaac Bakis
- Cardiovascular Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
| | - Miaoyan Qiu
- Cardiovascular Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
| | - Yingxi Cao
- Cardiovascular Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
| | - Ylan Guo
- Cardiovascular Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
| | - James V. Eichenbaum
- Alfred E. Mann Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90033, USA
| | - Megan L. McCain
- Alfred E. Mann Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90033, USA
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Junsu Kang
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin – Madison, Madison, WI 53705, USA
| | - Michael R. M. Harrison
- Cardiovascular Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
| | - Jingli Cao
- Cardiovascular Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
| |
Collapse
|
7
|
Fan J, Liu X, Duan Z, Zhao H, Chang Z, Li L. The Regulatory Role of miRNAs in Zebrafish Fin Regeneration. Int J Mol Sci 2024; 25:10542. [PMID: 39408869 PMCID: PMC11477159 DOI: 10.3390/ijms251910542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Since Teleostei fins have a strong regenerative capacity, further research was conducted on the regulation of gene expression during fin regeneration. This research focuses on miRNA, which is a key post-transcriptional regulatory molecule. In this study, a miRNA library for the fin regeneration of zebrafish was constructed to reveal the differential expression of miRNA during fin regeneration and to explore the regulatory pathway for fin regeneration. Following the injection of miRNA agomir into zebrafish, the proliferation of blastema cells and the overall fin regeneration area were significantly reduced. It was observed that the miRNAs impaired blastocyte formation by affecting fin regeneration through the inhibition of the expressions of genes and proteins associated with blastocyte formation (including yap1 and Smad1/5/9), which is an effect associated with the Hippo pathway. Furthermore, it has been demonstrated that miRNAs can impair the patterns and mineralization of newly formed fin rays. The miRNAs influenced fin regeneration by inhibiting the expression of a range of bone-related genes and proteins in osteoblast lineages, including sp7, runx2a, and runx2b. This study provides a valuable reference for the further exploration of morphological bone reconstruction in aquatic vertebrates.
Collapse
Affiliation(s)
| | | | | | | | | | - Li Li
- Molecular and Genetic Laboratory, College of Life Science, Henan Normal University, 46# East of Construction Road, Xinxiang 453007, China; (J.F.)
| |
Collapse
|
8
|
Ortega Granillo A, Zamora D, Schnittker RR, Scott AR, Spluga A, Russell J, Brewster CE, Ross EJ, Acheampong DA, Zhang N, Ferro K, Morrison JA, Rubinstein BY, Perera AG, Wang W, Sánchez Alvarado A. Positional information modulates transient regeneration-activated cell states during vertebrate appendage regeneration. iScience 2024; 27:110737. [PMID: 39286507 PMCID: PMC11404194 DOI: 10.1016/j.isci.2024.110737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 06/18/2024] [Accepted: 08/12/2024] [Indexed: 09/19/2024] Open
Abstract
Injury is common in the life of organisms. Because the extent of damage cannot be predicted, injured organisms must determine how much tissue needs to be restored. Although it is known that amputation position affects the regeneration speed of appendages, mechanisms conveying positional information remain unclear. We investigated tissue dynamics in regenerating caudal fins of the African killifish (Nothobranchius furzeri) and found position-specific, differential spatial distribution modulation, persistence, and magnitude of proliferation. Single-cell RNA sequencing revealed a transient regeneration-activated cell state (TRACS) in the basal epidermis that is amplified to match a given amputation position and expresses components and modifiers of the extracellular matrix (ECM). Notably, CRISPR-Cas9-mediated deletion of the ECM modifier sequestosome 1 (sqstm1) increased the regenerative capacity of distal injuries, suggesting that regeneration growth rate can be uncoupled from amputation position. We propose that basal epidermis TRACS transduce positional information to the regenerating blastema by remodeling the ECM.
Collapse
Affiliation(s)
| | - Daniel Zamora
- Stowers Institute for Medical Research, 1000 E 50th St, Kansas City, MO 64110, USA
| | - Robert R Schnittker
- Stowers Institute for Medical Research, 1000 E 50th St, Kansas City, MO 64110, USA
| | - Allison R Scott
- Stowers Institute for Medical Research, 1000 E 50th St, Kansas City, MO 64110, USA
| | - Alessia Spluga
- Stowers Institute for Medical Research, 1000 E 50th St, Kansas City, MO 64110, USA
| | - Jonathon Russell
- Stowers Institute for Medical Research, 1000 E 50th St, Kansas City, MO 64110, USA
| | - Carolyn E Brewster
- Stowers Institute for Medical Research, 1000 E 50th St, Kansas City, MO 64110, USA
| | - Eric J Ross
- Stowers Institute for Medical Research, 1000 E 50th St, Kansas City, MO 64110, USA
| | - Daniel A Acheampong
- Stowers Institute for Medical Research, 1000 E 50th St, Kansas City, MO 64110, USA
| | - Ning Zhang
- Stowers Institute for Medical Research, 1000 E 50th St, Kansas City, MO 64110, USA
| | - Kevin Ferro
- Stowers Institute for Medical Research, 1000 E 50th St, Kansas City, MO 64110, USA
| | - Jason A Morrison
- Stowers Institute for Medical Research, 1000 E 50th St, Kansas City, MO 64110, USA
| | - Boris Y Rubinstein
- Stowers Institute for Medical Research, 1000 E 50th St, Kansas City, MO 64110, USA
| | - Anoja G Perera
- Stowers Institute for Medical Research, 1000 E 50th St, Kansas City, MO 64110, USA
| | - Wei Wang
- National Institute of Biological Sciences, 7 Science Park Road ZGC Life Science Park, Beijing 102206, China
| | | |
Collapse
|
9
|
López-Cuevas P, Oates TCL, Tong Q, McGowan LM, Cross SJ, Xu C, Zhao Y, Yin Z, Toye AM, Boussahel A, Hammond CL, Mann S, Martin P. Reprogramming macrophages with R848-loaded artificial protocells to modulate skin and skeletal wound healing. J Cell Sci 2024; 137:jcs262202. [PMID: 39078119 PMCID: PMC11385641 DOI: 10.1242/jcs.262202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 07/15/2024] [Indexed: 07/31/2024] Open
Abstract
After tissue injury, inflammatory cells are rapidly recruited to the wound where they clear microbes and other debris, and coordinate the behaviour of other cell lineages at the repair site in both positive and negative ways. In this study, we take advantage of the translucency and genetic tractability of zebrafish to evaluate the feasibility of reprogramming innate immune cells in vivo with cargo-loaded protocells and investigate how this alters the inflammatory response in the context of skin and skeletal repair. Using live imaging, we show that protocells loaded with R848 cargo (which targets TLR7 and TLR8 signalling), are engulfed by macrophages resulting in their switching to a pro-inflammatory phenotype and altering their regulation of angiogenesis, collagen deposition and re-epithelialization during skin wound healing, as well as dampening osteoblast and osteoclast recruitment and bone mineralization during fracture repair. For infected skin wounds, R848-reprogrammed macrophages exhibited enhanced bactericidal activities leading to improved healing. We replicated our zebrafish studies in cultured human macrophages, and showed that R848-loaded protocells similarly reprogramme human cells, indicating how this strategy might be used to modulate wound inflammation in the clinic.
Collapse
Affiliation(s)
- Paco López-Cuevas
- School of Biochemistry, Biomedical Sciences Building, University Walk, University of Bristol, Bristol BS8 1TD, UK
| | - Tiah C L Oates
- School of Cellular and Molecular Medicine, Biomedical Sciences Building, University Walk, University of Bristol, Bristol BS8 1TD, UK
| | - Qiao Tong
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University Walk, University of Bristol, Bristol BS8 1TD, UK
| | - Lucy M McGowan
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University Walk, University of Bristol, Bristol BS8 1TD, UK
| | - Stephen J Cross
- Wolfson Bioimaging Facility, Biomedical Sciences Building, University Walk, University of Bristol, Bristol BS8 1TD, UK
| | - Can Xu
- Centre for Protolife Research, School of Chemistry, University of Bristol, Bristol BS8 1TS, UK
| | - Yu Zhao
- Centre for Protolife Research, School of Chemistry, University of Bristol, Bristol BS8 1TS, UK
| | - Zhuping Yin
- Centre for Protolife Research, School of Chemistry, University of Bristol, Bristol BS8 1TS, UK
| | - Ashley M Toye
- School of Biochemistry, Biomedical Sciences Building, University Walk, University of Bristol, Bristol BS8 1TD, UK
- National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Red Blood Cell Products, University of Bristol, Bristol BS34 7QH, UK
| | - Asme Boussahel
- School of Cellular and Molecular Medicine, Biomedical Sciences Building, University Walk, University of Bristol, Bristol BS8 1TD, UK
| | - Chrissy L Hammond
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University Walk, University of Bristol, Bristol BS8 1TD, UK
| | - Stephen Mann
- Centre for Protolife Research, School of Chemistry, University of Bristol, Bristol BS8 1TS, UK
- Max Planck Bristol Centre for Minimal Biology, School of Chemistry, University of Bristol, Bristol BS8 1TS, UK
| | - Paul Martin
- School of Biochemistry, Biomedical Sciences Building, University Walk, University of Bristol, Bristol BS8 1TD, UK
| |
Collapse
|
10
|
Cudak N, López-Delgado AC, Rost F, Kurth T, Lesche M, Reinhardt S, Dahl A, Rulands S, Knopf F. Compartmentalization and synergy of osteoblasts drive bone formation in the regenerating fin. iScience 2024; 27:108841. [PMID: 38318374 PMCID: PMC10838958 DOI: 10.1016/j.isci.2024.108841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 12/13/2023] [Accepted: 01/03/2024] [Indexed: 02/07/2024] Open
Abstract
Zebrafish regenerate their fins which involves a component of cell plasticity. It is currently unclear how regenerate cells divide labor to allow for appropriate growth and patterning. Here, we studied lineage relationships of fluorescence-activated cell sorting-enriched epidermal, bone-forming (osteoblast), and (non-osteoblast) blastemal fin regenerate cells by single-cell RNA sequencing, lineage tracing, targeted osteoblast ablation, and electron microscopy. Most osteoblasts in the outgrowing regenerate derive from osterix+ osteoblasts, while mmp9+ cells reside at segment joints. Distal blastema cells contribute to distal osteoblast progenitors, suggesting compartmentalization of the regenerating appendage. Ablation of osterix+ osteoblasts impairs segment joint and bone matrix formation and decreases regenerate length which is partially compensated for by distal regenerate cells. Our study characterizes expression patterns and lineage relationships of rare fin regenerate cell populations, indicates inherent detection and compensation of impaired regeneration, suggests variable dependence on growth factor signaling, and demonstrates zonation of the elongating fin regenerate.
Collapse
Affiliation(s)
- Nicole Cudak
- CRTD - Center for Regenerative Therapies TU Dresden, Dresden, Germany
- Center for Healthy Aging, Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Alejandra Cristina López-Delgado
- CRTD - Center for Regenerative Therapies TU Dresden, Dresden, Germany
- Center for Healthy Aging, Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Fabian Rost
- DRESDEN-concept Genome Center, DFG NGS Competence Center, c/o Center for Molecular and Cellular Bioengineering (CMCB), TU Dresden, Dresden, Germany
| | - Thomas Kurth
- Core Facility Electron Microscopy and Histology, Technology Platform, Center for Molecular and Cellular Bioengineering (CMCB), TU Dresden, Dresden, Germany
| | - Mathias Lesche
- DRESDEN-concept Genome Center, DFG NGS Competence Center, c/o Center for Molecular and Cellular Bioengineering (CMCB), TU Dresden, Dresden, Germany
| | - Susanne Reinhardt
- DRESDEN-concept Genome Center, DFG NGS Competence Center, c/o Center for Molecular and Cellular Bioengineering (CMCB), TU Dresden, Dresden, Germany
| | - Andreas Dahl
- DRESDEN-concept Genome Center, DFG NGS Competence Center, c/o Center for Molecular and Cellular Bioengineering (CMCB), TU Dresden, Dresden, Germany
| | - Steffen Rulands
- Max Planck Institute for the Physics of Complex Systems, Dresden, Germany
- Ludwig-Maximilians-Universität München, Arnold-Sommerfeld-Center for Theoretical Physics, München, Germany
| | - Franziska Knopf
- CRTD - Center for Regenerative Therapies TU Dresden, Dresden, Germany
- Center for Healthy Aging, Faculty of Medicine, TU Dresden, Dresden, Germany
| |
Collapse
|
11
|
Serra KM, Vyzas C, Shehreen S, Chipendo I, Clifford KM, Youngstrom DW, Devoto SH. Vertebral pattern and morphology is determined during embryonic segmentation. Dev Dyn 2024; 253:204-214. [PMID: 37688793 DOI: 10.1002/dvdy.649] [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: 01/03/2023] [Revised: 06/29/2023] [Accepted: 07/17/2023] [Indexed: 09/11/2023] Open
Abstract
BACKGROUND The segmented nature of the adult vertebral column is based on segmentation of the paraxial mesoderm during early embryogenesis. Disruptions to embryonic segmentation, whether caused by genetic lesions or environmental stress, result in adult vertebral pathologies. However, the mechanisms linking embryonic segmentation and the details of adult vertebral morphology are poorly understood. RESULTS We induced border defects using two approaches in zebrafish: heat stress and misregulation of embryonic segmentation genes tbx6, mesp-ba, and ripply1. We assayed vertebral length, regularity, and polarity using microscopic and radiological imaging. In population studies, we find a correlation between specific embryonic border defects and specific vertebral defects, and within individual fish, we trace specific adult vertebral defects to specific embryonic border defects. CONCLUSIONS Our data reveal that transient disruptions of embryonic segment border formation led to significant vertebral anomalies that persist through adulthood. The spacing of embryonic borders controls the length of the vertebra. The positions of embryonic borders control the positions of ribs and arches. Embryonic borders underlie fusions and divisions between adjacent spines and ribs. These data suggest that segment borders have a dominant role in vertebral development.
Collapse
Affiliation(s)
- Kevin M Serra
- Department of Biology, Wesleyan University, Middletown, Connecticut, USA
| | - Christina Vyzas
- Department of Biology, Wesleyan University, Middletown, Connecticut, USA
| | - Sarah Shehreen
- Department of Biology, Wesleyan University, Middletown, Connecticut, USA
| | - Iris Chipendo
- Department of Biology, Wesleyan University, Middletown, Connecticut, USA
| | - Katherine M Clifford
- Department of Biology, Wesleyan University, Middletown, Connecticut, USA
- Department of Neurology, Stanford University, Stanford, California, USA
| | - Daniel W Youngstrom
- Department of Orthopaedic Surgery, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Stephen H Devoto
- Department of Biology, Wesleyan University, Middletown, Connecticut, USA
| |
Collapse
|
12
|
Komiya H, Sato Y, Kimura H, Kawakami A. Independent mesenchymal progenitor pools respectively produce and maintain osteogenic and chondrogenic cells in zebrafish. Dev Growth Differ 2024; 66:161-171. [PMID: 38193362 PMCID: PMC11457501 DOI: 10.1111/dgd.12908] [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: 11/02/2023] [Revised: 12/18/2023] [Accepted: 12/21/2023] [Indexed: 01/10/2024]
Abstract
Skeletal tissues including cartilage and bones are characteristic features of vertebrates that are crucial for supporting body morphology and locomotion. Studies mainly in mice have shown that osteoblasts and chondroblasts are supplied from several progenitors like the sclerotome cells in the embryonic stage, osteo-chondroprogenitors in growing long bones, and skeletal stem cells of bone marrow in the postnatal period. However, the exact origins of progenitor cells, their lineage relationships, and their potential to differentiate into osteoblasts and chondroblasts from embryos to adult tissues are not well understood. In this study, we conducted clonal cell tracking in zebrafish and showed that sox9a+ cells are already committed to either chondrogenic or osteogenic fates during embryonic stages and that respective progenies are independently maintained as mesenchymal progenitor pools. Once committed, they never change their lineage identities throughout animal life, even through regeneration. In addition, we further revealed that only osteogenic mesenchymal cells replenish the osteoblast progenitor cells (OPCs), a population of reserved tissue stem cells found to be involved in the de novo production of osteoblasts during regeneration and homeostasis in zebrafish. Thus, our clonal cell tracking study in zebrafish firstly revealed that the mesenchymal progenitor cells that are fated to develop into either chondroblasts or osteoblasts serve as respective tissue stem cells to maintain skeletal tissue homeostasis. Such mesenchymal progenitors dedicated to producing either chondroblasts or osteoblasts would be important targets for skeletal tissue regeneration.
Collapse
Grants
- 19K22417 Japan Society for the Promotion of Science
- 22K19306 Japan Society for the Promotion of Science
- 21H04764 Ministry of Education, Culture, Sports, Science, and Technology
- JP23ama121020 Japan Agency for Medical Research and Development
- 19H03232 Ministry of Education, Culture, Sports, Science and Technology
- Japan Society for the Promotion of Science
- Japan Agency for Medical Research and Development
- Ministry of Education, Culture, Sports, Science and Technology
Collapse
Affiliation(s)
- Hiroaki Komiya
- School of Life Science and Technology, Tokyo Institute of TechnologyYokohamaJapan
| | - Yuko Sato
- Institute of Innovative Research, Tokyo Institute of TechnologyYokohamaJapan
| | - Hiroshi Kimura
- Institute of Innovative Research, Tokyo Institute of TechnologyYokohamaJapan
| | - Atsushi Kawakami
- School of Life Science and Technology, Tokyo Institute of TechnologyYokohamaJapan
| |
Collapse
|
13
|
Zhang Y, Bi J, Ning Y, Feng J. Methodology Advances in Vertebrate Age Estimation. Animals (Basel) 2024; 14:343. [PMID: 38275802 PMCID: PMC10812784 DOI: 10.3390/ani14020343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/09/2024] [Accepted: 01/18/2024] [Indexed: 01/27/2024] Open
Abstract
Age is a core metric in vertebrate management, and the correct estimation of the age of an individual plays a principal role in comprehending animal behavior, identifying genealogical information, and assessing the potential reproductive capacity of populations. Vertebrates have a vertebral column and a distinct head containing a developed brain; they have played an important role in the study of biological evolution. However, biological age estimations constantly exhibit large deviations due to the diversity of vertebrate taxon species, sample types, and determination methods. To systematically and comprehensively understand age estimation methods in different situations, we classify the degree of damage to vertebrates during sample collection, present the sample types and their applications, list commonly applied methods, present methodological recommendations based on the combination of accuracy and implementability, and, finally, predict future methods for vertebrate age assessments, taking into account the current level of research and requirements. Through comprehensive data gathering and compilation, this work serves as an introduction and summary for those who are eager to catch up on related fields and facilitates the rapid and accurate selection of an evaluation method for researchers engaged in related research. This is essential to promote animal conservation and guide the smooth implementation of conservation management plans.
Collapse
Affiliation(s)
- Yifei Zhang
- College of Life Science, Jilin Agricultural University, Changchun 130118, China; (Y.Z.); (J.B.)
- Jilin Provincial International Cooperation Key Laboratory for Biological Control of Agricultural Pests, Changchun 130118, China
| | - Jinping Bi
- College of Life Science, Jilin Agricultural University, Changchun 130118, China; (Y.Z.); (J.B.)
- Jilin Provincial International Cooperation Key Laboratory for Biological Control of Agricultural Pests, Changchun 130118, China
| | - Yao Ning
- College of Life Science, Jilin Agricultural University, Changchun 130118, China; (Y.Z.); (J.B.)
- Jilin Provincial International Cooperation Key Laboratory for Biological Control of Agricultural Pests, Changchun 130118, China
| | - Jiang Feng
- College of Life Science, Jilin Agricultural University, Changchun 130118, China; (Y.Z.); (J.B.)
- Jilin Provincial International Cooperation Key Laboratory for Biological Control of Agricultural Pests, Changchun 130118, China
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, Changchun 130117, China
- Key Laboratory of Vegetation Ecology of Education Ministry, Institute of Grassland Science, Northeast Normal University, Changchun 130024, China
| |
Collapse
|
14
|
Tan WH, Winkler C. Lineage Tracing of Bone Cells in the Regenerating Fin and During Repair of Bone Lesions. Methods Mol Biol 2024; 2707:99-110. [PMID: 37668907 DOI: 10.1007/978-1-0716-3401-1_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2023]
Abstract
Small teleost fishes such as zebrafish and medaka show remarkable regeneration capabilities upon tissue injury or amputation. To elucidate cellular mechanisms of teleost tissue repair and regeneration processes, the Cre/LoxP recombination system for cell lineage tracing is a widely used technique. In this chapter, we describe protocols used for inducible Cre/LoxP recombination-mediated lineage tracing of osteoblast progenitors during medaka fin regeneration as well as during the repair of osteoporosis-like bone lesions in the medaka vertebral column. Our approach can be adapted for lineage tracing of other cell populations in the regenerating teleost fin or in other tissues undergoing repair.
Collapse
Affiliation(s)
- Wen Hui Tan
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.
| | - Christoph Winkler
- Department of Biological Sciences and Centre for Bioimaging Sciences, National University of Singapore, Singapore, Singapore
| |
Collapse
|
15
|
De Simone A. Quantitative Live Imaging of Zebrafish Scale Regeneration: From Adult Fish to Signaling Patterns and Tissue Flows. Methods Mol Biol 2024; 2707:185-204. [PMID: 37668913 DOI: 10.1007/978-1-0716-3401-1_12] [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] [Indexed: 09/06/2023]
Abstract
In regeneration, a damaged body part grows back to its original form. Understanding the mechanisms and physical principles underlying this process has been limited by the difficulties of visualizing cell signals and behaviors in regeneration. Zebrafish scales are emerging as a model system to investigate morphogenesis during vertebrate regeneration using quantitative live imaging. Scales are millimeter-sized dermal bone disks forming a skeletal armor on the body of the fish. The scale bone is deposited by an adjacent monolayer of osteoblasts that, after scale loss, regenerates in about 2 weeks. This intriguing regenerative process is accessible to live confocal microscopy, quantifications, and mathematical modeling. Here, I describe methods to image scale regeneration live, tissue-wide and at sub-cellular resolution. Furthermore, I describe methods to process the resulting images and quantify cell, tissue, and signal dynamics.
Collapse
Affiliation(s)
- Alessandro De Simone
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA.
- Duke Regeneration Center, Duke University, Durham, NC, USA.
- Department of Genetics and Evolution, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
16
|
Carletti A, Rosa JT, Pes K, Borges I, Santos T, Barreira L, Varela J, Pereira H, Cancela ML, Gavaia PJ, Laizé V. The osteogenic and mineralogenic potential of the microalgae Skeletonema costatum and Tetraselmis striata CTP4 in fish models. Cell Mol Life Sci 2023; 80:310. [PMID: 37777592 PMCID: PMC10543572 DOI: 10.1007/s00018-023-04953-y] [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] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 09/04/2023] [Accepted: 09/05/2023] [Indexed: 10/02/2023]
Abstract
Skeletal disorders are problematic aspects for the aquaculture industry as skeletal deformities, which affect most species of farmed fish, increase production costs and affect fish welfare. Following recent findings that show the presence of osteoactive compounds in marine organisms, we evaluated the osteogenic and mineralogenic potential of commercially available microalgae strains Skeletonema costatum and Tetraselmis striata CTP4 in several fish systems. Ethanolic extracts increased extracellular matrix mineralization in gilthead seabream (Sparus aurata) bone-derived cell cultures and promoted osteoblastic differentiation in zebrafish (Danio rerio) larvae. Long-term dietary exposure to both extracts increased bone mineralization in zebrafish and upregulated the expression of genes involved in bone formation (sp7, col1a1a, oc1, and oc2), bone remodeling (acp5a), and antioxidant defenses (cat, sod1). Extracts also improved the skeletal status of zebrafish juveniles by reducing the incidence of skeletal anomalies. Our results indicate that both strains of microalgae contain osteogenic and mineralogenic compounds, and that ethanolic extracts have the potential for an application in the aquaculture sector as dietary supplements to support fish bone health. Future studies should also identify osteoactive compounds and establish whether they can be used in human health to broaden the therapeutic options for bone erosive disorders such as osteoporosis.
Collapse
Affiliation(s)
- Alessio Carletti
- Centre of Marine Sciences (CCMAR), University of Algarve, Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve, Faro, Portugal
| | - Joana T. Rosa
- Centre of Marine Sciences (CCMAR), University of Algarve, Faro, Portugal
- Present Address: Collaborative Laboratory for Sustainable and Smart Aquaculture (S2AQUAcoLAB), Olhão, Portugal
| | - Katia Pes
- Centre of Marine Sciences (CCMAR), University of Algarve, Faro, Portugal
| | - Inês Borges
- Centre of Marine Sciences (CCMAR), University of Algarve, Faro, Portugal
| | - Tamára Santos
- Centre of Marine Sciences (CCMAR), University of Algarve, Faro, Portugal
| | - Luísa Barreira
- Centre of Marine Sciences (CCMAR), University of Algarve, Faro, Portugal
- Associação Oceano Verde (GreenCoLab), Faro, Portugal
| | - João Varela
- Centre of Marine Sciences (CCMAR), University of Algarve, Faro, Portugal
- Associação Oceano Verde (GreenCoLab), Faro, Portugal
| | - Hugo Pereira
- Centre of Marine Sciences (CCMAR), University of Algarve, Faro, Portugal
- Associação Oceano Verde (GreenCoLab), Faro, Portugal
| | - M. Leonor Cancela
- Centre of Marine Sciences (CCMAR), University of Algarve, Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve, Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve, Faro, Portugal
| | - Paulo J. Gavaia
- Centre of Marine Sciences (CCMAR), University of Algarve, Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve, Faro, Portugal
- Associação Oceano Verde (GreenCoLab), Faro, Portugal
| | - Vincent Laizé
- Centre of Marine Sciences (CCMAR), University of Algarve, Faro, Portugal
- Present Address: Collaborative Laboratory for Sustainable and Smart Aquaculture (S2AQUAcoLAB), Olhão, Portugal
| |
Collapse
|
17
|
Craig EW, Black EC, Goo CE, Swearer AA, Yee NG, Rasmussen JP. Dendritic atoh1a+ cells serve as transient intermediates during zebrafish Merkel cell development and regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.14.557830. [PMID: 37745341 PMCID: PMC10515958 DOI: 10.1101/2023.09.14.557830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Sensory cells often adopt specific morphologies that aid in the detection of external stimuli. Merkel cells encode gentle touch stimuli in vertebrate skin and adopt a reproducible shape characterized by spiky, actin-rich microvilli that emanate from the cell surface. The mechanism by which Merkel cells acquire this stereotyped morphology from basal keratinocyte progenitors is unknown. Here, we establish that dendritic Merkel cells (dMCs) express atonal homolog 1a (atoh1a), extend dynamic filopodial processes, and arise in transient waves during zebrafish skin development and regeneration. We find that dMCs share molecular similarities with both basal keratinocytes and Merkel cells, yet display mesenchymal-like behaviors, including local cell motility and proliferation within the epidermis. Furthermore, dMCs can directly adopt the mature, microvilliated Merkel cell morphology through substantial remodeling of the actin cytoskeleton. Loss of Ectodysplasin A signaling alters the morphology of dMCs and Merkel cells within specific skin regions. Our results show that dMCs represent an intermediate state in the Merkel cell maturation program and identify Ectodysplasin A signaling as a key regulator of Merkel cell morphology.
Collapse
Affiliation(s)
- Evan W. Craig
- Department of Biology, University of Washington, Seattle, WA, USA
| | - Erik C. Black
- Department of Biology, University of Washington, Seattle, WA, USA
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA, USA
| | - Camille E.A. Goo
- Department of Biology, University of Washington, Seattle, WA, USA
| | - Avery Angell Swearer
- Department of Biology, University of Washington, Seattle, WA, USA
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA, USA
| | - Nathaniel G. Yee
- Department of Biology, University of Washington, Seattle, WA, USA
| | - Jeffrey P. Rasmussen
- Department of Biology, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
18
|
Schuster KJ, Christiaen L. The Chordate Origins of Heart Regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.19.558507. [PMID: 37781597 PMCID: PMC10541106 DOI: 10.1101/2023.09.19.558507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
The human heart is infamous for not healing after infarction in adults, prompting biomedical interest in species that can regenerate damaged hearts. In such animals as zebrafish and neonatal mice, cardiac repair relies on remaining heart tissue supporting cardiomyocyte proliferation. Natural de novo cardiogenesis in post-embryonic stages thus remains elusive. Here we show that the tunicate Ciona, an ascidian among the closest living relatives to the vertebrates, can survive complete chemogenetic ablation of the heart and loss of cardiac function, and recover both cardiac tissue and contractility. As in vertebrates, Ciona heart regeneration relies on Bone Morphogenetic Protein (BMP) signaling-dependent proliferation of cardiomyocytes, providing insights into the evolutionary origins of regenerative cardiogenesis in chordates. Remarkably, prospective lineage tracing by photoconversion of the fluorescent protein Kaede suggested that new cardiomyocytes can emerge from endodermal lineages in post-metamorphic animals, providing an unprecedented case of regenerative de novo cardiogenesis. Finally, while embryos cannot compensate for early losses of the cardiogenic lineage, forming heartless juveniles, developing animals gain their regenerative ability during metamorphosis, uncovering a fundamental transition between deterministic embryogenesis and regulative post-embryonic development.
Collapse
Affiliation(s)
- Keaton J Schuster
- Center for Developmental Genetics, Department of Biology, New York University, New York, NY, USA
| | - Lionel Christiaen
- Center for Developmental Genetics, Department of Biology, New York University, New York, NY, USA
- Center for Genomics and Systems Biology, Department of Biology, New York University, New York, NY, USA
- Michael Sars Centre, University of Bergen, Bergen, Norway
| |
Collapse
|
19
|
Anderson T, Mo J, Gagarin E, Sherwood D, Blumenkrantz M, Mao E, Leon G, Levitz H, Chen HJ, Tseng KC, Fabian P, Crump JG, Smeeton J. Ligament injury in adult zebrafish triggers ECM remodeling and cell dedifferentiation for scar-free regeneration. NPJ Regen Med 2023; 8:51. [PMID: 37726321 PMCID: PMC10509200 DOI: 10.1038/s41536-023-00329-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 08/31/2023] [Indexed: 09/21/2023] Open
Abstract
After traumatic injury, healing of mammalian ligaments is typically associated with fibrotic scarring as opposed to scar-free regeneration. In contrast, here we show that the ligament supporting the jaw joint of adult zebrafish is capable of rapid and complete scar-free healing. Following surgical transection of the jaw joint ligament, we observe breakdown of ligament tissue adjacent to the cut sites, expansion of mesenchymal tissue within the wound site, and then remodeling of extracellular matrix (ECM) to a normal ligament morphology. Lineage tracing of mature ligamentocytes following transection shows that they dedifferentiate, undergo cell cycle re-entry, and contribute to the regenerated ligament. Single-cell RNA sequencing of the regenerating ligament reveals dynamic expression of ECM genes in neural-crest-derived mesenchymal cells, as well as diverse immune cells expressing the endopeptidase-encoding gene legumain. Analysis of legumain mutant zebrafish shows a requirement for early ECM remodeling and efficient ligament regeneration. Our study establishes a new model of adult scar-free ligament regeneration and highlights roles of immune-mesenchyme cross-talk in ECM remodeling that initiates regeneration.
Collapse
Affiliation(s)
- Troy Anderson
- Columbia Stem Cell Initiative, Department of Rehabilitation and Regenerative Medicine, and Department of Genetics and Development, Columbia University Irving Medical Center, Columbia University, New York, NY, 10032, USA
| | - Julia Mo
- Columbia Stem Cell Initiative, Department of Rehabilitation and Regenerative Medicine, and Department of Genetics and Development, Columbia University Irving Medical Center, Columbia University, New York, NY, 10032, USA
| | - Ernesto Gagarin
- Columbia Stem Cell Initiative, Department of Rehabilitation and Regenerative Medicine, and Department of Genetics and Development, Columbia University Irving Medical Center, Columbia University, New York, NY, 10032, USA
| | - Desmarie Sherwood
- Columbia Stem Cell Initiative, Department of Rehabilitation and Regenerative Medicine, and Department of Genetics and Development, Columbia University Irving Medical Center, Columbia University, New York, NY, 10032, USA
| | - Maria Blumenkrantz
- Columbia Stem Cell Initiative, Department of Rehabilitation and Regenerative Medicine, and Department of Genetics and Development, Columbia University Irving Medical Center, Columbia University, New York, NY, 10032, USA
| | - Eric Mao
- Columbia Stem Cell Initiative, Department of Rehabilitation and Regenerative Medicine, and Department of Genetics and Development, Columbia University Irving Medical Center, Columbia University, New York, NY, 10032, USA
- Department of Biological Sciences, Columbia College, Columbia University, New York, NY, 10027, USA
| | - Gianna Leon
- Columbia Stem Cell Initiative, Department of Rehabilitation and Regenerative Medicine, and Department of Genetics and Development, Columbia University Irving Medical Center, Columbia University, New York, NY, 10032, USA
- Packer Collegiate Institute, New York, NY, 11201, USA
| | - Hailey Levitz
- Columbia Stem Cell Initiative, Department of Rehabilitation and Regenerative Medicine, and Department of Genetics and Development, Columbia University Irving Medical Center, Columbia University, New York, NY, 10032, USA
- Department of Chemistry, Barnard College, Columbia University, New York, NY, 10027, USA
| | - Hung-Jhen Chen
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Kuo-Chang Tseng
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Peter Fabian
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - J Gage Crump
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Joanna Smeeton
- Columbia Stem Cell Initiative, Department of Rehabilitation and Regenerative Medicine, and Department of Genetics and Development, Columbia University Irving Medical Center, Columbia University, New York, NY, 10032, USA.
| |
Collapse
|
20
|
Rebello D, Wohler E, Erfani V, Li G, Aguilera AN, Santiago-Cornier A, Zhao S, Hwang SW, Steiner RD, Zhang TJ, Gurnett CA, Raggio C, Wu N, Sobreira N, Giampietro PF, Ciruna B. COL11A2 as a candidate gene for vertebral malformations and congenital scoliosis. Hum Mol Genet 2023; 32:2913-2928. [PMID: 37462524 PMCID: PMC10508038 DOI: 10.1093/hmg/ddad117] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 05/08/2023] [Accepted: 07/13/2023] [Indexed: 07/23/2023] Open
Abstract
Human vertebral malformations (VMs) have an estimated incidence of 1/2000 and are associated with significant health problems including congenital scoliosis (CS) and recurrent organ system malformation syndromes such as VACTERL (vertebral anomalies; anal abnormalities; cardiac abnormalities; tracheo-esophageal fistula; renal anomalies; limb anomalies). The genetic cause for the vast majority of VMs are unknown. In a CS/VM patient cohort, three COL11A2 variants (R130W, R1407L and R1413H) were identified in two patients with cervical VM. A third patient with a T9 hemivertebra and the R130W variant was identified from a separate study. These substitutions are predicted to be damaging to protein function, and R130 and R1407 residues are conserved in zebrafish Col11a2. To determine the role for COL11A2 in vertebral development, CRISPR/Cas9 was used to create a nonsense mutation (col11a2L642*) as well as a full gene locus deletion (col11a2del) in zebrafish. Both col11a2L642*/L642* and col11a2del/del mutant zebrafish exhibit vertebral fusions in the caudal spine, which form due to mineralization across intervertebral segments. To determine the functional consequence of VM-associated variants, we assayed their ability to suppress col11a2del VM phenotypes following transgenic expression within the developing spine. While wildtype col11a2 expression suppresses fusions in col11a2del/+ and col11a2del/del backgrounds, patient missense variant-bearing col11a2 failed to rescue the loss-of-function phenotype in these animals. These results highlight an essential role for COL11A2 in vertebral development and support a pathogenic role for two missense variants in CS.
Collapse
Affiliation(s)
- Denise Rebello
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
- Department of Molecular Genetics, The University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Elizabeth Wohler
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Vida Erfani
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
- Department of Molecular Genetics, The University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Guozhuang Li
- Department of Orthopedic Surgery, Key Laboratory of Big Data for Spinal Deformities, Beijing Key Laboratory for Genetic Research of Skeletal Deformity, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Alexya N Aguilera
- Department of Pediatrics, University of Illinois-Chicago, Chicago, IL 60612, USA
| | - Alberto Santiago-Cornier
- Genetic Section, San Jorge Children’s and Women’s Hospital, San Juan, Puerto Rico 00912, USA
- Department of Public Health, Ponce Health Sciences University, Ponce, Puerto Rico 00912, USA
| | - Sen Zhao
- Department of Orthopedic Surgery, Key Laboratory of Big Data for Spinal Deformities, Beijing Key Laboratory for Genetic Research of Skeletal Deformity, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Steven W Hwang
- Shriners Children’s-Philadelphia, Philadelphia, PA 19140, USA
| | - Robert D Steiner
- Department of Pediatrics, University of Wisconsin, Madison, WI 54449, USA
- Marshfield Clinic Health System, Marshfield, WI 54449, USA
| | - Terry Jianguo Zhang
- Department of Orthopedic Surgery, Key Laboratory of Big Data for Spinal Deformities, Beijing Key Laboratory for Genetic Research of Skeletal Deformity, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Christina A Gurnett
- Department of Neurology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | | | - Nan Wu
- Department of Orthopedic Surgery, Key Laboratory of Big Data for Spinal Deformities, Beijing Key Laboratory for Genetic Research of Skeletal Deformity, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Nara Sobreira
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Philip F Giampietro
- Department of Pediatrics, University of Illinois-Chicago, Chicago, IL 60612, USA
| | - Brian Ciruna
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
- Department of Molecular Genetics, The University of Toronto, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
21
|
Allen RS, Biswas SK, Seifert AW. Neural crest cells give rise to non-myogenic mesenchymal tissue in the adult murid ear pinna. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.06.552195. [PMID: 37609220 PMCID: PMC10441307 DOI: 10.1101/2023.08.06.552195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Despite being a major target of reconstructive surgery, development of the external ear pinna remains poorly studied. As a craniofacial organ highly accessible to manipulation and highly conserved among mammals, the ear pinna represents a valuable model for the study of appendage development and wound healing in the craniofacial complex. Here we provide a cellular characterization of late gestational and postnatal ear pinna development in Mus musculus and Acomys cahirinus and demonstrate that ear pinna development is largely conserved between these species. Using Wnt1-cre;ROSAmT/mG mice we find that connective tissue fibroblasts, elastic cartilage, dermal papilla cells, dermal sheath cells, vasculature, and adipocytes in the adult pinna are derived from cranial crest. In contrast, we find that skeletal muscle and hair follicles are not derived from neural crest cells. Cellular analysis using the naturally occurring short ear mouse mutant shows that elastic cartilage does not develop properly in distal pinna due to impaired chondroprogenitor proliferation. Interestingly, while chondroprogenitors develop in a mostly continuous sheet, the boundaries of cartilage loss in the short ear mutant strongly correlate with locations of vasculature-conveying foramen. Concomitant with loss of elastic cartilage we report increased numbers of adipocytes, but this seems to be a state acquired in adulthood rather than a developmental abnormality. In addition, chondrogenesis remains impaired in the adult mid-distal ear pinna of these mutants. Together these data establish a developmental basis for the study of the ear pinna with intriguing insights into the development of elastic cartilage.
Collapse
Affiliation(s)
- Robyn S. Allen
- Department of Biology, University of Kentucky, Lexington, KY 40506, USA
| | - Shishir K. Biswas
- Department of Biology, University of Kentucky, Lexington, KY 40506, USA
| | - Ashley W. Seifert
- Department of Biology, University of Kentucky, Lexington, KY 40506, USA
| |
Collapse
|
22
|
Medina-Gomez C, Mullin BH, Chesi A, Prijatelj V, Kemp JP, Shochat-Carvalho C, Trajanoska K, Wang C, Joro R, Evans TE, Schraut KE, Li-Gao R, Ahluwalia TS, Zillikens MC, Zhu K, Mook-Kanamori DO, Evans DS, Nethander M, Knol MJ, Thorleifsson G, Prokic I, Zemel B, Broer L, McGuigan FE, van Schoor NM, Reppe S, Pawlak MA, Ralston SH, van der Velde N, Lorentzon M, Stefansson K, Adams HHH, Wilson SG, Ikram MA, Walsh JP, Lakka TA, Gautvik KM, Wilson JF, Orwoll ES, van Duijn CM, Bønnelykke K, Uitterlinden AG, Styrkársdóttir U, Akesson KE, Spector TD, Tobias JH, Ohlsson C, Felix JF, Bisgaard H, Grant SFA, Richards JB, Evans DM, van der Eerden B, van de Peppel J, Ackert-Bicknell C, Karasik D, Kague E, Rivadeneira F. Bone mineral density loci specific to the skull portray potential pleiotropic effects on craniosynostosis. Commun Biol 2023; 6:691. [PMID: 37402774 PMCID: PMC10319806 DOI: 10.1038/s42003-023-04869-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 04/25/2023] [Indexed: 07/06/2023] Open
Abstract
Skull bone mineral density (SK-BMD) provides a suitable trait for the discovery of key genes in bone biology, particularly to intramembranous ossification, not captured at other skeletal sites. We perform a genome-wide association meta-analysis (n ~ 43,800) of SK-BMD, identifying 59 loci, collectively explaining 12.5% of the trait variance. Association signals cluster within gene-sets involved in skeletal development and osteoporosis. Among the four novel loci (ZIC1, PRKAR1A, AZIN1/ATP6V1C1, GLRX3), there are factors implicated in intramembranous ossification and as we show, inherent to craniosynostosis processes. Functional follow-up in zebrafish confirms the importance of ZIC1 on cranial suture patterning. Likewise, we observe abnormal cranial bone initiation that culminates in ectopic sutures and reduced BMD in mosaic atp6v1c1 knockouts. Mosaic prkar1a knockouts present asymmetric bone growth and, conversely, elevated BMD. In light of this evidence linking SK-BMD loci to craniofacial abnormalities, our study provides new insight into the pathophysiology, diagnosis and treatment of skeletal diseases.
Collapse
Grants
- UL1 TR000128 NCATS NIH HHS
- U01 AG042124 NIA NIH HHS
- U01 AG042145 NIA NIH HHS
- U01 AG042168 NIA NIH HHS
- U01 AG042140 NIA NIH HHS
- U24 AG051129 NIA NIH HHS
- R01 AR051124 NIAMS NIH HHS
- U01 AG027810 NIA NIH HHS
- U01 AR066160 NIAMS NIH HHS
- MC_UU_00007/10 Medical Research Council
- R01 HD058886 NICHD NIH HHS
- RC2 AR058973 NIAMS NIH HHS
- Wellcome Trust
- M01 RR000240 NCRR NIH HHS
- U01 AG042143 NIA NIH HHS
- UL1 RR026314 NCRR NIH HHS
- U01 AG042139 NIA NIH HHS
- EC | EU Framework Programme for Research and Innovation H2020 | H2020 Priority Excellent Science | H2020 European Research Council (H2020 Excellent Science - European Research Council)
- European Cooperation in Science and Technology (COST)
- Wellcome Trust (Wellcome)
- Department of Health | National Health and Medical Research Council (NHMRC)
- U.S. Department of Health & Human Services | NIH | Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD)
- ZonMw (Netherlands Organisation for Health Research and Development)
- EC | EC Seventh Framework Programm | FP7 Ideas: European Research Council (FP7-IDEAS-ERC - Specific Programme: "Ideas" Implementing the Seventh Framework Programme of the European Community for Research, Technological Development and Demonstration Activities (2007 to 2013))
- Vetenskapsrådet (Swedish Research Council)
- U.S. Department of Health & Human Services | NIH | National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS)
- Gouvernement du Canada | Canadian Institutes of Health Research (Instituts de Recherche en Santé du Canada)
- Nederlandse Organisatie voor Wetenschappelijk Onderzoek (Netherlands Organisation for Scientific Research)
- NCHA (Netherlands Consortium Healthy Ageing) Leiden/ Rotterdam; Dutch Ministry of Economic Affairs, Agriculture and Innovation (project KB-15-004-003); the Research Institute for Diseases in the Elderly [Netherlands] (014-93-015; RIDE2)
- Clinical and Translational Research Center (5-MO1-RR-000240 and UL1 RR-026314); U.S. Department of Health & Human Services | NIH | Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD) GrantRecipient="Au50"
- European Commission FP6 STRP grant number 018947 (LSHG-CT-2006-01947); Netherlands Organization for Scientific Research and the Russian Foundation for Basic Research (NWO-RFBR 047.017.043); Netherlands Brain Foundation (project number F2013(1)-28) GrantRecipient="Au40"
- Chief Scientist Office of the Scottish Government (CZB/4/276, CZB/4/710) GrantRecipient="Au28"
- Chief Scientist Office of the Scottish Government (CZB/4/276, CZB/4/710) GrantRecipient="Au38"
- The Pawsey Supercomputing Centre (with Funding from the Australian Government and the Government of Western Australia; PG 16/0162, PG 17/director2025) GrantRecipient="Au45”
- European Commission (EC)
- U.S. Department of Health & Human Services | NIH | National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS);NIH Roadmap for Medical Research [USA]: U01 AG027810, U01 AG042124, U01 AG042139, U01 AG042140, U01 AG042143, U01 AG042145, U01 AG042168, U01 AR066160, and UL1 TR000128 GrantRecipient="Au39”
- Versus Arthritis [USA] 21937 GrantRecipient="Au57”
Collapse
Affiliation(s)
- Carolina Medina-Gomez
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, 3000 CA, Rotterdam, The Netherlands
| | - Benjamin H Mullin
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Nedlands, WA, 6009, Australia
- School of Biomedical Sciences, University of Western Australia, Nedlands, WA, 6009, Australia
| | - Alessandra Chesi
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Vid Prijatelj
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, 3000 CA, Rotterdam, The Netherlands
| | - John P Kemp
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, QLD, 4102, Australia
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol, BS8 2BN, UK
| | | | - Katerina Trajanoska
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, 3000 CA, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, 3000 CA, Rotterdam, The Netherlands
| | - Carol Wang
- School of Women's and Infants' Health, University of Western Australia, Crawley, WA, 6009, Australia
| | - Raimo Joro
- Institute of Biomedicine, Physiology, University of Eastern Finland, Kuopio, 70211, Finland
| | - Tavia E Evans
- Department of Clinical Genetics, Erasmus MC, University Medical Center Rotterdam, 3000 CA, Rotterdam, The Netherlands
- Department of Radiology & Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, 3000 CA, Rotterdam, The Netherlands
| | - Katharina E Schraut
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, EH16 4UX, Scotland
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, EH8 9AG, Scotland
| | - Ruifang Li-Gao
- Department of Clinical Epidemiology, Leiden University Medical Centre, 2333 ZA, Leiden, The Netherlands
| | - Tarunveer S Ahluwalia
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, 2820, Denmark
- Steno Diabetes Center Copenhagen, Herlev, 2820, Denmark
- The Bioinformatics Center, Department of Biology, University of Copenhagen, Copenhagen, 2200, Denmark
| | - M Carola Zillikens
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, 3000 CA, Rotterdam, The Netherlands
| | - Kun Zhu
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Nedlands, WA, 6009, Australia
- Medical School, University of Western Australia, Perth, WA, 6009, Australia
| | - Dennis O Mook-Kanamori
- Department of Clinical Epidemiology, Leiden University Medical Centre, 2333 ZA, Leiden, The Netherlands
- Department of Public Health and Primary Care, Leiden University Medical Centre, 2333 ZA, Leiden, The Netherlands
| | - Daniel S Evans
- California Pacific Medical Center Research Institute, San Francisco, CA, 94107, USA
| | - Maria Nethander
- Bioinformatics Core Facility, Sahlgrenska Academy, University of Gothenburg, 413 90, Gothenburg, Sweden
- Center for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 90, Gothenburg, Sweden
| | - Maria J Knol
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, 3000 CA, Rotterdam, The Netherlands
| | | | - Ivana Prokic
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, 3000 CA, Rotterdam, The Netherlands
| | - Babette Zemel
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Division of GI, Hepatology, and Nutrition, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Linda Broer
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, 3000 CA, Rotterdam, The Netherlands
| | - Fiona E McGuigan
- Clinical and Molecular Osteoporosis Research Unit, Department of Clinical Sciences Malmö, Lund University, 205 02, Malmö, Sweden
| | - Natasja M van Schoor
- Department of Epidemiology and Data Science, Amsterdam UMC, 1081 HV, Amsterdam, The Netherlands
| | - Sjur Reppe
- Department of Plastic and Reconstructive Surgery, Oslo University Hospital, 0372, Oslo, Norway
- Department of Medical Biochemistry, Oslo University Hospital, 0372, Oslo, Norway
- Unger-Vetlesen Institute, Lovisenberg Diaconal Hospital, 0456, Oslo, Norway
| | - Mikolaj A Pawlak
- Department of Clinical Genetics, Erasmus MC, University Medical Center Rotterdam, 3000 CA, Rotterdam, The Netherlands
- Department of Neurology, Poznan University of Medical Sciences, 61-701, Poznan, Poland
| | - Stuart H Ralston
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU, Scotland
| | - Nathalie van der Velde
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, 3000 CA, Rotterdam, The Netherlands
- Department of Geriatric Medicine, Amsterdam Public Health Research Institute, Amsterdam UMC, 1105 AZ, Amsterdam, The Netherlands
| | - Mattias Lorentzon
- Center for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 90, Gothenburg, Sweden
- Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, VIC, 3000, Australia
| | | | - Hieab H H Adams
- Department of Clinical Genetics, Erasmus MC, University Medical Center Rotterdam, 3000 CA, Rotterdam, The Netherlands
- Department of Radiology & Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, 3000 CA, Rotterdam, The Netherlands
- Latin American Brain Health (BrainLat), Universidad Adolfo Ibáñez, Santiago, Chile
| | - Scott G Wilson
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Nedlands, WA, 6009, Australia
- School of Biomedical Sciences, University of Western Australia, Nedlands, WA, 6009, Australia
- Department of Twin Research & Genetic Epidemiology, King's College London, London, SE1 7EH, UK
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, 3000 CA, Rotterdam, The Netherlands
| | - John P Walsh
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Nedlands, WA, 6009, Australia
- Medical School, University of Western Australia, Perth, WA, 6009, Australia
| | - Timo A Lakka
- Institute of Biomedicine, Physiology, University of Eastern Finland, Kuopio, 70211, Finland
- Kuopio Research Institute of Exercise Medicine, Kuopio, 70100, Finland
- Department of Clinical Physiology and Nuclear Medicine, University of Eastern Finland, Kuopio, 70210, Finland
| | - Kaare M Gautvik
- Unger-Vetlesen Institute, Lovisenberg Diaconal Hospital, 0456, Oslo, Norway
| | - James F Wilson
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, EH16 4UX, Scotland
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU, Scotland
| | - Eric S Orwoll
- Department of Public Health & Preventive Medicine, Oregon Health & Science University, Portland, OR, OR97239, USA
| | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, 3000 CA, Rotterdam, The Netherlands
| | - Klaus Bønnelykke
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, 2820, Denmark
| | - Andre G Uitterlinden
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, 3000 CA, Rotterdam, The Netherlands
| | | | - Kristina E Akesson
- Clinical and Molecular Osteoporosis Research Unit, Department of Clinical Sciences Malmö, Lund University, 205 02, Malmö, Sweden
- Department of Orthopedics Malmö, Skåne University Hospital, S-21428, Malmö, Sweden
| | - Timothy D Spector
- Department of Twin Research & Genetic Epidemiology, King's College London, London, SE1 7EH, UK
| | - Jonathan H Tobias
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol, BS8 2BN, UK
- Musculoskeletal Research Unit, Translational Health Sciences, Bristol Medical School, Bristol, BS10 5NB, UK
| | - Claes Ohlsson
- Center for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 90, Gothenburg, Sweden
- Department of Drug Treatment, Sahlgrenska University Hospital, Region Västra Götaland, SE-413 45, Gothenburg, Sweden
| | - Janine F Felix
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, 3000 CA, Rotterdam, The Netherlands
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, 3000 CA, Rotterdam, The Netherlands
| | - Hans Bisgaard
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, 2820, Denmark
| | - Struan F A Grant
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Division of Endocrinology, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - J Brent Richards
- Department of Twin Research & Genetic Epidemiology, King's College London, London, SE1 7EH, UK
- Lady Davis Institute, Jewish General Hospital, Montreal, H3T 1E2, QC, Canada
| | - David M Evans
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, QLD, 4102, Australia
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol, BS8 2BN, UK
| | - Bram van der Eerden
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, 3000 CA, Rotterdam, The Netherlands
| | - Jeroen van de Peppel
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, 3000 CA, Rotterdam, The Netherlands
| | | | - David Karasik
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, 1311502, Israel
- Marcus Institute for Aging Research, Hebrew SeniorLife, Roslindale, MA, 02131, USA
| | - Erika Kague
- The School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences, University of Bristol, Bristol, BS8 1TD, UK
| | - Fernando Rivadeneira
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, 3000 CA, Rotterdam, The Netherlands.
| |
Collapse
|
23
|
Kemmler CL, Moran HR, Murray BF, Scoresby A, Klem JR, Eckert RL, Lepovsky E, Bertho S, Nieuwenhuize S, Burger S, D'Agati G, Betz C, Puller AC, Felker A, Ditrychova K, Bötschi S, Affolter M, Rohner N, Lovely CB, Kwan KM, Burger A, Mosimann C. Next-generation plasmids for transgenesis in zebrafish and beyond. Development 2023; 150:dev201531. [PMID: 36975217 PMCID: PMC10263156 DOI: 10.1242/dev.201531] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 03/10/2023] [Indexed: 03/29/2023]
Abstract
Transgenesis is an essential technique for any genetic model. Tol2-based transgenesis paired with Gateway-compatible vector collections has transformed zebrafish transgenesis with an accessible modular system. Here, we establish several next-generation transgenesis tools for zebrafish and other species to expand and enhance transgenic applications. To facilitate gene regulatory element testing, we generated Gateway middle entry vectors harboring the small mouse beta-globin minimal promoter coupled to several fluorophores, CreERT2 and Gal4. To extend the color spectrum for transgenic applications, we established middle entry vectors encoding the bright, blue-fluorescent protein mCerulean and mApple as an alternative red fluorophore. We present a series of p2A peptide-based 3' vectors with different fluorophores and subcellular localizations to co-label cells expressing proteins of interest. Finally, we established Tol2 destination vectors carrying the zebrafish exorh promoter driving different fluorophores as a pineal gland-specific transgenesis marker that is active before hatching and through adulthood. exorh-based reporters and transgenesis markers also drive specific pineal gland expression in the eye-less cavefish (Astyanax). Together, our vectors provide versatile reagents for transgenesis applications in zebrafish, cavefish and other models.
Collapse
Affiliation(s)
- Cassie L. Kemmler
- University of Colorado, School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, 12801 E 17th Avenue, Aurora, CO 80045, USA
| | - Hannah R. Moran
- University of Colorado, School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, 12801 E 17th Avenue, Aurora, CO 80045, USA
| | - Brooke F. Murray
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Aaron Scoresby
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - John R. Klem
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Rachel L. Eckert
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Elizabeth Lepovsky
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Sylvain Bertho
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Susan Nieuwenhuize
- University of Colorado, School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, 12801 E 17th Avenue, Aurora, CO 80045, USA
- Department of Molecular Life Sciences, University of Zurich, 8057 Zürich, Switzerland
| | - Sibylle Burger
- Department of Molecular Life Sciences, University of Zurich, 8057 Zürich, Switzerland
| | - Gianluca D'Agati
- Department of Molecular Life Sciences, University of Zurich, 8057 Zürich, Switzerland
| | - Charles Betz
- Growth & Development, Biozentrum, Spitalstrasse 41, University of Basel, 4056 Basel, Switzerland
| | - Ann-Christin Puller
- Department of Molecular Life Sciences, University of Zurich, 8057 Zürich, Switzerland
| | - Anastasia Felker
- Department of Molecular Life Sciences, University of Zurich, 8057 Zürich, Switzerland
| | - Karolina Ditrychova
- Department of Molecular Life Sciences, University of Zurich, 8057 Zürich, Switzerland
| | - Seraina Bötschi
- Department of Molecular Life Sciences, University of Zurich, 8057 Zürich, Switzerland
| | - Markus Affolter
- Growth & Development, Biozentrum, Spitalstrasse 41, University of Basel, 4056 Basel, Switzerland
| | - Nicolas Rohner
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - C. Ben Lovely
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Kristen M. Kwan
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Alexa Burger
- University of Colorado, School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, 12801 E 17th Avenue, Aurora, CO 80045, USA
| | - Christian Mosimann
- University of Colorado, School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, 12801 E 17th Avenue, Aurora, CO 80045, USA
| |
Collapse
|
24
|
Sapède D, Bahraoui S, Abou Nassif L, Barthelaix A, Mathieu M, Jorgensen C, Djouad F. Cartilage regeneration in zebrafish depends on Nrg1/ErbB signaling pathway. Front Cell Dev Biol 2023; 11:1123299. [PMID: 37215080 PMCID: PMC10192884 DOI: 10.3389/fcell.2023.1123299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 04/19/2023] [Indexed: 05/24/2023] Open
Abstract
Objective: Cartilage, as the majority of adult mammalian tissues, has limited regeneration capacity. Cartilage degradation consecutive to joint injury or aging then leads to irreversible joint damage and diseases. In contrast, several vertebrate species such as the zebrafish have the remarkable capacity to spontaneously regenerate skeletal structures after severe injuries. The objective of our study was to test the regenerative capacity of Meckel's cartilage (MC) upon mechanical injury in zebrafish and to identify the mechanisms underlying this process. Methods and Results: Cartilage regenerative capacity in zebrafish larvae was investigated after mechanical injuries of the lower jaw MC in TgBAC(col2a1a:mCherry), to visualize the loss and recovery of cartilage. Confocal analysis revealed the formation of new chondrocytes and complete regeneration of MC at 14 days post-injury (dpi) via chondrocyte cell cycle re-entry and proliferation of pre-existing MC chondrocytes near the wound. Through expression analyses, we showed an increase of nrg1 expression in the regenerating lower jaw, which also expresses Nrg1 receptors, ErbB3 and ErbB2. Pharmacological inhibition of the ErbB pathway and specific knockdown of Nrg1 affected MC regeneration indicating the pivotal role of this pathway for cartilage regeneration. Finally, addition of exogenous NRG1 in an in vitro model of osteoarthritic (OA)-like chondrocytes induced by IL1β suggests that Nrg1/ErbB pathway is functional in mammalian chondrocytes and alleviates the increased expression of catabolic markers characteristic of OA-like chondrocytes. Conclusion: Our results show that the Nrg1/ErbB pathway is required for spontaneous cartilage regeneration in zebrafish and is of interest to design new therapeutic approaches to promote cartilage regeneration in mammals.
Collapse
Affiliation(s)
- Dora Sapède
- IRMB, University Montpellier, INSERM, Montpellier, France
| | - Sarah Bahraoui
- IRMB, University Montpellier, INSERM, Montpellier, France
| | | | | | - Marc Mathieu
- IRMB, University Montpellier, INSERM, Montpellier, France
| | - Christian Jorgensen
- IRMB, University Montpellier, INSERM, Montpellier, France
- CHU Montpellier, Montpellier, France
| | - Farida Djouad
- IRMB, University Montpellier, INSERM, Montpellier, France
| |
Collapse
|
25
|
Anderson T, Mo J, Gagarin E, Sherwood D, Blumenkrantz M, Mao E, Leon G, Chen HJ, Tseng KC, Fabian P, Crump JG, Smeeton J. Ligament injury in adult zebrafish triggers ECM remodeling and cell dedifferentiation for scar-free regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.03.527039. [PMID: 36778403 PMCID: PMC9915717 DOI: 10.1101/2023.02.03.527039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
After traumatic injury, healing of mammalian ligaments is typically associated with fibrotic scarring as opposed to scar-free regeneration. In contrast, here we show that the ligament supporting the jaw joint of adult zebrafish is capable of rapid and complete scar-free healing. Following surgical transection of the jaw joint ligament, we observe breakdown of ligament tissue adjacent to the cut sites, expansion of mesenchymal tissue within the wound site, and then remodeling of extracellular matrix (ECM) to a normal ligament morphology. Lineage tracing of mature ligamentocytes following transection shows that they dedifferentiate, undergo cell cycle re-entry, and contribute to the regenerated ligament. Single-cell RNA sequencing of the regenerating ligament reveals dynamic expression of ECM genes in neural-crest-derived mesenchymal cells, as well as diverse immune cells expressing the endopeptidase-encoding gene legumain . Analysis of legumain mutant zebrafish shows a requirement for early ECM remodeling and efficient ligament regeneration. Our study establishes a new model of adult scar-free ligament regeneration and highlights roles of immune-mesenchyme cross-talk in ECM remodeling that initiates regeneration. Highlights Rapid regeneration of the jaw joint ligament in adult zebrafishDedifferentiation of mature ligamentocytes contributes to regenerationscRNAseq reveals dynamic ECM remodeling and immune activation during regenerationRequirement of Legumain for ECM remodeling and ligament healing.
Collapse
Affiliation(s)
- Troy Anderson
- Columbia Stem Cell Initiative, Department of Rehabilitation and Regenerative Medicine, and Department of Genetics and Development, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA
| | - Julia Mo
- Columbia Stem Cell Initiative, Department of Rehabilitation and Regenerative Medicine, and Department of Genetics and Development, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA
| | - Ernesto Gagarin
- Columbia Stem Cell Initiative, Department of Rehabilitation and Regenerative Medicine, and Department of Genetics and Development, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA
| | - Desmarie Sherwood
- Columbia Stem Cell Initiative, Department of Rehabilitation and Regenerative Medicine, and Department of Genetics and Development, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA
| | - Maria Blumenkrantz
- Columbia Stem Cell Initiative, Department of Rehabilitation and Regenerative Medicine, and Department of Genetics and Development, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA
| | - Eric Mao
- Columbia Stem Cell Initiative, Department of Rehabilitation and Regenerative Medicine, and Department of Genetics and Development, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA
- Department of Biological Sciences, Columbia College, Columbia University NY 10027, USA
| | - Gianna Leon
- Columbia Stem Cell Initiative, Department of Rehabilitation and Regenerative Medicine, and Department of Genetics and Development, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA
- Packer Collegiate Institute, New York, NY 11201, USA
| | - Hung-Jhen Chen
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Kuo-Chang Tseng
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Peter Fabian
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - J. Gage Crump
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Joanna Smeeton
- Columbia Stem Cell Initiative, Department of Rehabilitation and Regenerative Medicine, and Department of Genetics and Development, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA
| |
Collapse
|
26
|
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
|
27
|
Brown TL, Horton EC, Craig EW, Goo CEA, Black EC, Hewitt MN, Yee NG, Fan ET, Raible DW, Rasmussen JP. Dermal appendage-dependent patterning of zebrafish atoh1a+ Merkel cells. eLife 2023; 12:85800. [PMID: 36648063 PMCID: PMC9901935 DOI: 10.7554/elife.85800] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Touch system function requires precise interactions between specialized skin cells and somatosensory axons, as exemplified by the vertebrate mechanosensory Merkel cell-neurite complex. Development and patterning of Merkel cells and associated neurites during skin organogenesis remain poorly understood, partly due to the in utero development of mammalian embryos. Here, we discover Merkel cells in the zebrafish epidermis and identify Atonal homolog 1a (Atoh1a) as a marker of zebrafish Merkel cells. We show that zebrafish Merkel cells derive from basal keratinocytes, express neurosecretory and mechanosensory machinery, extend actin-rich microvilli, and complex with somatosensory axons, all hallmarks of mammalian Merkel cells. Merkel cells populate all major adult skin compartments, with region-specific densities and distribution patterns. In vivo photoconversion reveals that Merkel cells undergo steady loss and replenishment during skin homeostasis. Merkel cells develop concomitant with dermal appendages along the trunk and loss of Ectodysplasin signaling, which prevents dermal appendage formation, reduces Merkel cell density by affecting cell differentiation. By contrast, altering dermal appendage morphology changes the distribution, but not density, of Merkel cells. Overall, our studies provide insights into touch system maturation during skin organogenesis and establish zebrafish as an experimentally accessible in vivo model for the study of Merkel cell biology.
Collapse
Affiliation(s)
- Tanya L Brown
- Department of Biology, University of WashingtonSeattleUnited States
| | - Emma C Horton
- Department of Biology, University of WashingtonSeattleUnited States
| | - Evan W Craig
- Department of Biology, University of WashingtonSeattleUnited States
| | - Camille EA Goo
- Department of Biology, University of WashingtonSeattleUnited States
| | - Erik C Black
- Department of Biology, University of WashingtonSeattleUnited States
- Molecular and Cellular Biology Program, University of WashingtonSeattleUnited States
| | - Madeleine N Hewitt
- Molecular and Cellular Biology Program, University of WashingtonSeattleUnited States
- Department of Biological Structure, University of WashingtonSeattleUnited States
| | - Nathaniel G Yee
- Department of Biology, University of WashingtonSeattleUnited States
| | - Everett T Fan
- Department of Biology, University of WashingtonSeattleUnited States
| | - David W Raible
- Department of Biological Structure, University of WashingtonSeattleUnited States
- Department of Otolaryngology - Head and Neck Surgery, University of WashingtonSeattleUnited States
- Institute for Stem Cell and Regenerative Medicine, University of WashingtonSeattleUnited States
| | - Jeffrey P Rasmussen
- Department of Biology, University of WashingtonSeattleUnited States
- Institute for Stem Cell and Regenerative Medicine, University of WashingtonSeattleUnited States
| |
Collapse
|
28
|
Xia Y, Duca S, Perder B, Dündar F, Zumbo P, Qiu M, Yao J, Cao Y, Harrison MRM, Zangi L, Betel D, Cao J. Activation of a transient progenitor state in the epicardium is required for zebrafish heart regeneration. Nat Commun 2022; 13:7704. [PMID: 36513650 PMCID: PMC9747719 DOI: 10.1038/s41467-022-35433-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 12/02/2022] [Indexed: 12/15/2022] Open
Abstract
The epicardium, a mesothelial cell tissue that encompasses vertebrate hearts, supports heart regeneration after injury through paracrine effects and as a source of multipotent progenitors. However, the progenitor state in the adult epicardium has yet to be defined. Through single-cell RNA-sequencing of isolated epicardial cells from uninjured and regenerating adult zebrafish hearts, we define the epithelial and mesenchymal subsets of the epicardium. We further identify a transiently activated epicardial progenitor cell (aEPC) subpopulation marked by ptx3a and col12a1b expression. Upon cardiac injury, aEPCs emerge from the epithelial epicardium, migrate to enclose the wound, undergo epithelial-mesenchymal transition (EMT), and differentiate into mural cells and pdgfra+hapln1a+ mesenchymal epicardial cells. These EMT and differentiation processes are regulated by the Tgfβ pathway. Conditional ablation of aEPCs blocks heart regeneration through reduced nrg1 expression and mesenchymal cell number. Our findings identify a transient progenitor population of the adult epicardium that is indispensable for heart regeneration and highlight it as a potential target for enhancing cardiac repair.
Collapse
Affiliation(s)
- Yu Xia
- Cardiovascular Research Institute, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
- Department of Cell and Developmental Biology, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
| | - Sierra Duca
- Cardiovascular Research Institute, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
- Department of Cell and Developmental Biology, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
| | - Björn Perder
- Cardiovascular Research Institute, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
- Department of Cell and Developmental Biology, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
| | - Friederike Dündar
- Department of Physiology and Biophysics, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
- Applied Bioinformatics Core, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
| | - Paul Zumbo
- Department of Physiology and Biophysics, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
- Applied Bioinformatics Core, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
| | - Miaoyan Qiu
- Cardiovascular Research Institute, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
- Department of Cell and Developmental Biology, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
| | - Jun Yao
- Cardiovascular Research Institute, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
- Department of Cell and Developmental Biology, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
| | - Yingxi Cao
- Cardiovascular Research Institute, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
- Department of Cell and Developmental Biology, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
| | - Michael R M Harrison
- Cardiovascular Research Institute, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
- Department of Cell and Developmental Biology, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
| | - Lior Zangi
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Doron Betel
- Applied Bioinformatics Core, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
- Division of Hematology and Oncology, Department of Medicine, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
- Institute for Computational Biomedicine, Department of Medicine, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
| | - Jingli Cao
- Cardiovascular Research Institute, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA.
- Department of Cell and Developmental Biology, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA.
| |
Collapse
|
29
|
Fin ray branching is defined by TRAP + osteolytic tubules in zebrafish. Proc Natl Acad Sci U S A 2022; 119:e2209231119. [PMID: 36417434 PMCID: PMC9889879 DOI: 10.1073/pnas.2209231119] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The shaping of bone structures relies on various cell types and signaling pathways. Here, we use the zebrafish bifurcating fin rays during regeneration to investigate bone patterning. We found that the regenerating fin rays form via two mineralization fronts that undergo an osteoblast-dependent fusion/stitching until the branchpoint, and that bifurcation is not simply the splitting of one unit into two. We identified tartrate-resistant acid phosphatase-positive osteolytic tubular structures at the branchpoints, hereafter named osteolytic tubules (OLTs). Chemical inhibition of their bone-resorbing activity strongly impairs ray bifurcation, indicating that OLTs counteract the stitching process. Furthermore, by testing different osteoactive compounds, we show that the position of the branchpoint depends on the balance between bone mineralization and resorption activities. Overall, these findings provide a unique perspective on fin ray formation and bifurcation, and reveal a key role for OLTs in defining the proximo-distal position of the branchpoint.
Collapse
|
30
|
Saelens JW, Sweeney MI, Viswanathan G, Xet-Mull AM, Jurcic Smith KL, Sisk DM, Hu DD, Cronin RM, Hughes EJ, Brewer WJ, Coers J, Champion MM, Champion PA, Lowe CB, Smith CM, Lee S, Stout JE, Tobin DM. An ancestral mycobacterial effector promotes dissemination of infection. Cell 2022; 185:4507-4525.e18. [PMID: 36356582 PMCID: PMC9691622 DOI: 10.1016/j.cell.2022.10.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 08/27/2022] [Accepted: 10/16/2022] [Indexed: 11/11/2022]
Abstract
The human pathogen Mycobacterium tuberculosis typically causes lung disease but can also disseminate to other tissues. We identified a M. tuberculosis (Mtb) outbreak presenting with unusually high rates of extrapulmonary dissemination and bone disease. We found that the causal strain carried an ancestral full-length version of the type VII-secreted effector EsxM rather than the truncated version present in other modern Mtb lineages. The ancestral EsxM variant exacerbated dissemination through enhancement of macrophage motility, increased egress of macrophages from established granulomas, and alterations in macrophage actin dynamics. Reconstitution of the ancestral version of EsxM in an attenuated modern strain of Mtb altered the migratory mode of infected macrophages, enhancing their motility. In a zebrafish model, full-length EsxM promoted bone disease. The presence of a derived nonsense variant in EsxM throughout the major Mtb lineages 2, 3, and 4 is consistent with a role for EsxM in regulating the extent of dissemination.
Collapse
Affiliation(s)
- Joseph W Saelens
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Mollie I Sweeney
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Gopinath Viswanathan
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ana María Xet-Mull
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Kristen L Jurcic Smith
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Dana M Sisk
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Daniel D Hu
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Rachel M Cronin
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Erika J Hughes
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - W Jared Brewer
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Jörn Coers
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA; Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Matthew M Champion
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA; Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Patricia A Champion
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA; Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Craig B Lowe
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Clare M Smith
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Sunhee Lee
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Jason E Stout
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA; Division of Infectious Diseases and International Health, Duke University School of Medicine, Durham, NC 27710, USA.
| | - David M Tobin
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA; Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA.
| |
Collapse
|
31
|
Cao Z, Liu G, Zhang H, Wang M, Xu Y. Nox4 promotes osteoblast differentiation through TGF-beta signal pathway. Free Radic Biol Med 2022; 193:595-609. [PMID: 36372285 DOI: 10.1016/j.freeradbiomed.2022.11.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/08/2022] [Accepted: 11/09/2022] [Indexed: 11/12/2022]
Abstract
NADPH oxidase 4 (Nox4) is the main source of reactive oxygen species, which promote osteoclast formation and lead to bone loss, thereby causing osteoporosis. However, the role of Nox4 in osteoblasts during early development remains unclear. We used zebrafish to study the effect of Nox4 deletion on bone mineralization in early development. nox4-/- zebrafish showed decreased bone mineralization during early development and significantly reduced numbers of osteoblasts, osteoclasts, and chondrocytes. Transcriptome sequencing showed that the TGF-β signaling pathway was significantly disrupted in nox4-/- zebrafish. Inhibiting TGF-β signaling rescued the abnormal bone development caused by nox4 deletion and increased the number of osteoblasts. We used Saos-2 human osteosarcoma cells to confirm our results, which clarified the role of Nox4 in human osteoblasts. Our results demonstrate the mechanism of reduced bone mineralization in early development and provide a basis for the clinical treatment of osteoporosis.
Collapse
Affiliation(s)
- Zihou Cao
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Gongwen Liu
- Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, China
| | - Hui Zhang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Mingyong Wang
- Murui Biological Technology Co., Ltd., Suzhou Industrial Park, No.11 Jinpu Road, Suzhou, China.
| | - Youjia Xu
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
32
|
Brandão AS, Borbinha J, Pereira T, Brito PH, Lourenço R, Bensimon-Brito A, Jacinto A. A regeneration-triggered metabolic adaptation is necessary for cell identity transitions and cell cycle re-entry to support blastema formation and bone regeneration. eLife 2022; 11:e76987. [PMID: 35993337 PMCID: PMC9395193 DOI: 10.7554/elife.76987] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 07/29/2022] [Indexed: 11/29/2022] Open
Abstract
Regeneration depends on the ability of mature cells at the injury site to respond to injury, generating tissue-specific progenitors that incorporate the blastema and proliferate to reconstitute the original organ architecture. The metabolic microenvironment has been tightly connected to cell function and identity during development and tumorigenesis. Yet, the link between metabolism and cell identity at the mechanistic level in a regenerative context remains unclear. The adult zebrafish caudal fin, and bone cells specifically, have been crucial for the understanding of mature cell contribution to tissue regeneration. Here, we use this model to explore the relevance of glucose metabolism for the cell fate transitions preceding new osteoblast formation and blastema assembly. We show that injury triggers a modulation in the metabolic profile at early stages of regeneration to enhance glycolysis at the expense of mitochondrial oxidation. This metabolic adaptation mediates transcriptional changes that make mature osteoblast amenable to be reprogramed into pre-osteoblasts and induces cell cycle re-entry and progression. Manipulation of the metabolic profile led to severe reduction of the pre-osteoblast pool, diminishing their capacity to generate new osteoblasts, and to a complete abrogation of blastema formation. Overall, our data indicate that metabolic alterations have a powerful instructive role in regulating genetic programs that dictate fate decisions and stimulate proliferation, thereby providing a deeper understanding on the mechanisms regulating blastema formation and bone regeneration.
Collapse
Affiliation(s)
- Ana S Brandão
- CEDOC, NOVA Medical School, Universidade Nova de LisboaLisbonPortugal
| | - Jorge Borbinha
- CEDOC, NOVA Medical School, Universidade Nova de LisboaLisbonPortugal
| | - Telmo Pereira
- CEDOC, NOVA Medical School, Universidade Nova de LisboaLisbonPortugal
| | - Patrícia H Brito
- UCIBIO, Dept. Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de LisboaLisbonPortugal
| | - Raquel Lourenço
- CEDOC, NOVA Medical School, Universidade Nova de LisboaLisbonPortugal
| | | | - Antonio Jacinto
- CEDOC, NOVA Medical School, Universidade Nova de LisboaLisbonPortugal
| |
Collapse
|
33
|
Hui SP, Sugimoto K, Sheng DZ, Kikuchi K. Regulatory T cells regulate blastemal proliferation during zebrafish caudal fin regeneration. Front Immunol 2022; 13:981000. [PMID: 36059461 PMCID: PMC9429828 DOI: 10.3389/fimmu.2022.981000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 07/27/2022] [Indexed: 11/26/2022] Open
Abstract
The role of T cells in appendage regeneration remains unclear. In this study, we revealed an important role for regulatory T cells (Tregs), a subset of T cells that regulate tolerance and tissue repair, in the epimorphic regeneration of zebrafish caudal fin tissue. Upon amputation, fin tissue-resident Tregs infiltrate into the blastema, a population of progenitor cells that produce new fin tissues. Conditional genetic ablation of Tregs attenuates blastemal cell proliferation during fin regeneration. Blastema-infiltrating Tregs upregulate the expression of igf2a and igf2b, and pharmacological activation of IGF signaling restores blastemal proliferation in Treg-ablated zebrafish. These findings further extend our understandings of Treg function in tissue regeneration and repair.
Collapse
Affiliation(s)
- Subhra P. Hui
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- S. N. Pradhan Centre for Neurosciences, University of Calcutta, Kolkata, West Bengal, India
- *Correspondence: Subhra P. Hui, ; Kazu Kikuchi,
| | - Kotaro Sugimoto
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- Department of Basic Pathology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Delicia Z. Sheng
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
| | - Kazu Kikuchi
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- St. Vincent’s Clinical School, University of New South Wales, Kensington, NSW, Australia
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
- *Correspondence: Subhra P. Hui, ; Kazu Kikuchi,
| |
Collapse
|
34
|
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
|
35
|
Sun J, Peterson EA, Jiao C, Chen X, Zhao Y, Wang J. Zebrafish heart regeneration after coronary dysfunction-induced cardiac damage. Dev Biol 2022; 487:57-66. [PMID: 35490764 PMCID: PMC11017783 DOI: 10.1016/j.ydbio.2022.04.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 04/21/2022] [Accepted: 04/25/2022] [Indexed: 11/03/2022]
Abstract
Over the past 20 years, various zebrafish injury models demonstrated efficient heart regeneration after cardiac tissue loss. However, no established coronary vessel injury methods exist in the zebrafish model, despite coronary endothelial dysfunction occurring in most patients with acute coronary syndrome. This is due to difficulties performing surgery on small coronary vessels and a lack of genetic tools to precisely manipulate coronary cells in zebrafish. We determined that the Notch ligand gene deltaC regulatory sequences drive gene expression in zebrafish coronary endothelial cells, enabling us to overcome these obstacles. We created a deltaC fluorescent reporter line and visualized robust coronary growth during heart development and regeneration. Importantly, this reporter facilitated the visualization of coronary growth without an endocardial background. Moreover, we visualized robust coronary growth on the surface of juvenile hearts and regrowth in the wounded area of adult hearts ex vivo. With this approach, we observed growth inhibition by reported vascular growth antagonists of the VEGF, EGF and Notch signaling pathways. Furthermore, we established a coronary genetic ablation system and observed that severe coronary endothelial cell loss resulted in fish death, whereas fish survived mild coronary cell loss. Coronary cell depletion triggered regenerative responses, which resulted in the restoration of damaged cardiac tissues within several weeks. Overall, our work demonstrated the efficacy of using deltaC regulatory elements for high-resolution visualization of the coronary endothelium; screening small molecules for coronary growth effects; and revealed complete recovery in adult zebrafish after coronary-induced heart damage.
Collapse
Affiliation(s)
- Jisheng Sun
- Division of Cardiology, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Elizabeth A Peterson
- Division of Cardiology, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Cheng Jiao
- Division of Cardiology, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Xin Chen
- Division of Cardiology, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Yun Zhao
- Division of Cardiology, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Jinhu Wang
- Division of Cardiology, School of Medicine, Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
36
|
Sehring IM, Mohammadi HF, Haffner-Luntzer M, Ignatius A, Huber-Lang M, Weidinger G. Zebrafish fin regeneration involves generic and regeneration-specific osteoblast injury responses. eLife 2022; 11:77614. [PMID: 35748539 PMCID: PMC9259016 DOI: 10.7554/elife.77614] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 06/23/2022] [Indexed: 11/13/2022] Open
Abstract
Successful regeneration requires the coordinated execution of multiple cellular responses to injury. In amputated zebrafish fins, mature osteoblasts dedifferentiate, migrate towards the injury and form proliferative osteogenic blastema cells. We show that osteoblast migration is preceded by cell elongation and alignment along the proximodistal axis, which require actomyosin, but not microtubule turnover. Surprisingly, osteoblast dedifferentiation and migration can be uncoupled. Using pharmacological and genetic interventions, we found that NF-ĸB and retinoic acid signalling regulate dedifferentiation without affecting migration, while the complement system and actomyosin dynamics affect migration but not dedifferentiation. Furthermore, by removing bone at two locations within a fin ray, we established an injury model containing two injury sites. We found that osteoblasts dedifferentiate at and migrate towards both sites, while accumulation of osteogenic progenitor cells and regenerative bone formation only occur at the distal-facing injury. Together, these data indicate that osteoblast dedifferentiation and migration represent generic injury responses that are differentially regulated and can occur independently of each other and of regenerative growth. We conclude that successful fin bone regeneration appears to involve the coordinated execution of generic and regeneration-specific responses of osteoblasts to injury.
Collapse
Affiliation(s)
| | | | | | - Anita Ignatius
- Institute of Orthopaedic Research and Biomechanics, University Hospital Ulm, Ulm, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology (ITI), University Hospital Ulm, Ulm, Germany
| | - Gilbert Weidinger
- Institute of Biochemistry and Molecular Biology, University of Ulm, Ulm, Germany
| |
Collapse
|
37
|
Das RN, Tevet Y, Safriel S, Han Y, Moshe N, Lambiase G, Bassi I, Nicenboim J, Brückner M, Hirsch D, Eilam-Altstadter R, Herzog W, Avraham R, Poss KD, Yaniv K. Generation of specialized blood vessels via lymphatic transdifferentiation. Nature 2022; 606:570-575. [PMID: 35614218 PMCID: PMC9875863 DOI: 10.1038/s41586-022-04766-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 04/14/2022] [Indexed: 01/27/2023]
Abstract
The lineage and developmental trajectory of a cell are key determinants of cellular identity. In the vascular system, endothelial cells (ECs) of blood and lymphatic vessels differentiate and specialize to cater to the unique physiological demands of each organ1,2. Although lymphatic vessels were shown to derive from multiple cellular origins, lymphatic ECs (LECs) are not known to generate other cell types3,4. Here we use recurrent imaging and lineage-tracing of ECs in zebrafish anal fins, from early development to adulthood, to uncover a mechanism of specialized blood vessel formation through the transdifferentiation of LECs. Moreover, we demonstrate that deriving anal-fin vessels from lymphatic versus blood ECs results in functional differences in the adult organism, uncovering a link between cell ontogeny and functionality. We further use single-cell RNA-sequencing analysis to characterize the different cellular populations and transition states involved in the transdifferentiation process. Finally, we show that, similar to normal development, the vasculature is rederived from lymphatics during anal-fin regeneration, demonstrating that LECs in adult fish retain both potency and plasticity for generating blood ECs. Overall, our research highlights an innate mechanism of blood vessel formation through LEC transdifferentiation, and provides in vivo evidence for a link between cell ontogeny and functionality in ECs.
Collapse
Affiliation(s)
- Rudra N. Das
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel, Corresponding Authors Karina Yaniv Department of Biological Regulation, Weizmann Institute of Science, Rehovot, 76100, Israel, , Rudra N. Das Department of Biological Regulation, Weizmann Institute of Science, Rehovot, 76100, Israel,
| | - Yaara Tevet
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Stav Safriel
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Yanchao Han
- Duke Regeneration Center, Department of Cell Biology, Duke University School of Medicine, Durham, United States, Institute for Cardiovascular Science, Medical College, Soochow University, Suzhou, China
| | - Noga Moshe
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Giuseppina Lambiase
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Ivan Bassi
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Julian Nicenboim
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Matthias Brückner
- University of Muenster and Max Plank Institute for Molecular Biomedicine, Muenster, Germany
| | - Dana Hirsch
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | | | - Wiebke Herzog
- University of Muenster and Max Plank Institute for Molecular Biomedicine, Muenster, Germany
| | - Roi Avraham
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Kenneth D. Poss
- Duke Regeneration Center, Department of Cell Biology, Duke University School of Medicine, Durham, United States
| | - Karina Yaniv
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel, Corresponding Authors Karina Yaniv Department of Biological Regulation, Weizmann Institute of Science, Rehovot, 76100, Israel, , Rudra N. Das Department of Biological Regulation, Weizmann Institute of Science, Rehovot, 76100, Israel,
| |
Collapse
|
38
|
Carletti A, Cardoso C, Lobo-Arteaga J, Sales S, Juliao D, Ferreira I, Chainho P, Dionísio MA, Gaudêncio MJ, Afonso C, Lourenço H, Cancela ML, Bandarra NM, Gavaia PJ. Antioxidant and Anti-inflammatory Extracts From Sea Cucumbers and Tunicates Induce a Pro-osteogenic Effect in Zebrafish Larvae. Front Nutr 2022; 9:888360. [PMID: 35614979 PMCID: PMC9125325 DOI: 10.3389/fnut.2022.888360] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 03/31/2022] [Indexed: 12/24/2022] Open
Abstract
Bone metabolic disorders such as osteoporosis are characterized by the loss of mineral from the bone tissue leading to its structural weakening and increased susceptibility to fractures. A growing body of evidence suggests that inflammation and oxidative stress play an important role in the pathophysiological processes involved in the rise of these conditions. As the currently available therapeutic strategies are often characterized by toxic effects associated with their long-term use, natural antioxidants and anti-inflammatory compounds such as polyphenols promise to be a valuable alternative for the prevention and treatment of these disorders. In this scope, the marine environment is becoming an important source of bioactive compounds with potential pharmacological applications. Here, we explored the bioactive potential of three species of holothurians (Echinodermata) and four species of tunicates (Chordata) as sources of antioxidant and anti-inflammatory compounds with a particular focus on polyphenolic substances. Hydroethanolic and aqueous extracts were obtained from animals' biomass and screened for their content of polyphenols and their antioxidant and anti-inflammatory properties. Hydroethanolic fractions of three species of tunicates displayed high polyphenolic content associated with strong antioxidant potential and anti-inflammatory activity. Extracts were thereafter tested for their capacity to promote bone formation and mineralization by applying an assay that uses the developing operculum of zebrafish (Danio rerio) to assess the osteogenic activity of compounds. The same three hydroethanolic fractions from tunicates were characterized by a strong in vivo osteogenic activity, which positively correlated with their anti-inflammatory potential as measured by COX-2 inhibition. This study highlights the therapeutic potential of polyphenol-rich hydroethanolic extracts obtained from three species of tunicates as a substrate for the development of novel drugs for the treatment of bone disorders correlated to oxidative stress and inflammatory processes.
Collapse
Affiliation(s)
- Alessio Carletti
- Faculty of Biomedical Sciences and Medicine (FCBM), University of Algarve, Faro, Portugal
- Centre of Marine Sciences, University of Algarve, Faro, Portugal
| | - Carlos Cardoso
- Division of Aquaculture, Upgrading and Bioprospection, Portuguese Institute for the Sea and Atmosphere (IPMA), Algés, Portugal
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto, Porto, Portugal
| | - Jorge Lobo-Arteaga
- Division of Environmental Oceanography, Portuguese Institute for the Sea and Atmosphere, Algés, Portugal
- Marine and Environmental Sciences Centre (MARE), NOVA University of Lisbon, Lisbon, Portugal
| | - Sabrina Sales
- Division of Aquaculture, Upgrading and Bioprospection, Portuguese Institute for the Sea and Atmosphere (IPMA), Algés, Portugal
| | - Diana Juliao
- Division of Aquaculture, Upgrading and Bioprospection, Portuguese Institute for the Sea and Atmosphere (IPMA), Algés, Portugal
| | - Inês Ferreira
- Division of Aquaculture, Upgrading and Bioprospection, Portuguese Institute for the Sea and Atmosphere (IPMA), Algés, Portugal
| | - Paula Chainho
- Marine and Environmental Sciences Centre (MARE), NOVA University of Lisbon, Lisbon, Portugal
| | - Maria Ana Dionísio
- Marine and Environmental Sciences Centre (MARE), NOVA University of Lisbon, Lisbon, Portugal
| | - Maria J. Gaudêncio
- Division of Environmental Oceanography, Portuguese Institute for the Sea and Atmosphere, Algés, Portugal
| | - Cláudia Afonso
- Division of Aquaculture, Upgrading and Bioprospection, Portuguese Institute for the Sea and Atmosphere (IPMA), Algés, Portugal
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto, Porto, Portugal
| | - Helena Lourenço
- Division of Aquaculture, Upgrading and Bioprospection, Portuguese Institute for the Sea and Atmosphere (IPMA), Algés, Portugal
| | - M. Leonor Cancela
- Faculty of Biomedical Sciences and Medicine (FCBM), University of Algarve, Faro, Portugal
- Centre of Marine Sciences, University of Algarve, Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve, Faro, Portugal
- Centre for BioMedical Research (CBMR), University of Algarve, Faro, Portugal
| | - Narcisa M. Bandarra
- Division of Aquaculture, Upgrading and Bioprospection, Portuguese Institute for the Sea and Atmosphere (IPMA), Algés, Portugal
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto, Porto, Portugal
| | - Paulo J. Gavaia
- Faculty of Biomedical Sciences and Medicine (FCBM), University of Algarve, Faro, Portugal
- Centre of Marine Sciences, University of Algarve, Faro, Portugal
| |
Collapse
|
39
|
Probiotics Enhance Bone Growth and Rescue BMP Inhibition: New Transgenic Zebrafish Lines to Study Bone Health. Int J Mol Sci 2022; 23:ijms23094748. [PMID: 35563140 PMCID: PMC9102566 DOI: 10.3390/ijms23094748] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/21/2022] [Accepted: 04/22/2022] [Indexed: 02/07/2023] Open
Abstract
Zebrafish larvae, especially gene-specific mutants and transgenic lines, are increasingly used to study vertebrate skeletal development and human pathologies such as osteoporosis, osteopetrosis and osteoarthritis. Probiotics have been recognized in recent years as a prophylactic treatment for various bone health issues in humans. Here, we present two new zebrafish transgenic lines containing the coding sequences for fluorescent proteins inserted into the endogenous genes for sp7 and col10a1a with larvae displaying fluorescence in developing osteoblasts and the bone extracellular matrix (mineralized or non-mineralized), respectively. Furthermore, we use these transgenic lines to show that exposure to two different probiotics, Bacillus subtilis and Lactococcus lactis, leads to an increase in osteoblast formation and bone matrix growth and mineralization. Gene expression analysis revealed the effect of the probiotics, particularly Bacillus subtilis, in modulating several skeletal development genes, such as runx2, sp7, spp1 and col10a1a, further supporting their ability to improve bone health. Bacillus subtilis was the more potent probiotic able to significantly reverse the inhibition of bone matrix formation when larvae were exposed to a BMP inhibitor (LDN212854).
Collapse
|
40
|
Geurtzen K, López-Delgado AC, Duseja A, Kurzyukova A, Knopf F. Laser-mediated osteoblast ablation triggers a pro-osteogenic inflammatory response regulated by reactive oxygen species and glucocorticoid signaling in zebrafish. Development 2022; 149:275194. [DOI: 10.1242/dev.199803] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 02/22/2022] [Indexed: 12/31/2022]
Abstract
ABSTRACT
In zebrafish, transgenic labeling approaches, robust regenerative responses and excellent in vivo imaging conditions enable precise characterization of immune cell behavior in response to injury. Here, we monitored osteoblast-immune cell interactions in bone, a tissue which is particularly difficult to in vivo image in tetrapod species. Ablation of individual osteoblasts leads to recruitment of neutrophils and macrophages in varying numbers, depending on the extent of the initial insult, and initiates generation of cathepsin K+ osteoclasts from macrophages. Osteoblast ablation triggers the production of pro-inflammatory cytokines and reactive oxygen species, which are needed for successful macrophage recruitment. Excess glucocorticoid signaling as it occurs during the stress response inhibits macrophage recruitment, maximum speed and changes the macrophage phenotype. Although osteoblast loss is compensated for within a day by contribution of committed osteoblasts, macrophages continue to populate the region. Their presence is required for osteoblasts to fill the lesion site. Our model enables visualization of bone repair after microlesions at single-cell resolution and demonstrates a pro-osteogenic function of tissue-resident macrophages in non-mammalian vertebrates.
Collapse
Affiliation(s)
- Karina Geurtzen
- Center for Regenerative Therapies TU Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), TU Dresden, 01307 Dresden, Germany
- Center for Healthy Aging, Medical Faculty Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, 3000 Leuven, Belgium
| | - Alejandra Cristina López-Delgado
- Center for Regenerative Therapies TU Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), TU Dresden, 01307 Dresden, Germany
- Center for Healthy Aging, Medical Faculty Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Ankita Duseja
- Center for Regenerative Therapies TU Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), TU Dresden, 01307 Dresden, Germany
- Center for Healthy Aging, Medical Faculty Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
- Department of Oncology and Metabolism, Metabolic Bone Centre, Sorby Wing, Northern General Hospital, Sheffield S5 7AU, UK
| | - Anastasia Kurzyukova
- Center for Regenerative Therapies TU Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), TU Dresden, 01307 Dresden, Germany
- Center for Healthy Aging, Medical Faculty Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
- Faculty of Health and Medical Sciences, Biotech Research & Innovation Centre (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark
| | - Franziska Knopf
- Center for Regenerative Therapies TU Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), TU Dresden, 01307 Dresden, Germany
- Center for Healthy Aging, Medical Faculty Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| |
Collapse
|
41
|
Kraus JM, Giovannone D, Rydzik R, Balsbaugh JL, Moss IL, Schwedler JL, Bertrand JY, Traver D, Hankenson KD, Crump JG, Youngstrom DW. Notch signaling enhances bone regeneration in the zebrafish mandible. Development 2022; 149:dev199995. [PMID: 35178545 PMCID: PMC8959151 DOI: 10.1242/dev.199995] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 01/21/2022] [Indexed: 12/12/2022]
Abstract
Loss or damage to the mandible caused by trauma, treatment of oral malignancies, and other diseases is treated using bone-grafting techniques that suffer from numerous shortcomings and contraindications. Zebrafish naturally heal large injuries to mandibular bone, offering an opportunity to understand how to boost intrinsic healing potential. Using a novel her6:mCherry Notch reporter, we show that canonical Notch signaling is induced during the initial stages of cartilage callus formation in both mesenchymal cells and chondrocytes following surgical mandibulectomy. We also show that modulation of Notch signaling during the initial post-operative period results in lasting changes to regenerate bone quantity one month later. Pharmacological inhibition of Notch signaling reduces the size of the cartilage callus and delays its conversion into bone, resulting in non-union. Conversely, conditional transgenic activation of Notch signaling accelerates conversion of the cartilage callus into bone, improving bone healing. Given the conserved functions of this pathway in bone repair across vertebrates, we propose that targeted activation of Notch signaling during the early phases of bone healing in mammals may both augment the size of the initial callus and boost its ossification into reparative bone.
Collapse
Affiliation(s)
- Jessica M. Kraus
- Department of Orthopaedic Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Dion Giovannone
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA
| | - Renata Rydzik
- Department of Orthopaedic Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Jeremy L. Balsbaugh
- Proteomics & Metabolomics Facility, Center for Open Research Resources & Equipment, University of Connecticut, Storrs, CT 06269, USA
| | - Isaac L. Moss
- Department of Orthopaedic Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Jennifer L. Schwedler
- Section of Cell and Developmental Biology, University of California San Diego, La Jolla, CA 92093, USA
| | - Julien Y. Bertrand
- Section of Cell and Developmental Biology, University of California San Diego, La Jolla, CA 92093, USA
| | - David Traver
- Section of Cell and Developmental Biology, University of California San Diego, La Jolla, CA 92093, USA
| | - Kurt D. Hankenson
- Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - J. Gage Crump
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA
| | - Daniel W. Youngstrom
- Department of Orthopaedic Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA
| |
Collapse
|
42
|
Paulissen SM, Castranova DM, Krispin SM, Burns MC, Menéndez J, Torres-Vázquez J, Weinstein BM. Anatomy and development of the pectoral fin vascular network in the zebrafish. Development 2022; 149:dev199676. [PMID: 35132436 PMCID: PMC8959142 DOI: 10.1242/dev.199676] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 01/24/2022] [Indexed: 12/15/2022]
Abstract
The pectoral fins of teleost fish are analogous structures to human forelimbs, and the developmental mechanisms directing their initial growth and patterning are conserved between fish and tetrapods. The forelimb vasculature is crucial for limb function, and it appears to play important roles during development by promoting development of other limb structures, but the steps leading to its formation are poorly understood. In this study, we use high-resolution imaging to document the stepwise assembly of the zebrafish pectoral fin vasculature. We show that fin vascular network formation is a stereotyped, choreographed process that begins with the growth of an initial vascular loop around the pectoral fin. This loop connects to the dorsal aorta to initiate pectoral vascular circulation. Pectoral fin vascular development continues with concurrent formation of three elaborate vascular plexuses, one in the distal fin that develops into the fin-ray vasculature and two near the base of the fin in association with the developing fin musculature. Our findings detail a complex, yet highly choreographed, series of steps involved in the development of a complete, functional, organ-specific vascular network.
Collapse
Affiliation(s)
- Scott M. Paulissen
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Daniel M. Castranova
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Shlomo M. Krispin
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Margaret C. Burns
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Javier Menéndez
- Department of Cell Biology, Skirball Institute of Biomolecular Medicine, New York University Langone Medical Center, NY 10016, USA
| | - Jesús Torres-Vázquez
- Department of Cell Biology, Skirball Institute of Biomolecular Medicine, New York University Langone Medical Center, NY 10016, USA
| | - Brant M. Weinstein
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| |
Collapse
|
43
|
Liu R, Imangali N, Ethiraj LP, Carney TJ, Winkler C. Transcriptome Profiling of Osteoblasts in a Medaka ( Oryzias latipes) Osteoporosis Model Identifies Mmp13b as Crucial for Osteoclast Activation. Front Cell Dev Biol 2022; 10:775512. [PMID: 35281094 PMCID: PMC8911226 DOI: 10.3389/fcell.2022.775512] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 01/14/2022] [Indexed: 11/13/2022] Open
Abstract
Matrix metalloproteases (MMPs) play crucial roles in extracellular matrix (ECM) modulation during osteoclast-driven bone remodeling. In the present study, we used transcriptome profiling of bone cells in a medaka model for osteoporosis and bone regeneration to identify factors critical for bone remodeling and homeostasis. This identified mmp13b, which was strongly expressed in osteoblast progenitors and upregulated under osteoporotic conditions and during regeneration of bony fin rays. To characterize the role of mmp13b in bone remodeling, we generated medaka mmp13b mutants by CRISPR/Cas9. We found that mmp13b mutants form normal numbers of osteoblasts and osteoclasts. However, osteoclast activity was severely impaired under osteoporotic conditions. In mmp13b mutants and embryos treated with the MMP13 inhibitor CL-82198, unmineralized collagens and mineralized bone matrix failed to be degraded. In addition, the dynamic migratory behavior of activated osteoclasts was severely affected in mmp13b mutants. Expression analysis showed that maturation genes were downregulated in mmp13b deficient osteoclasts suggesting that they remain in an immature and non-activated state. We also found that fin regeneration was delayed in mmp13b mutants with a concomitant alteration of the ECM and reduced numbers of osteoblast progenitors in regenerating joint regions. Together, our findings suggest that osteoblast-derived Mmp13b alters the bone ECM to allow the maturation and activation of osteoclasts during bone remodeling in a paracrine manner. Mmp13b-induced ECM alterations are also required to facilitate osteoblast progenitor recruitment and full regeneration of bony fin rays.
Collapse
Affiliation(s)
- Ranran Liu
- Department of Biological Sciences and Centre for Bioimaging Sciences, National University of Singapore, Singapore, Singapore
| | - Nurgul Imangali
- Department of Biological Sciences and Centre for Bioimaging Sciences, National University of Singapore, Singapore, Singapore
| | - Lalith Prabha Ethiraj
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Tom James Carney
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Christoph Winkler
- Department of Biological Sciences and Centre for Bioimaging Sciences, National University of Singapore, Singapore, Singapore
| |
Collapse
|
44
|
Tang WJ, Watson CJ, Olmstead T, Allan CH, Kwon RY. Single-cell resolution of MET- and EMT-like programs in osteoblasts during zebrafish fin regeneration. iScience 2022; 25:103784. [PMID: 35169687 PMCID: PMC8829776 DOI: 10.1016/j.isci.2022.103784] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 10/15/2021] [Accepted: 01/14/2022] [Indexed: 12/04/2022] Open
Abstract
Zebrafish regenerate fin rays following amputation through epimorphic regeneration, a process that has been proposed to involve the epithelial-to-mesenchymal transition (EMT). We performed single-cell RNA sequencing (scRNA-seq) to elucidate osteoblastic transcriptional programs during zebrafish caudal fin regeneration. We show that osteoprogenitors are enriched with components associated with EMT and its reverse, mesenchymal-to-epithelial transition (MET), and provide evidence that the EMT markers cdh11 and twist2 are co-expressed in dedifferentiating cells at the amputation stump at 1 dpa, and in differentiating osteoblastic cells in the regenerate, the latter of which are enriched in EMT signatures. We also show that esrp1, a regulator of alternative splicing in epithelial cells that is associated with MET, is expressed in a subset of osteoprogenitors during outgrowth. This study provides a single cell resource for the study of osteoblastic cells during zebrafish fin regeneration, and supports the contribution of MET- and EMT-associated components to this process.
Collapse
Affiliation(s)
- W. Joyce Tang
- Department of Orthopaedics and Sports Medicine, University of Washington School of Medicine, Seattle, WA 98105, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | - Claire J. Watson
- Department of Orthopaedics and Sports Medicine, University of Washington School of Medicine, Seattle, WA 98105, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | - Theresa Olmstead
- Department of Orthopaedics and Sports Medicine, University of Washington School of Medicine, Seattle, WA 98105, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | - Christopher H. Allan
- Department of Orthopaedics and Sports Medicine, University of Washington School of Medicine, Seattle, WA 98105, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | - Ronald Y. Kwon
- Department of Orthopaedics and Sports Medicine, University of Washington School of Medicine, Seattle, WA 98105, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| |
Collapse
|
45
|
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
|
46
|
Bump RG, Goo CEA, Horton EC, Rasmussen JP. Osteoblasts pattern endothelium and somatosensory axons during zebrafish caudal fin organogenesis. Development 2022; 149:dev200172. [PMID: 35129199 PMCID: PMC8918783 DOI: 10.1242/dev.200172] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 12/23/2021] [Indexed: 12/18/2022]
Abstract
Skeletal elements frequently associate with vasculature and somatosensory nerves, which regulate bone development and homeostasis. However, the deep, internal location of bones in many vertebrates has limited in vivo exploration of the neurovascular-bone relationship. Here, we use the zebrafish caudal fin, an optically accessible organ formed of repeating bony ray skeletal units, to determine the cellular relationship between nerves, bones and endothelium. In adult zebrafish, we establish the presence of somatosensory axons running through the inside of the bony fin rays, juxtaposed with osteoblasts on the inner hemiray surface. During development we show that the caudal fin progresses through sequential stages of endothelial plexus formation, bony ray addition, ray innervation and endothelial remodeling. Surprisingly, the initial stages of fin morphogenesis proceed normally in animals lacking either fin endothelium or somatosensory nerves. Instead, we find that sp7+ osteoblasts are required for endothelial remodeling and somatosensory axon innervation in the developing fin. Overall, this study demonstrates that the proximal neurovascular-bone relationship in the adult caudal fin is established during fin organogenesis and suggests that ray-associated osteoblasts pattern axons and endothelium.
Collapse
Affiliation(s)
- Rosalind G Bump
- Department of Biology, University of Washington, Seattle, WA 98195, USA
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - Camille E A Goo
- Department of Biology, University of Washington, Seattle, WA 98195, USA
| | - Emma C Horton
- Department of Biology, University of Washington, Seattle, WA 98195, USA
| | - Jeffrey P Rasmussen
- Department of Biology, University of Washington, Seattle, WA 98195, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| |
Collapse
|
47
|
Bergen DJM, Tong Q, Shukla A, Newham E, Zethof J, Lundberg M, Ryan R, Youlten SE, Frysz M, Croucher PI, Flik G, Richardson RJ, Kemp JP, Hammond CL, Metz JR. Regenerating zebrafish scales express a subset of evolutionary conserved genes involved in human skeletal disease. BMC Biol 2022; 20:21. [PMID: 35057801 PMCID: PMC8780716 DOI: 10.1186/s12915-021-01209-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 12/07/2021] [Indexed: 12/23/2022] Open
Abstract
Background Scales are mineralised exoskeletal structures that are part of the dermal skeleton. Scales have been mostly lost during evolution of terrestrial vertebrates whilst bony fish have retained a mineralised dermal skeleton in the form of fin rays and scales. Each scale is a mineralised collagen plate that is decorated with both matrix-building and resorbing cells. When removed, an ontogenetic scale is quickly replaced following differentiation of the scale pocket-lining cells that regenerate a scale. Processes promoting de novo matrix formation and mineralisation initiated during scale regeneration are poorly understood. Therefore, we performed transcriptomic analysis to determine gene networks and their pathways involved in dermal scale regeneration. Results We defined the transcriptomic profiles of ontogenetic and regenerating scales of zebrafish and identified 604 differentially expressed genes (DEGs). These were enriched for extracellular matrix, ossification, and cell adhesion pathways, but not in enamel or dentin formation processes indicating that scales are reminiscent to bone. Hypergeometric tests involving monogenetic skeletal disorders showed that DEGs were strongly enriched for human orthologues that are mutated in low bone mass and abnormal bone mineralisation diseases (P< 2× 10−3). The DEGs were also enriched for human orthologues associated with polygenetic skeletal traits, including height (P< 6× 10−4), and estimated bone mineral density (eBMD, P< 2× 10−5). Zebrafish mutants of two human orthologues that were robustly associated with height (COL11A2, P=6× 10−24) or eBMD (SPP1, P=6× 10−20) showed both exo- and endo- skeletal abnormalities as predicted by our genetic association analyses; col11a2Y228X/Y228X mutants showed exoskeletal and endoskeletal features consistent with abnormal growth, whereas spp1P160X/P160X mutants predominantly showed mineralisation defects. Conclusion We show that scales have a strong osteogenic expression profile comparable to other elements of the dermal skeleton, enriched in genes that favour collagen matrix growth. Despite the many differences between scale and endoskeletal developmental processes, we also show that zebrafish scales express an evolutionarily conserved sub-population of genes that are relevant to human skeletal disease. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-021-01209-8.
Collapse
|
48
|
Tarasco M, Gavaia PJ, Bensimon-Brito A, Cardeira-da-Silva J, Ramkumar S, Cordelières FP, Günther S, Bebianno MJ, Stainier DYR, Cancela ML, Laizé V. New insights into benzo[⍺]pyrene osteotoxicity in zebrafish. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 226:112838. [PMID: 34607190 DOI: 10.1016/j.ecoenv.2021.112838] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/22/2021] [Accepted: 09/23/2021] [Indexed: 06/13/2023]
Abstract
Persistent and ubiquitous organic pollutants, such as the polycyclic aromatic hydrocarbon benzo[⍺]pyrene (BaP), represent a major threat to aquatic organisms and human health. Beside some well-documented adverse effects on the development and reproduction of aquatic organisms, BaP was recently shown to affect fish bone formation and skeletal development through mechanisms that remain poorly understood. In this work, zebrafish bone-related in vivo assays were used to evaluate the osteotoxic effects of BaP during bone development and regeneration. Acute exposure of zebrafish larvae to BaP from 3 to 6 days post-fertilization (dpf) induced a dose-dependent reduction of the opercular bone size and a depletion of osteocalcin-positive cells, indicating an effect on osteoblast maturation. Chronic exposure of zebrafish larvae to BaP from 3 to 30 dpf affected the development of the axial skeleton and increased the incidence and severity of skeletal deformities. In young adults, BaP affected the mineralization of newly formed fin rays and scales, and impaired fin ray patterning and scale shape, through mechanisms that involve an imbalanced bone remodeling. Gene expression analyses indicated that BaP induced the activation of xenobiotic and metabolic pathways, while negatively impacting extracellular matrix formation and organization. Interestingly, BaP exposure positively regulated inflammation markers in larvae and increased the recruitment of neutrophils. A direct interaction between neutrophils and bone extracellular matrix or bone forming cells was observed in vivo, suggesting a role for neutrophils in the mechanisms underlying BaP osteotoxicity. Our work provides novel data on the cellular and molecular players involved in BaP osteotoxicity and brings new insights into a possible role for neutrophils in inflammatory bone reduction.
Collapse
Affiliation(s)
- Marco Tarasco
- Centre of Marine Sciences (CCMAR), University of Algarve, Faro, Portugal
| | - Paulo J Gavaia
- Centre of Marine Sciences (CCMAR), University of Algarve, Faro, Portugal; Faculty of Medicine and Biomedical Sciences (FMCB) and Algarve Biomedical Center (ABC), University of Algarve, Faro, Portugal
| | - Anabela Bensimon-Brito
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany; DZHK German Centre for Cardiovascular Research, Partner Site Rhine-Main, Bad Nauheim, Germany; INSERM, ATIP-Avenir, Aix Marseille University, Marseille Medical Genetics, Marseille, France
| | - João Cardeira-da-Silva
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
| | - Srinath Ramkumar
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany; Department of Life Sciences, Goethe University, Frankfurt am Main, Germany
| | - Fabrice P Cordelières
- Bordeaux Imaging Center (BIC), UMS 3420 CNRS - Université de Bordeaux - US4 INSERM, Pôle d'imagerie photonique, Centre Broca Nouvelle-Aquitaine, Bordeaux, France
| | - Stefan Günther
- DZHK German Centre for Cardiovascular Research, Partner Site Rhine-Main, Bad Nauheim, Germany; Max Planck Institute for Heart and Lung Research, Bioinformatics and Deep Sequencing Platform, Bad Nauheim, Germany
| | - Maria J Bebianno
- Centre of Marine and Environmental Research (CIMA), University of Algarve, Faro, Portugal
| | - Didier Y R Stainier
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
| | - M Leonor Cancela
- Centre of Marine Sciences (CCMAR), University of Algarve, Faro, Portugal; Faculty of Medicine and Biomedical Sciences (FMCB) and Algarve Biomedical Center (ABC), University of Algarve, Faro, Portugal
| | - Vincent Laizé
- Centre of Marine Sciences (CCMAR), University of Algarve, Faro, Portugal.
| |
Collapse
|
49
|
Foessl I, Bassett JHD, Bjørnerem Å, Busse B, Calado Â, Chavassieux P, Christou M, Douni E, Fiedler IAK, Fonseca JE, Hassler E, Högler W, Kague E, Karasik D, Khashayar P, Langdahl BL, Leitch VD, Lopes P, Markozannes G, McGuigan FEA, Medina-Gomez C, Ntzani E, Oei L, Ohlsson C, Szulc P, Tobias JH, Trajanoska K, Tuzun Ş, Valjevac A, van Rietbergen B, Williams GR, Zekic T, Rivadeneira F, Obermayer-Pietsch B. Bone Phenotyping Approaches in Human, Mice and Zebrafish - Expert Overview of the EU Cost Action GEMSTONE ("GEnomics of MusculoSkeletal traits TranslatiOnal NEtwork"). Front Endocrinol (Lausanne) 2021; 12:720728. [PMID: 34925226 PMCID: PMC8672201 DOI: 10.3389/fendo.2021.720728] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 10/21/2021] [Indexed: 12/16/2022] Open
Abstract
A synoptic overview of scientific methods applied in bone and associated research fields across species has yet to be published. Experts from the EU Cost Action GEMSTONE ("GEnomics of MusculoSkeletal Traits translational Network") Working Group 2 present an overview of the routine techniques as well as clinical and research approaches employed to characterize bone phenotypes in humans and selected animal models (mice and zebrafish) of health and disease. The goal is consolidation of knowledge and a map for future research. This expert paper provides a comprehensive overview of state-of-the-art technologies to investigate bone properties in humans and animals - including their strengths and weaknesses. New research methodologies are outlined and future strategies are discussed to combine phenotypic with rapidly developing -omics data in order to advance musculoskeletal research and move towards "personalised medicine".
Collapse
Affiliation(s)
- Ines Foessl
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Endocrine Lab Platform, Medical University of Graz, Graz, Austria
| | - J. H. Duncan Bassett
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Åshild Bjørnerem
- Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway
- Norwegian Research Centre for Women’s Health, Oslo University Hospital, Oslo, Norway
| | - Björn Busse
- Department of Osteology and Biomechanics, University Medical Center, Hamburg-Eppendorf, Hamburg, Germany
| | - Ângelo Calado
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisboa, Portugal
| | | | - Maria Christou
- Department of Hygiene and Epidemiology, Medical School, University of Ioannina, Ioannina, Greece
| | - Eleni Douni
- Institute for Bioinnovation, Biomedical Sciences Research Center “Alexander Fleming”, Vari, Greece
- Department of Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Imke A. K. Fiedler
- Department of Osteology and Biomechanics, University Medical Center, Hamburg-Eppendorf, Hamburg, Germany
| | - João Eurico Fonseca
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisboa, Portugal
- Rheumatology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte (CHULN), Lisbon Academic Medical Centre, Lisbon, Portugal
| | - Eva Hassler
- Division of Neuroradiology, Vascular and Interventional Radiology, Department of Radiology, Medical University Graz, Graz, Austria
| | - Wolfgang Högler
- Department of Paediatrics and Adolescent Medicine, Johannes Kepler University Linz, Linz, Austria
| | - Erika Kague
- The School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences, University of Bristol, Bristol, United Kingdom
| | - David Karasik
- Azrieli Faculty of Medicine, Bar-Ilan University, Ramat Gan, Israel
| | - Patricia Khashayar
- Center for Microsystems Technology, Imec and Ghent University, Ghent, Belgium
| | - Bente L. Langdahl
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Victoria D. Leitch
- Innovative Manufacturing Cooperative Research Centre, Royal Melbourne Institute of Technology, School of Engineering, Carlton, VIC, Australia
| | - Philippe Lopes
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Georgios Markozannes
- Department of Hygiene and Epidemiology, Medical School, University of Ioannina, Ioannina, Greece
| | | | | | - Evangelia Ntzani
- Department of Hygiene and Epidemiology, Medical School, University of Ioannina, Ioannina, Greece
- Department of Health Services, Policy and Practice, Center for Research Synthesis in Health, School of Public Health, Brown University, Providence, RI, United States
| | - Ling Oei
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Claes Ohlsson
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Department of Drug Treatment, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Pawel Szulc
- INSERM UMR 1033, University of Lyon, Lyon, France
| | - Jonathan H. Tobias
- Musculoskeletal Research Unit, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- MRC Integrative Epidemiology Unit, Bristol Medical School, Bristol, University of Bristol, Bristol, United Kingdom
| | - Katerina Trajanoska
- Department of Internal Medicine, Erasmus MC Rotterdam, Rotterdam, Netherlands
| | - Şansın Tuzun
- Physical Medicine & Rehabilitation Department, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Amina Valjevac
- Department of Human Physiology, School of Medicine, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Bert van Rietbergen
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Graham R. Williams
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Tatjana Zekic
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, Clinical Hospital Center Rijeka, Rijeka, Croatia
| | | | - Barbara Obermayer-Pietsch
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Endocrine Lab Platform, Medical University of Graz, Graz, Austria
| |
Collapse
|
50
|
Stewart S, Le Bleu HK, Yette GA, Henner AL, Robbins AE, Braunstein JA, Stankunas K. longfin causes cis-ectopic expression of the kcnh2a ether-a-go-go K+ channel to autonomously prolong fin outgrowth. Development 2021; 148:dev199384. [PMID: 34061172 PMCID: PMC8217709 DOI: 10.1242/dev.199384] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 04/19/2021] [Indexed: 12/11/2022]
Abstract
Organs stop growing to achieve a characteristic size and shape in scale with the body of an animal. Likewise, regenerating organs sense injury extents to instruct appropriate replacement growth. Fish fins exemplify both phenomena through their tremendous diversity of form and remarkably robust regeneration. The classic zebrafish mutant longfint2 develops and regenerates dramatically elongated fins and underlying ray skeleton. We show longfint2 chromosome 2 overexpresses the ether-a-go-go-related voltage-gated potassium channel kcnh2a. Genetic disruption of kcnh2a in cis rescues longfint2, indicating longfint2 is a regulatory kcnh2a allele. We find longfint2 fin overgrowth originates from prolonged outgrowth periods by showing Kcnh2a chemical inhibition during late stage regeneration fully suppresses overgrowth. Cell transplantations demonstrate longfint2-ectopic kcnh2a acts tissue autonomously within the fin intra-ray mesenchymal lineage. Temporal inhibition of the Ca2+-dependent phosphatase calcineurin indicates it likewise entirely acts late in regeneration to attenuate fin outgrowth. Epistasis experiments suggest longfint2-expressed Kcnh2a inhibits calcineurin output to supersede growth cessation signals. We conclude ion signaling within the growth-determining mesenchyme lineage controls fin size by tuning outgrowth periods rather than altering positional information or cell-level growth potency.
Collapse
Affiliation(s)
- Scott Stewart
- Institute of Molecular Biology, University of Oregon, 273 Onyx Bridge, 1318 Franklin Blvd, Eugene, OR 97403-1229, USA
| | - Heather K. Le Bleu
- Institute of Molecular Biology, University of Oregon, 273 Onyx Bridge, 1318 Franklin Blvd, Eugene, OR 97403-1229, USA
- Department of Biology, University of Oregon, 77 Klamath Hall, Eugene, OR 97403-1210, USA
| | - Gabriel A. Yette
- Institute of Molecular Biology, University of Oregon, 273 Onyx Bridge, 1318 Franklin Blvd, Eugene, OR 97403-1229, USA
- Department of Biology, University of Oregon, 77 Klamath Hall, Eugene, OR 97403-1210, USA
| | - Astra L. Henner
- Institute of Molecular Biology, University of Oregon, 273 Onyx Bridge, 1318 Franklin Blvd, Eugene, OR 97403-1229, USA
| | - Amy E. Robbins
- Institute of Molecular Biology, University of Oregon, 273 Onyx Bridge, 1318 Franklin Blvd, Eugene, OR 97403-1229, USA
- Department of Biology, University of Oregon, 77 Klamath Hall, Eugene, OR 97403-1210, USA
| | - Joshua A. Braunstein
- Institute of Molecular Biology, University of Oregon, 273 Onyx Bridge, 1318 Franklin Blvd, Eugene, OR 97403-1229, USA
| | - Kryn Stankunas
- Institute of Molecular Biology, University of Oregon, 273 Onyx Bridge, 1318 Franklin Blvd, Eugene, OR 97403-1229, USA
- Department of Biology, University of Oregon, 77 Klamath Hall, Eugene, OR 97403-1210, USA
| |
Collapse
|